Chapter Eleven - SLC41 Transporters—Molecular Identification and Functional Role

https://doi.org/10.1016/B978-0-12-800223-0.00011-6Get rights and content

Abstract

The solute carrier family 41 (SLC41) encompasses three members A1, A2, and A3. Based on their distant homology to the bacterial Mg2 + channel MgtE, all have been linked to Mg2 + transport. There is only very limited knowledge on the molecular biology and exact functions of SLC41A2 and SLC41A3. SLC41A1 is ubiquitously expressed and data on its functional and molecular properties, regulation, complex-forming ability, and spectrum of binding partners are available. SLC41A1 was recently identified as being the Na+/Mg2 + exchanger (NME)—a predominant Mg2 + efflux system. Mg2 +-dependent and hormonal regulation of NME activity is now known to depend on the intracellular N terminus of SLC41A1 that is involved in Mg2 + sensing and contains phosphorylation sites for protein kinase (PK) A and PKC. Data showing a link between SLC41A1 and human disorders such as Parkinson's disease, nephronophthisis (induced by the null mutation c.698G>T in renal SLC41A1), and preeclampsia make the protein a candidate therapeutic target.

Introduction

Magnesium (Mg2 +), in particular, its free intracellular form, plays an important role as a cofactor for many enzymes, as a modulator of ion channels, and in numerous other processes such as protein synthesis, nucleic acid stability, neuromuscular excitability, secretion of hormones, and in antagonizing the actions of Ca2 + (Bara and Guiet-Bara, 2001, Garban, 1997, Hartwig, 2001, Jóna et al., 2001, Mooren et al., 2001; Watanabe & Dreyfus, 1972; White & Hartzell, 1989). Therefore, the regulation of extracellular and intracellular Mg2 + levels by transmembrane and transepithelial transport processes is critical for numerous cellular and organ functions. At a functional level, the presence of such active plasma membrane Mg2 + transport mechanisms has been indicated for a long time, but the identification of their molecular entities has started only recently (reviewed in Quamme, 2010).

Among Mg2 + transporters are the three members A1, A2, and A3 of solute carrier family 41 (SLC41) with which this review deals. All of them are distantly homologous to the unique bacterial Mg2 + transport protein MgtE (Smith, Thompson, & Maguire, 1995), suggesting that they also may be involved in transport of the ion across cell membranes (Quamme, 2010, Wabakken et al., 2003). Indeed, the group of Quamme (Goytain and Quamme, 2005a, Goytain and Quamme, 2005b, Quamme, 2010) demonstrated that all three SLC41 isoforms mediate electrogenic transport of Mg2 + when cloned from renal mouse distal tubule cells and heterologously expressed in Xenopus laevis oocytes. Furthermore, in a Salmonella enterica strain (MM281) displaying disruption of all natural Mg2 + transporters, overexpression of hSLC41A1 functionally substitutes these transporters and thereby restores bacterial growth (Kolisek et al., 2008).

Using a Mag-Fura2-based fluorescence spectroscopic assay on HEK293 cells overexpressing human SLC41A1, our group performed the first direct studies showing SLC41A1-linked Mg2 + transport in a mammalian cell system (Kolisek et al., 2008, Kolisek et al., 2012). According to our data, SLC41A1 is a functional Mg2 + carrier that normally mediates efflux of the ion (Kolisek et al., 2008) and functions as a Na+/Mg2 + exchanger (NME). The NME is the best characterized Mg2 + transporter of nonepithelial cells and tissues (Günther and Vormann, 1995, Hintz et al., 1999, Xu and Willis, 1994, Yoshimura et al., 1995). The NME was first described in chicken and turkey red blood cells by the laboratory of Günther (Günther and Vormann, 1985, Günther et al., 1984). This laboratory (Büttner et al., 1998, Günther and Vormann, 1990a) and others (Feray and Garay, 1986, Flatman and Smith, 1996, Xu and Willis, 1994) studied the Na+-dependent Mg2 + transporter in numerous vertebrate erythrocytes. Over the years, NME activity has been identified in the majority of examined mammalian cell types (Cefaratti and Ruse, 2007, Fagan and Romani, 2001, Handy et al., 1996, He et al., 2005, Schweigel et al., 2006, Tessman and Romani, 1998).

Regulated Mg2 + efflux by the NME is essential for the maintenance of free intracellular magnesium concentrations ([Mg2 +]i) at levels between 0.4 and 1.2 mM (Schweigel et al., 2006, Standley and Standley, 2002), and for Mg2 + transport across intestinal, renal, placental, and mammary epithelia (Leonhard-Marek et al., 2005, Quamme, 1989, Shaw et al., 1991, Wolf et al., 2009). Additionally, hormones/mediators activate the NME to induce an [Mg2 +]i decrease, which directly or indirectly influences cellular transport mechanisms and physiological functions (Fagan and Romani, 2001, He et al., 2005, Kolisek, Sponder, et al., 2013). NME disruption or dysregulation may alter systemic and intracellular Mg2 + homeostasis and participate in the pathogenesis of various diseases and associated events such as primary hypertension (Kisters, Krefting, Barenbrock, Spieker, & Rahn, 1999), ischemic heart disease (Altura & Altura, 1995), diabetes (Saris, Mervaala, Karppanen, Khawaja, & Lewenstam, 2000), cystic fibrosis (Vormann, 1992), neurodegeneration (Barbagallo et al., 2011, Lovitt et al., 2010), inflammation (Weglicki & Phillips, 1992), and preeclampsia/eclampsia (Weaver, 1986).

Although members A2 and A3 of the SLC41 family have also been linked to Mg2 + transport, their molecular and biochemical properties (including membrane topology, cellular localization, transcriptomics, and proteomics) and physiology compared to SLC41A1 are only poorly explored. Therefore, this review concentrates on the molecular and functional characteristics of SLC41A1, in addition to its regulation and involvement in the pathophysiology of human diseases.

Section snippets

SLCA1 expression profile

Transcripts of the human and mice gene SLC41A1 have been identified in various organs and tissues. It is abundantly expressed in heart, testis, skeletal muscle, brain, kidney, liver, the adrenal and thyroid glands, and also in prostate and ovaries; lower levels have been detected in all other tested tissues, for example, colon and small intestine (Goytain & Quamme, 2005a). Transcript has been shown to be expressed in numerous cell lines such as HEK-293, Tom-1, BV173, Reh, DT40, Jurkat, and

Molecular Characteristics of SLC41A1

SLC41A1 consists of 513 amino acids with a molecular mass of ~ 56 kDa. In both human and mouse cells, SLC41A1 was predicted and experimentally confirmed to be a protein integral to the cytoplasmic membrane (Goytain and Quamme, 2005a, Kolisek et al., 2008, Kolisek et al., 2012, Mandt et al., 2011).

Initially, a model of SLC41A1 topology comprising 10 transmembrane helices (TM) with the N-inside/C-inside configuration was predicted (Fig. 11.1). Using various membrane protein prediction software

Inhibitors

No highly selective blockers for SLC41A1 are available. A reversible inhibition was observed after application of amiloride (Ki = 0.6 mM), quinidine (Ki = 0.5 mM), or imipramine (Ki = 0.5 mM) (Fagan and Romani, 2001, Günther, 2007, Günther and Vormann, 1995, Schweigel et al., 2006, Schweigel et al., 2000, Shaw et al., 1991, Tessman and Romani, 1998, Wolf et al., 1997, Zhang and Melvin, 1995).

Imipramine acts on the extracellular Na+-binding site of the NME and competition between these two compounds

Physiological Functions of SLC41A1-Related NME Activity

The NME has been shown to be responsible for the maintenance of an optimal [Mg2 +]i (Schweigel et al., 2006) for a variety of processes critical for the cell such as bioenergetics, the regulation of cellular pH, volume, and total ion balance, and cell proliferation and differentiation (Jiang et al., 2007, Romani and Scarpa, 1992, Rubin, 2005, Wolf and Trapani, 2008, Yamaguchi and Ishikawa, 2008).

Implications for a Role of Disturbed SLC41A1-Related NME Activity in Disease States

Evidence for an involvement of SLC41A1 in the pathophysiology of Parkinson's disease (PD), preeclampsia, and nephronophthisis (NPHP)-related ciliopathies has been reported recently (Hurd et al., 2013, Kolisek, Galaviz-Hernández, et al., 2013, Kolisek, Sponder, et al., 2013, Tucci et al., 2010, Yan et al., 2011).

SLC41A1 has been detected as being a part of a novel PD susceptibility locus named PARK16 (Satake et al., 2009). Subsequently, rare PD-associated coding variants of SLC41A1, namely

Molecular and Functional Characteristics of SLC41A2 and SLC41A3

The SLC41 members A2 and A3 are products of the human 12q23.3 and 3q21.2 genes and their mRNA has been identified in various organs and tissues (reviewed in Sahni and Scharenberg, 2013, Schmitz et al., 2007). In a recent review, Sahni and Scharenberg (2013) summarized SLC41A2 expression data obtained from http://www.proteinatlas.org/ENSG00000136052. According to these data, SLC41A2 shows the strongest expression in lymph nodes, stomach, lungs, testis, and skin, and is moderately expressed in

Conclusion

Over the past decade, the field of Mg2 + homeostasis has undergone a dramatic expansion with the identification of several genes encoding putative Mg2 + transporters, among them the three members of the SLC41 family. At the molecular and physiological level, SLC41A1 is the best characterized protein, whereas not much is known about the exact function(s) of SLC41A2 and SLC41A3 (e.g., as channels or carriers). A possible role of SLC41A2 in organellar Mg2 + transport must be further validated. In

Acknowledgment

The work of the authors was supported by research grants from the German Research Foundation (DFG); Schw 642/1 to M. S. R. and KO-3586/3-2 to M. K. M. S. R. and M. K. are inventors on the pending patent EP2011/065979 “Na+/Mg2 + exchanger.”

References (125)

  • R.D. Handy et al.

    Na-dependent regulation of intracellular free magnesium concentration in isolated rat ventricular myocytes

    Journal of Molecular and Cellular Cardiology

    (1996)
  • A. Hartwig

    Role of magnesium in genomic stability

    Mutation Research

    (2001)
  • A. Ikari et al.

    Up-regulation of Na+-dependent Mg2 + transport by nitric oxide and cyclic GMP pathway in renal epithelial cells

    European Journal of Pharmacology

    (2002)
  • D. Keenan et al.

    Regulation of Mg2 + homeostasis by insulin in perfused rat livers and isolated hepatocytes

    FEBS Letters

    (1996)
  • K. Kisters et al.

    Na+ and Mg2 + contents in smooth muscle cells in spontaneously hypertensive rats

    American Journal of Hypertension

    (1999)
  • M. Kolisek et al.

    SLC41A1 is a novel mammalian Mg2 + carrier

    The Journal of Biological Chemistry

    (2008)
  • T. Kubota et al.

    Na+/Mg2 + transporter acts as a Mg2 + buffering mechanism in PC12 cells

    Biochemical and Biophysical Research Communications

    (2003)
  • L.M. Kucharski et al.

    Cation hexaammines are selective and potent inhibitors of the CorA magnesium transport system

    Journal of Biological Chemistry

    (2000)
  • T. Matsuura et al.

    cAMP-induced changes of intracellular free Mg2 + levels in human erythrocytes

    Biochimica et Biophysica Acta

    (1993)
  • M.P. Ödblom et al.

    A novel DIDS-sensitive, anion-dependent Mg2 + efflux pathway in rat ventricular myocytes

    Biochemical and Biophysical Research Communications

    (1999)
  • A. Okorodudu et al.

    Ionized magnesium in the homeostasis of cells: Intracellular threshold for Mg2 + in human platelets

    Clinica Chimica Acta

    (2001)
  • A. Romani et al.

    Cyclic AMP induced Mg2 + release from rat liver hepatocytes, permeabilized hepatocytes and isolated mitochondria

    Journal of Biological Chemistry

    (1991)
  • A. Romani et al.

    Norepinephrine evokes a marked Mg2 + efflux from liver cells

    FEBS Letters

    (1990)
  • A. Romani et al.

    Regulation of cell magnesium

    Archives of Biochemistry and Biophysics

    (1992)
  • J. Sahni et al.

    The SLC41 family of MgtE-like magnesium transporters

    Molecular Aspects of Medicine

    (2013)
  • N.E. Saris et al.

    Magnesium. An update on physiological, clinical and analytical aspects

    Clinica Chimica Acta

    (2000)
  • P.R. Standley et al.

    Identification of a functional Na+/Mg2 + exchanger in human trophoblast cells

    American Journal of Hypertension

    (2002)
  • M. Tashiro et al.

    Metabolic inhibition strongly inhibits Na+-dependent Mg2 + efflux in rat ventricular myocytes

    Biophysical Journal

    (2009)
  • M. Tashiro et al.

    Na+ gradient-dependent Mg2 + transport in smooth muscle cells of guinea pig tenia cecum

    Biophysical Journal

    (1997)
  • M. Tashiro et al.

    Intracellular and extracellular concentrations of Na+ modulate Mg2 + transport in rat ventricular myocytes

    Biophysical Journal

    (2005)
  • B.M. Altura et al.

    Magnesium and cardiovascular biology: An important link between cardiovascular risk factors and atherogenesis

    Cellular & Molecular Biology Research

    (1995)
  • M. Bara et al.

    Magnesium regulation of Ca2 + channels in smooth muscle and endothelial cells of human allantochorial placental vessels

    Magnesium Research

    (2001)
  • M. Barbagallo et al.

    Altered ionized magnesium levels in mild-to-moderate Alzheimer's disease

    Magnesium Research

    (2011)
  • S. Büttner et al.

    Magnesium metabolism in erythrocytes of various species

    Magnesium-Bulletin

    (1998)
  • C. Cefaratti et al.

    Characterization of two Mg2 + transporters in sealed plasma membrane vesicles from rat liver

    American Journal of Physiology

    (1998)
  • C. Cefaratti et al.

    Protein kinase A dependent phosphorylation activates Mg2 + efflux in the basolateral region of the liver

    Molecular and Cellular Biochemistry

    (2007)
  • Ch. De Rouffignac et al.

    Renal magnesium handling and its hormonal control

    Physiological Reviews

    (1994)
  • T.E. Fagan et al.

    Streptozotocin-induced diabetes impairs Mg2 + homeostasis and uptake in rat liver cells

    American Journal of Physiology

    (2004)
  • T.E. Fagan et al.

    Activation of Na+- and Ca2 +-dependent Mg2 + extrusion by α1- and β1-adrenergic agonists in rat liver cells

    American Journal of Physiology

    (2000)
  • T.E. Fagan et al.

    Alpha(1)-Adrenoceptor-induced Mg2 + extrusion from rat hepatocytes occurs via Na+-dependent transport mechanism

    American Journal of Physiology. Gastrointestinal and Liver Physiology

    (2001)
  • M. Fatholati et al.

    Relationship between total and free cellular Mg2 + during metabolic stimulation of rat cardiac myocytes and perfused hearts

    Archives of Biochemistry and Biophysics

    (2000)
  • C. Feillet-Coudray et al.

    Erythrocyte magnesium fluxes in mice with nutritionally and genetically low magnesium status

    European Journal of Nutrition

    (2006)
  • J.C. Féray et al.

    Demonstration of Na+: Mg2+ exchange in human red cells by its sensivity to tricyclic antidepressant drugs

    Naunyn-Schmiedeberg's Archives of Pharmacology

    (1988)
  • A. Ferreira et al.

    Na+/Mg2 + exchange is functionally coupled to the insulin receptor

    Journal of Cellular Physiology

    (2004)
  • P.W. Flatman et al.

    Sodium-dependent magnesium uptake by ferret red cells

    Journal of Physiology

    (1991)
  • P.W. Flatman et al.

    Magnesium transport in magnesium-loaded ferret red blood cells

    Pflügers Archiv - European Journal of Physiology

    (1996)
  • A. Fleig et al.

    Solute carrier family SLC41, what do we really know about it?

    WIREs Membrane Transport and Signaling

    (2013)
  • E.J. Frenkel et al.

    ATP requirement of the sodium-dependent magnesium extrusion from human red blood cells

    Journal of Physiology

    (1989)
  • Z. Garban

    Implications of the chronobiochemistry-metabolism relationship in the induction of homeostasis changes. V. The action of Mg2 + and Zn2 + ions on DNA

  • A. Goytain et al.

    Functional characterization of human SLC41A1, a Mg2 + transporter with similarity to prokaryotic MgtE Mg2 + transporters

    Physiological Genomics

    (2005)
  • Cited by (25)

    • Functional characteristics and therapeutic potential of SLC41 transporters

      2023, Journal of Pharmacological Sciences
      Citation Excerpt :

      Human SLC41A2 (GenBank Accession No. NM_032148) consists of 573 amino acids and its molecular weight is approximately 62 kDa (Table 1). The human SLC41A2 is approximately 70% homologous to SLC41A1 and is estimated by Susui program (hydropathy analysis) and TMpred program to possess 12-TM and 10-TM topologies, respectively, with both extracellular N- and C-terminals.7,14 In contrast, independent epitope-tag analysis suggested that SLC41A2 may form an 11-TM topology with extracellular N-terminal and intracellular C-terminal.15

    • Ion selectivity mechanism of the MgtE channel for Mg<sup>2+</sup> over Ca<sup>2+</sup>

      2022, iScience
      Citation Excerpt :

      Therefore, cellular Mg2+ homeostasis is vital to all domains of life and is therefore strictly controlled by Mg2+ channels and transporters.4,5,6,7 MgtE is a bacterial member of the MgtE/SLC41 superfamily of Mg2+ channels and transporters, whose orthologs are widely conserved from bacteria to eukaryotes, including humans.8,9,10,11,12 MgtE is a Mg2+-selective ion channel implicated in cellular Mg2+ homeostasis13,14 and is involved in bacterial survival upon exposure to antibiotics.15

    • Dysregulation of Mg<sup>2+</sup> homeostasis contributes to acquisition of cancer hallmarks

      2019, Cell Calcium
      Citation Excerpt :

      The solute carrier family 41 (SLC41) encompasses three members: A1, A2, and A3. Based on their distant homology to the bacterial Mg2+ channel MgtE, all have been linked to Mg2+ transport [78]. There is only very limited knowledge on the molecular biology and exact functions of SLC41A2.

    • Expression of magnesium transporter SLC41A1 in the striatum of 6-hydroxydopamine-induced parkinsonian rats

      2018, Brain Research Bulletin
      Citation Excerpt :

      Although here we did not show that the results were not the effect of SO4, our parallel study obtained the same results when MgSO4 was replaced with MgCl2 (data not shown). SLC41A1 was recently identified to function as a Na+/Mg2+exchanger (Kolisek et al., 2012, 2013) and SLC41A1 is ubiquitously expressed (Schweigel-Röntgen and Kolisek, 2014). Our data showed that the expression levels of SLC41A1 mRNA and protein in the lesioned STR were downregulated by approximately 72% and 81% of the basal levels, respectively, at 7 days post-lesion induction.

    View all citing articles on Scopus
    View full text