REVIEW ARTICLE

Magnesium in aging and aging-related disease

Zhiguo Zou1, Qifan Lu1, Yifan Wang1, Xing Gao2, Xinye Zhu1, Xiyuan Lu1* and Jun Pu1*

1Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; 2Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK

Abstract

Magnesium (Mg2+) is an essential divalent cation in human body. Its balance is tightly controlled via a balanced interplay among intestinal absorption, storage, and renal excretion, involving multiple transporters across cell membrane that regulate Mg2+ influx and efflux. Mg2+ is involved in a variety of physiological and pathological processes such as enzymatic reactions, energy metabolism, cell proliferation, apoptosis, oxidative stress, and inflammation. In particular, Mg2+ contributes to the molecular hallmarks of aging. Emerging evidence demonstrates that altered Mg2+ status has been associated with many aging-related diseases, including cancer, cardiovascular disease, neurodegenerative disease, musculoskeletal function, metabolic syndrome, and COVID-19. In this review, we focus on Mg2+ and its association with molecular hallmarks of aging. We also summarize recent findings supporting an important role of Mg2+ in aging-related disease including the COVID-19 pandemic.

Keywords: magnesium; magnesium transporters; aging; human disease

 

Citation: STEMedicine 2022, 3(2): e119 - http://dx.doi.org/10.37175/stemedicine.v3.i2.119

Copyright: © 2022 Zhiguo Zou et al. This is an Open Access article distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material for any purpose, even commercially, provided the original work is properly cited and states its license.

Received: 4 January 2022; Revised: 14 February 2022; Accepted: 21 February 2022; Published: 4 April 2022

Competing interests and funding: The authors declare no conflict of interest. This research was funded by the National Natural Science Foundation of China (81873504 and 82170379), the National Key Research and Development Program of China (2018YFC1312802), and Shanghai Pujiang Program (21PJD039).

*Xiyuan Lu, Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Shanghai, 200127, China. Email: luxiyuan2021@126.com;

Jun Pu, Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Shanghai, 200127, China. Email: pujun310@hotmail.com

These authors are equal contribution to this work.

 

Magnesium (Mg2+) is the eighth most common element in the earth’s crust (1). It is involved in various cellular activities, including signal transduction, ion transport, nucleic acid, protein synthesis, genomic stability, energy supply, and metabolic homeostasis (2). Due to its physiological significance, Mg2+ balance in human body and intracellular Mg2+ homeostasis needs to be tightly controlled via a dynamic interplay between intestinal absorption and renal excretion, involving multiple transporters across cell membrane that regulate Mg2+ influx and efflux (1). Mg2+ transporters include claudins, transient receptor potential melastatin 7 (TRPM7), TRPM6, solute carrier family 41 (SLC41), ancient conserved domain protein/cyclin M (CNNM), magnesium transporter 1 (MagT1), and mitochondrial RNA splicing 2 (Mrs2) (3, 4). Both hypomagnesemia and hypermagnesemia lead to human diseases. Chronic Mg2+ deficiency contributes to an increased risk of a variety of clinical disorders, such as cardiovascular diseases (CVDs), including hypertension and cardiac arrhythmias, stroke, type 2 diabetes, metabolic syndrome, depression, and neuropsychiatric disorders (5).

Aging is a progressive loss of physiological integrity and characterized by an increasing susceptibility to the development of multiple chronic diseases (6). The aging process involves various changes at the cellular, organ, and body levels. In general, aging encompasses multiple modes induced by DNA damage, oxidative stress, drug damage, mitochondrial dysfunction, paracrine secretion, or telomere shortening (7). Aging also leads to an increased incidence of numerous human diseases, such as CVDs, neurodegenerative diseases, immune disorders, respiratory diseases, cancers, and infections (8).

In this present review, we provide an illustrative overview of Mg2+ balance in human body and the underlying mechanisms involving various Mg2+ transporters. We also discuss the involvement of Mg2+ in molecular hallmarks of aging, with a focus on the important role of Mg2+ in aging-related human diseases.

Mg2+ and human health

Mg2+ basics

Mg2+ is highly soluble and, therefore, readily available to living organisms (9). As the fourth most abundant cation in the body behind sodium, potassium, and calcium, Mg2+ is present in three forms, free, ionized (55–70%), protein-bound (20–30%), and forming complexes with anions such as phosphate, bicarbonate, and citrate (5–15%). In healthy individuals, serum concentration of total Mg2+ is maintained tightly within a range between 0.65 and 1.05 mmol/L (1). Bone functions as a large buffer pool contribute to the maintenance of serum Mg2+ concentrations (10). Total Mg2+ concentration in mammalian cells is in the 17–20 mM range, mostly located in mitochondria, nucleus, and endoplasmic reticulum (11). Within these organelles, Mg2+ is bound to phospholipid, proteins, nucleic acids, chromatin, and nucleotides. Cytoplasmic Mg2+ is the last and well detectable pool of intracellular Mg2+, with the majority complexed with Adenosine Triphosphate (ATP) phosphonucleotides, and phosphometabolites (11, 12). With the application of novel Mg2+-selective fluorescent probes and the development of new imaging techniques, cellular Mg2+ homeostasis awaits more accurate and dynamic evaluation (13). Mg2+ is an essential element for normal life activities, and Mg2+ homeostasis is precisely regulated to maintain functional stability of cells.

Mg2+ transporters and Mg2+ homeostasis

Mg2+ is absorbed mainly in the small intestine (mainly in the jejunum and the ileum), stored in bone mineral, and excreted through urine and feces (14). In the small intestine, paracellular absorption is driven by higher luminal Mg2+ concentrations (1.0–5.0 mmol/L) and accounts for 80–90% of Mg2+ uptake (15). The kidneys regulate serum Mg2+ concentrations through excretion and reabsorption. Mg2+ excretion follows a circadian rhythm and only 3–5% of serum Mg2+ is excreted, and the thick ascending limb of the loop of Henle reabsorbs 60–70% of the filtered Mg2+, with about 10% reabsorbed in the distal tubule (3, 16). Under physiological conditions, the tightly regulated balance of Mg2+ intestinal uptake, intracellular storage, and renal excretion is achieved through a specialized transport system consisting of Mg2+ transporters across biological membranes (Fig. 1).

Fig 1
Fig. 1. Mechanism of Mg2+ balance in the body. Mg2+ is absorbed mainly in the intestine and excreted through the kidney. Claudin-16, Claudin-19, TRPM6/7, Mrs2, SLC41A1/3, and CNNM2/4 are transporters of Mg2+. Receptor tyrosine kinase (RTK) and its ligands such as epidermal growth factor (EGF) influence Mg2+ homeostasis by regulating Mg2+ transporters.

Biological function of Mg2+

Mg2+, as a cofactor, has been involved in more than 600 enzymatic reactions and exerts critical effects in a variety of cellular biological activities (11, 17). Mg2+ is indispensable for major cellular processes, including energy metabolism, apoptosis, and proliferation. Mg2+ acting as an allosteric modulator or as a cofactor in the form of Mg-ATP modulates the activity of enzymes implicated in glycolysis, the Krebs cycle, and the respiratory chain, core processes of energy metabolism (18). Mechanisms whereby Mg2+ regulates cell proliferation involve the cell cycle inhibitor p27 and p53, and other negative modulators of cell proliferation such as Jumonji and numblike (18, 19). Evidence for the role of Mg2+ in cell apoptosis remains inconclusive. In many experimental models, Mg2+ deprivation induced cell death by apoptosis, and dietary Mg2+ restriction accelerated apoptosis (18, 20, 21).

X-ray crystal structure analysis reveals that Mg2+ specifically binds to the major and minor grooves of DNA (22, 23). Mg2+ stabilizes the DNA conformation by electrostatic force or hydrogen bonds and contributes to the secondary and tertiary structure of DNA (24, 25). DNA aggregation study showed that DNA fragments strongly aggregated on Mg2+-treated glass and in Mg2+ solution in a concentration-dependent manner (26). Thus, the maintenance of intracellular Mg2+ at a physiological level is important for DNA stability. Additionally, abnormal Mg2+ concentrations can also lead to oxidative stress and damage to the double-stranded structure, thus weakening the DNA stability (27, 28). Moreover, Mg2+ is required for a variety of enzymes involved in DNA repair pathways, such as nucleotide excision repair (NER), base excision repair (BER), and mismatch repair (MMR), indicating the important role of Mg2+ in maintaining genome stability (2931).

Both in vitro and in vivo experiments have demonstrated that a decreased level of Mg2+ affects the behavior of leukocytes and vascular endothelial cells and activates the production of inflammatory factors in acute inflammation (32). The underlying mechanisms involve the upregulation of stress proteins and Nuclear Factor -kappa B (NF-kB) pathways, and the impairment of Ca2+ dynamics (33). Increased reactive oxygen species (ROS) and altered Ca2+ dynamics induced by low Mg2+ further lead to impaired mitochondrial function (34). In chronic diseases, Mg2+ inhibits oxygen free radical production and mast cell degranulation, protects epithelial cells, and alleviates the inflammatory and oxidative damage to cells and blood vessels (35). Moreover, reduced Mg2+ has been observed in a variety of diseases, such as CVD, asthma, preeclampsia, osteoporosis, inflammatory bowel disease, mental health disorders, and neurodegenerative disease (3638).

Mg2+ and aging

Aging is a progressive reduction of the body’s physiological and psychological adaptability to the environment, leading to increased vulnerability to death. Studies have linked aging to molecular cross-linking, free radicals-induced damage, changes in immune function, telomere shortening, and the presence of senescence genes in DNA (7, 39). However, there is not a single theory that can completely explain the aging process, indicating that it is a complicated process regulated by multiple factors at the genetic and cellular levels, and by the environment and society. Previous studies have demonstrated several cellular and molecular hallmarks of aging, including telomere attrition, mitochondrial dysfunction, genome instability, epigenetic alterations, a loss of proteostasis, dysregulated nutrient sensing, cellular senescence, stem cell exhaustion, and altered intracellular communication (Fig. 2) (40). In this review, we focus on several hallmarks of aging, which have been closely associated with Mg2+.

Fig 2
Fig. 2. The role of Mg2+ in cell physiology, aging, and aging-related disease. Mg2+ contributes to the regulation of intracellular environment, signaling pathways, cation concentration, and mitochondrial function. Mg2+ is critically linked to molecular hallmarks of aging and is involved in the development of aging-related disease.

Mg2+ and telomere attrition

Telomere consists of a 6-bp repeat sequence, TTAGGG. It is located at the ends of chromosomes, which shortens gradually with each cell division and ultimately limits cellular proliferative capacity (41). Telomeric chromatin structure and integrity are influenced by Mg2+. Telomere (>50%) is localized in the nuclear laminae, and their trailing laminin-binding proteins are dependent on the presence of Mg2+ (42). In addition to maintaining the telomere structure, Mg2+ also contributes to the regulation of telomerase; the enzyme catalyzes the addition of guanine-rich repetitive sequences to maintain telomeres (43). Telomerase reverse transcriptase (TERT) is the catalytic component of telomerase (44). TERT is known to exerts its biological effects through an interaction with the mammalian target of rapamycin (mTOR) pathway, which is sensitive to changes of Mg2+ status (45). Liu et al. found that Mg2+ can activate the mTOR signaling, which consequently protects against the age-related decline in muscle regenerative potential and muscle mass (46). Additionally, circadian fluctuation of mTOR is regulated through Mg2+ oscillations in a MgATP-sensitive manner (45). Altered circadian rhythms affect the expression of TERT mRNA and accelerate the aging process, and fluctuations in Mg2+ are critically implicated in the modulation of cellular clock and play a role in aging-related diseases (47).

Mg2+ and mitochondrial dysfunction

Intracellular Mg2+ deficiency has been shown to affect coupled respiration, increase ROS production, and inhibit the antioxidant defense systems, including superoxide dismutase (SOD), catalase, and glutathione, leading to the disruption of mitochondrial function (48). A reduction in intracellular Mg2+ also disturbs mitochondrial Mg2+ homeostasis through modulating mitochondrial RNA splicing 2 (Mrs2), a Mg2+ transporter specifically involved in mitochondrial Mg2+ influx and promoting mitochondrial Mg2+ efflux via the 41st family of solute carrier member 3 (SLC41A3) (49). The regulation of mitochondrial Mg2+ by Mrs2 has significantly influenced cellular energy status and cellular vulnerability. Mrs2 knockdown induces loss of electron transport chain complex I, decreases cellular and nuclear ATP levels, depolarizes ΔΨm, and renders cells sensitive to oxidative stress inducers and apoptotic stimuli (48). In line with this, the overexpression of Mrs2 enhances cellular resistance to apoptosis-inducing drugs (50).

SLC41A3 is a novel transporter important for mitochondrial Mg2+ efflux, and its mRNA expression was increased under Mg2+-deficient conditions (51). Intriguingly, intracellular ATP levels were reduced in cells with SLC41A3 overexpression, indicating that the transporter contributes to mitochondrial ATP production (52). Studies based on obesity models show that during ischemia and hypoxia, the increase of extracellular Mg2+ was associated with decreased ATP levels and TRPM7 inhibition, leading to exacerbated cell damage (53, 54). Moreover, reduced intake of Mg2+ and consequent low serum Mg2+ level induce oxidative stress injury through decreasing antioxidant enzyme activity, activating the inflammatory pathways, lipid peroxidation, and endothelial dysfunction (55). Dietary Mg2+ supplementation in a mouse model of premature aging has been shown to enhance the mitochondrial membrane potential and consequently increase H+-coupled mitochondrial NADPH and ATP productions, leading to an extended life expectancy (56).

Mg2+ in genomic instability and epigenetic alterations

As early as 1976, it was shown that Mg2+ in DNA polymerase is essential for the fidelity of DNA replication (57). Acting as an essential cofactor for the DNA damage repair process, Mg2+ contributes to the stabilization of the chromatin structure during the cell cycle (58). However, in cells undergoing apoptosis, intracellular levels of free Mg2+ are increased, constituting an early event in the process of apoptosis (59). At the protein level, Ca2+- and Mg2+-dependent endonucleases have been implicated in DNA breaks during apoptosis (60). Furthermore, Mg2+ has been linked to epigenetics, and DNA methylation is associated with chromatin compaction and gene silencing.

In the offspring of Mg2+-deficient dams, 11β-hydroxysteroid dehydrogenase-2 (Hsd11b2) CpG promoters displayed substantial hypermethylation, contributing to downstream down-regulated gene expression (61). In pregnant rats, Mg2+ deficiency induced by a low Mg2+ diet is able to affect the methylation of specific cytosines in the hepatic glucocorticoid genes and consequently cause metabolic complications in the neonatal offspring (62). In addition, even a short-term deprivation of dietary Mg2+ has been shown to greatly upregulate neutral-sphingomyelinase (N-SMAse) and p53 in cardiomyocytes, which is associated with genomic changes that are important in the development of aging (63).

Mg2+ and metabolic disturbances

Many studies have demonstrated metabolic characteristics of aged animals, such as glucose intolerance, insulin resistance, decreased fatty acid oxidation, mitochondrial biosynthesis, and impaired oxidative phosphorylation (64). Mg2+ exerts its significant impact on the metabolic state through functioning as a cofactor of critical enzymes in mitochondria, where Mg2+ binds to ATP and forms Mg-ATP complex to regulate the glycolytic enzymes (65). Mg2+ contributes to the activation of mitochondrial dehydrogenases that are important in energy metabolism, including pyruvate dehydrogenase complex (PDH) (66), isocitrate dehydrogenase (IDH), and 2-oxoglutarate dehydrogenase complex (OGDH), rate-limiting enzymes of the Krebs cycle (67, 68). Mg2+ also functions as a second messenger in the regulation of insulin secretion and release, influencing insulin downstream signaling pathways (6971). Calorie restriction (CR) is known to improve lifespan and age-associated deteriorations by changing metabolic state (72). Studies have found that Mg2+ mediates the beneficial effects of CR via R-loops suppressors Rnh1/201 and Pif1, and Mg2+ supplementation protects against the accumulation of R-loops (RNA–DNA hybrids), which contributes to genomic instability and lifespan-shortening (73). It has been shown that CR increases intracellular Mg2+ by upregulating Mg2+ transporter TRPM7, while disruption of the transporter reduces environmental Mg2+ levels and compromises CR-induced repression of the lifespan-shortening formation of RNA-DNA hybrids (73).

Mg2+, protein stability, and intracellular communication

Low levels of Mg2+ have been observed in the brain tissue of patients with neurological disorders, such as migraine, epilepsy, and Parkinson’s and Alzheimer’s diseases (AD). Underlying mechanisms of these diseases might include an abnormal aggregation of extracellular amyloid β-protein (Aβ), tau phosphorylation, and neuroinflammation characterized by increased TNF-α and IL-1β expressions (74). It was observed that at the cellular level and in animal models, Mg2+ was able to downregulate TNF-α and IL-1β and reduce the accumulation of amyloid β precursor protein in the brain (75, 76). In addition, Mg2+ was documented to promote Aβ clearance, through participating in the proteasomal degradation pathway and decreasing the permeability of blood–brain barrier (77).

Mg2+ and its transporters are crucial modulators of the communication between intracellular signaling pathways. The N-methyl-D-aspartate (NMDA) receptor is involved in excitatory neurotransmission, neuroplasticity, neuroexcitotoxicity, and memory and circadian clock rhythm, important process associated with aging (78). Mg2+ is able to inhibit NMDA receptors, and a decrease in extracellular Mg2+ depolarizes the membrane potential, leading to hyperexcitability (79). The ability of TRPM7 to act as a kinase also suggests that it is able to influence intracellular signaling. Several TRPM7 kinase substrates have been identified, including annexin-1, myosin IIA heavy chain, and calpain, supporting a role for TRPM7 in cell function, such as contraction, dilation, growth, migration, apoptosis, cell adhesion, and anti-inflammatory responses (8083). In lymphocytes, Mg2+ influx through TRPM7 contributes to the functioning of the phosphoinositide 3-kinase (PI3K)/Akt/mTOR signaling pathway, which is critical to prevent axonal overgrowth and induce cellular response to membrane stretch and fluid shear force (2, 84). In addition, the overexpression of SLC41A1, a Na+/Mg2+ exchanger responsible for Mg2+ efflux, was observed to remarkably weaken the phosphorylation of Akt/PKB on Thr308 and Ser473, and ERK1/2 on Thr202/Tyr204 (85).

Mg2+ and aging-related diseases

Aging is one of the primary risk factors associated with the development of multiple human diseases, including cancer, CVD, neurodegenerative disease, osteoporosis, musculoskeletal disorders, and COVID-19 (86). The prevalence of these diseases is remarkably increased in the elderly, significantly affecting the life expectancy. For examples, approximately 80 million people in the United States have at least one form of CVD, with almost one-half aged ≥60 years (87). As of today, the majority of new cancers occur in susceptible populations aged 55 years or older, and the incidence is still rapidly increasing as compared to younger adults (88). An epidemiological study in France shows that deaths specifically related to cancer patients aged over 65 account for 75.3% from all cancers (89). Similar epidemiological data are also observed for other aging-related disease. In this review, we will throw light on the critical role of Mg2+ in these common human diseases based on recent experimental and epidemiological data.

Mg2+ and cancer

It has been shown that elevated intracellular Mg2+ concentrations favor tumor proliferation, due to its involvement in the regulation of tumor-associated telomerase and protein phosphatase 1D, a Mg2+-dependent enzyme (90). Mg2+ deficiency impairs cell migration and growth by inducing cell cycle arrest. Upon Mg2+ restriction, cancer cells undergo cell cycle arrest in the G0/G1 through the up-regulation of p27, p21, and p16 (91, 92). Additionally, Mg2+ selectively enhanced the stability G-quadruplex of oncogene promoters and consequently impacts transcription of target genes, which provides a new insight on the observation that Mg2+ deficiency promotes the occurrence of cancers (93).

In addition to Mg2+ itself, Mg2+ transporters are also importantly implicated in the development of cancer. Many studies have already shown that the Mg2+ transporter TRPM7 exerts important effects on cellular proliferation, survival, cell cycle progression, migration, and invasion in various cell lines of cancer. An aberrant expression of TRPM7 has been observed in different types of cancer, especially pancreatic adenocarcinoma. The TRPM7 expression in the tissue of human pancreatic adenocarcinoma was positively correlated with the primary tumor size, stage, and progression (94). In line with this, the down-regulation of TRPM7 in human pancreatic cancer cells inhibits cell proliferation, effect that could be diminished by Mg2+ supplementation (95). In breast cancer tissues, the CNNM3 expression is increased and promotes Mg2+ entry into cells through binding to the phosphatase of regenerating liver 2 (PRL-2), consequently contributing to oncogenesis (96).

Mg2+ and cardiovascular disease

Mild to moderate Mg2+ deficiency associates with an increased risk of atherosclerosis, ischemic heart disease, and congestive heart failure (HF), while severe Mg2+ deficiency can cause ventricular arrhythmias that might lead to even sudden cardiac death (97). Mg2+ is important in regulating membrane potential and contractility of cardiomyocytes and autoregulatory cells (98). Severe Mg2+ loss may induce a prolongation of QT interval and the widening of QRS waves, resulting in ventricular arrhythmias (99). In the vasculature, Mg2+ is known to exert vasoprotective effects through regulating vascular tone and cytosolic Ca2+ (100). Epidemiological and experimental data show that Mg2+ has an inverse association with blood pressure, and the Mg2+ supplement may decrease peripheral vascular resistance and blood pressure (101). A meta-analysis of 34 randomized clinical trials involving 2028 participants shows that daily intake of 300 mg Mg2+ for 1 month is sufficient to elevate serum Mg2+ and reduce blood pressure (102). It is worth noting that TRPM7, acting as a vascular Mg2+ regulator, is critically implicated in hypertension. In a mouse model of TRPM7 deficiency, angiotensin II (Ang II)-induced blood pressure elevation was exaggerated, and the deteriorate effects on cardiac remodeling and left ventricular dysfunction were also amplified (103).

Mg2+ was found to influence the development of atherosclerosis by regulating the production of prostacyclin and NO (104106). Low Mg2+ status might promote the expression of proinflammatory and prothrombotic factors, such as interleukin-1β (IL-1β), IL-6, and vascular cell adhesion molecule 1 (VCAM-1), important molecules for the progress of atherosclerosis (107). Vascular calcification is one of the main features of atherosclerosis (108). Mg2+ regulated by TRPM7 increases the expression of inhibitors of calcification such as matrix Gla protein, osteopontin, and bone morphogenetic protein (BMP7) and reduces the formation of osteogenic VSMCs and vascular calcification (109). Moreover, clinical studies have confirmed that low serum Mg2+ levels are found in patients with coronary artery disease (CAD), an atherosclerotic disease that typically affects the heart (110).

Mg2+ and neurodegenerative disease

Mg2+ abundance in cerebrospinal fluid is higher than that in blood and is positively correlated with cognitive function (2, 111). In an in vitro blood–brain barrier model, TRPM7 and MagT1 are functionally active and involved in the transport of Mg2+ (77). Neuronal growth depends on changes of the cytoskeleton in the growth cone (112). High TRPM7 expression was observed in the tips of the growth cone, which mediates Mg2+ influx to fulfil the energy requirements of the neuronal network (113). In addition, Mg2+ promotes the differentiation of neural stem cells into neurons, and TRPM7 was able to influence astrocyte proliferation and migration by regulating extracellular regulated protein kinase (ERK) and c-Jun N-terminal kinase (JNK) activities (114).

Mg2+ deficiency causes emotional memory impairment and worsens the symptoms presented in AD (115). Mg2+ protects neuronal function in AD by reducing the TNF-α expression in glial cells with mechanisms involving PI3K/Akt and nuclear factor-kappa B (NF-κB) (76). In an animal model of Parkinson’s disease (PD), the expression level of SLC41A1 was reduced contributing to apomorphine (APO)-induced rotational behavior, while Mg2+ supplement significantly improved the behavior (116). Additionally, function loss of Mrs2 transporter important for mitochondrial Mg2+ homeostasis was observed in rats with demyelinating mutations. (117). It is believed that the disruption of mitochondrial Mg2+ dynamic dysregulates ATP production, leading to abnormal cell metabolism, thus triggering demyelination and neurological dysfunctions (49). Furthermore, neurological diseases such as migraine, seizures, anxiety, depression, and stroke have also been associated with a low Mg2+ abundance in serum and cerebrospinal fluid (49, 118, 119). However, further well-designed Randomized Controlled Trial (RCT) studies are still needed to confirm whether Mg2+ supplementation can improve the prognosis of these diseases.

Mg2+ and musculoskeletal function

Mg2+ is important for skeletal muscle energy metabolism as a cofactor for enzymes related to ATP synthesis, and Mg2+ deficiency is associated with fibromyalgia, a condition characterized by chronic widespread musculoskeletal pain (120). In healthy older women, daily Mg2+ supplementation over 12 weeks significantly improved physical performance assessed by Short-Physical Performance Battery (SPPB) score and a 4-m walking test, suggesting that Mg2+ may serve as a complementary treatment for aging-related physical deconditioning (121). In addition, Mg2+ enhances the activity of vitamin D and is involved in osteoblast proliferation and bone mineralization, and Mg2+ deficiency leads to reduced parathyroid hormone (PTH) synthesis and secretion, causing low Ca2+ (122). Therefore, Mg2+ supplement might be promising in the prevention of age-related osteoporosis.

Mg2+ and abnormal metabolism

Mg2+ has been found to modulate the activation of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, lipoprotein lipase, and lecithin-cholesterol acyltransferase, enzymes critically implicated in lipid metabolism (123). Mg2+ is also required to regulate genes that are involved in adipogenesis, lipolysis, and inflammation, such as PPAR-γ (124). Animal studies have shown that Mg2+ decreases lipid accumulation in hepatocytes by regulating enzymatic activities and transcriptional genes related to lipid metabolism (125). In type 2 diabetic patients, reduced serum Mg2+ levels were observed, and Mg2+ is believed to mediate the development of diabetes through insulin resistance (126). The effects of Mg2+ on glucose, lipids, and blood pressure suggest a role of Mg2+ in metabolic syndrome, and in line with this, Mg2+ supplementation was reported to be effective in treating metabolic syndrome in patients with comorbid hypomagnesemia (127).

Mg2+ and COVID-19

COVID-19 is one type of zoonotic epidemic exploded in late December in 2019, which shares a high degree of homology with Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) (128). COVID-19 is more prevalent in the elderly, and elderly patients are more likely to develop severe or critical pneumonia, with a high mortality rate (129, 130). Intriguingly, more and more clinical evidence showed that some aspects of COVID-19 pathogenesis are similar with the symptoms presented in Mg2+ deficiency (131). It has been demonstrated that up to 60% of critically ill patients in the ICU present with some degree of Mg2+ deficiency (132). In addition, people with hypertension, CVD, diabetes, and obesity are at high risk of developing COVID-19 (133). It is well known that these aging-related diseases are all characterized by hypomagnesemia and are associated with low-grade inflammation. Low Mg2+ status is believed to stimulate granulocyte oxidative burst, activate endothelial cells, and upregulate the production of cytokines, thus promoting inflammation that might lead to diseases (33). Mg2+ also leads to increased plasminogen activator inhibitor-1 (PAI-1) production and the inhibition of fibrinolysis, which may be associated with the elevated D-D dimer observed in COVID-19 (134). Moreover, COVID-19 could directly infect endothelial cells via ACE2 receptor, which induces cytokine storm, thus increasing permeability, vasoconstriction, and fostering thrombogenesis, while Mg2+ is significant in maintaining endothelial function and vascular integrity (135, 136). Collectively, a low Mg2+ status might accelerate the progression of COVID-19 from mild to serve stages, and Mg2+ supplementation might be one of the feasible treatment modalities, especially in severe patients (137).

Conclusion and perspectives

Mg2+ is an indispensable cation in human body regulating a variety of physiological and pathological processes. Under normal conditions, intracellular and extracellular Mg2+ levels are tightly controlled through a complicated Mg2+ transport system, and dynamic Mg2+ homeostasis is critical for human health. However, altered Mg2+ status also contributes to the molecular hallmarks of the aging process. Emerging evidence indicates that Mg2+ plays an important role in multiple aging-related diseases including the COVID-19 pandemic. The modulation of Mg2+ status might be a promising therapeutic option for human disease, which deserved further investigation.

References

  1. Jahnen-Dechent W, Ketteler M. Magnesium basics. Clin Kidney J 2012; 5(Suppl 1): i3–14. doi: 10.1093/ndtplus/sfr163
  2. Yamanaka R, Shindo Y, Oka K. Magnesium is a key player in neuronal maturation and neuropathology. Int J Mol Sci 2019; 20(14): 3439. doi: 10.3390/ijms20143439
  3. de Baaij JH, Hoenderop JG, Bindels RJ. Magnesium in man: implications for health and disease. Physiol Rev 2015; 95(1): 1–46. doi: 10.1152/physrev.00012.2014
  4. Giménez-Mascarell P, Schirrmacher CE, Martínez-Cruz LA, Müller D, Runnels LW. Novel aspects of renal magnesium homeostasis TRPM6 and TRPM7: a mul-TRP-PLIK-cation of channel functions. Front Pediatr 2018; 6(1): 77. doi: 10.3389/fped.2018.00077
  5. Gröber U, Schmidt J, Kisters K. Magnesium in prevention and therapy. Nutrients 2015; 7(9): 8199–226. doi: 10.3390/nu7095388
  6. Fabbri E, Zoli M, Gonzalez-Freire M, Salive ME, Studenski SA, Ferrucci L. Aging and multimorbidity: new tasks, priorities, and frontiers for integrated gerontological and clinical research. J Am Med Dir Assoc 2015; 16(8): 640–7. doi: 10.1016/j.jamda.2015.03.013
  7. Da Costa JP, Vitorino R, Silva GM, Vogel C, Duarte AC, Rocha-Santos T. A synopsis on aging-Theories, mechanisms and future prospects. Ageing Res Rev 2016; 29: 90–112. doi: 10.1016/j.arr.2016.06.005
  8. Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of cellular senescence. Trends Cell Biol 2018; 28(6): 436–53. doi: 10.1016/j.tcb.2018.02.001
  9. Maguire ME, Cowan JA. Magnesium chemistry and biochemistry. Biometals 2002; 15(3): 203–10. doi: 10.1023/A:1016058229972
  10. Alfrey AC, Miller NL. Bone magnesium pools in uremia. J Clin Investig 1973; 52(12): 3019–27. doi: 10.1172/JCI107500
  11. Romani AM. Cellular magnesium homeostasis. Arch Biochem Biophys 2011; 512(1): 1–23. doi: 10.1016/j.abb.2011.05.010
  12. Yamagami R, Bingaman JL, Frankel EA, Bevilacqua PC. Cellular conditions of weakly chelated magnesium ions strongly promote RNA stability and catalysis. Nat Commun 2018; 9(1): 2149. doi: 10.1038/s41467-018-04415-1
  13. Sargenti A, Farruggia G, Zaccheroni N, Marraccini C, Sgarzi M, Cappadone C, et al. Synthesis of a highly Mg2+-selective fluorescent probe and its application to quantifying and imaging total intracellular magnesium. Nat Protoc 2017; 12(3): 461–71. doi: 10.1038/nprot.2016.183
  14. Saris NE, Mervaala E, Karppanen H, Khawaja JA, Lewenstam A. Magnesium. An update on physiological, clinical and analytical aspects. Clin Chim Acta 2000; 294(1–2): 1–26. doi: 10.1016/S0009-8981(99)00258-2
  15. de Baaij JH, Hoenderop JG, Bindels RJ. Regulation of magnesium balance: lessons learned from human genetic disease. Clin Kidney J 2012; 5(Suppl 1): i15–24. doi: 10.1093/ndtplus/sfr164
  16. De Baaij JH. The art of magnesium transport. Magnes Res 2015; 28(3): 85–91. doi: 10.1684/mrh.2015.0388
  17. Swaminathan R. Magnesium metabolism and its disorders. Clin Biochem Rev 2003; 24(2): 47–66.
  18. Wolf FI, Trapani V. Cell (patho)physiology of magnesium. Clin Sci 2008; 114(1): 27–35. doi: 10.1042/CS20070129
  19. Ikari A, Sawada H, Sanada A, Tonegawa C, Yamazaki Y, Sugatani J. Magnesium deficiency suppresses cell cycle progression mediated by increase in transcriptional activity of p21(Cip1) and p27(Kip1) in renal epithelial NRK-52E cells. J Cell Biochem 2011; 112(12): 3563–72. doi: 10.1002/jcb.23284
  20. Martin H, Richert L, Berthelot A. Magnesium deficiency induces apoptosis in primary cultures of rat hepatocytes. J Nutr 2003; 133(8): 2505–11. doi: 10.1093/jn/133.8.2505
  21. Feng H, Guo L, Gao H, Li XA. Deficiency of calcium and magnesium induces apoptosis via scavenger receptor BI. Life Sci 2011; 88(13–14): 606–12. doi: 10.1016/j.lfs.2011.01.020
  22. Chen Y, Gao T, Wang Y, Yang G. Investigating the influence of magnesium ions on p53-DNA binding using atomic force microscopy. Int J Mol Sci 2017; 18(7): 1585. doi: 10.3390/ijms18071585
  23. Chiu TK, Dickerson RE. 1 A crystal structures of B-DNA reveal sequence-specific binding and groove-specific bending of DNA by magnesium and calcium. J Mol Biol 2000; 301(4): 915–45. doi: 10.1006/jmbi.2000.4012
  24. Price MA, Tullius TD. Using hydroxyl radical to probe DNA structure. Methods Enzymol 1992; 212: 194–219. doi: 10.1016/0076-6879(92)12013-G
  25. SantaLucia J, Jr, Hicks D. The thermodynamics of DNA structural motifs. Annu Rev Biophys Biomol Struct 2004; 33: 415–40. doi: 10.1146/annurev.biophys.32.110601.141800
  26. Bui VC, Nguyen TH. DNA aggregation induced by Mg(2+) ions under different conditions. J Mol Recognit 2018; 31(9): e2721. doi: 10.1002/jmr.2721
  27. Anastassopoulou J, Theophanides T. Magnesium-DNA interactions and the possible relation of magnesium to carcinogenesis. Irradiation and free radicals. Crit Rev Oncol Hematol 2002; 42(1): 79–91. doi: 10.1016/S1040-8428(02)00006-9
  28. Kielar C, Xin Y, Shen B, Kostiainen MA, Grundmeier G, Linko V, et al. On the stability of DNA origami nanostructures in low-magnesium buffers. Angew Chem Int Ed Engl 2018; 57(30): 9470–4. doi: 10.1002/anie.201802890
  29. Calsou P, Salles B. Properties of damage-dependent DNA incision by nucleotide excision repair in human cell-free extracts. Nucleic Acids Res 1994; 22(23): 4937–42. doi: 10.1093/nar/22.23.4937
  30. Ban C, Junop M, Yang W. Transformation of MutL by ATP binding and hydrolysis: a switch in DNA mismatch repair. Cell 1999; 97(1): 85–97. doi: 10.1016/S0092-8674(00)80717-5
  31. Hartwig A. Role of magnesium in genomic stability. Mutat Res 2001; 475(1–2): 113–21. doi: 10.1016/S0027-5107(01)00074-4
  32. Stankovic MS, Janjetovic K, Velimirovic M, Milenkovic M, Stojkovic T, Puskas N, et al. Effects of IL-33/ST2 pathway in acute inflammation on tissue damage, antioxidative parameters, magnesium concentration and cytokines profile. Exp Mol Pathol 2016; 101(1): 31–7. doi: 10.1016/j.yexmp.2016.05.012
  33. Maier JA, Castiglioni S, Locatelli L, Zocchi M, Mazur A. Magnesium and inflammation: advances and perspectives. Semin Cell Dev Biol 2021; 115: 37–44. doi: 10.1016/j.semcdb.2020.11.002
  34. Kolisek M, Touyz RM, Romani A, Barbagallo M. Magnesium and other biometals in oxidative medicine and redox biology. Oxid Med Cell Longev 2017; 2017: 7428796. doi: 10.1155/2017/7428796
  35. Locatelli L, Fedele G, Castiglioni S, Maier JA. Magnesium deficiency induces lipid accumulation in vascular endothelial cells via oxidative stress-the potential contribution of EDF-1 and PPARgamma. Int J Mol Sci 2021; 22(3): 1050. doi: 10.3390/ijms22031050
  36. Castiglioni S, Cazzaniga A, Maier JA. Potential interplay between NFκB and PPARγ in human dermal microvascular endothelial cells cultured in low magnesium. Magnes Res 2014; 27(2): 86–93. doi: 10.1684/mrh.2014.0365
  37. Cairns CB, Kraft M. Magnesium attenuates the neutrophil respiratory burst in adult asthmatic patients. Acad Emerg Med 1996; 3(12): 1093–7. doi: 10.1111/j.1553-2712.1996.tb03366.x
  38. Shahi A, Aslani S, Ataollahi M, Mahmoudi M. The role of magnesium in different inflammatory diseases. Inflammopharmacology 2019; 27(4): 649–61. doi: 10.1007/s10787-019-00603-7
  39. Tower J. Programmed cell death in aging. Ageing Res Rev 2015; 23(Pt A): 90–100. doi: 10.1016/j.arr.2015.04.002
  40. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell 2013; 153(6): 1194–217. doi: 10.1016/j.cell.2013.05.039
  41. Muraki K, Nyhan K, Han L, Murnane JP. Mechanisms of telomere loss and their consequences for chromosome instability. Front Oncol 2012; 2: 135. doi: 10.3389/fonc.2012.00135
  42. Ganesh S, Qin Z, Spagnol ST, Biegler MT, Coffey KA, Kalinowski A, et al. The tail domain of lamin B1 is more strongly modulated by divalent cations than lamin A. Nucleus 2015; 6(3): 203–11. doi: 10.1080/19491034.2015.1031436
  43. Zvereva MI, Shcherbakova DM, Dontsova OA. Telomerase: structure, functions, and activity regulation. Biochemistry 2010; 75(13): 1563–83. doi: 10.1134/S0006297910130055
  44. Zhou J, Ding D, Wang M, Cong YS. Telomerase reverse transcriptase in the regulation of gene expression. BMB Rep 2014; 47(1): 8–14. doi: 10.5483/BMBRep.2014.47.1.284
  45. Feeney KA, Hansen LL, Putker M, Olivares-Yañez C, Day J, Eades LJ, et al. Daily magnesium fluxes regulate cellular timekeeping and energy balance. Nature 2016; 532(7599): 375–9. doi: 10.1038/nature17407
  46. Liu Y, Wang Q, Zhang Z, Fu R, Zhou T, Long C, et al. Magnesium supplementation enhances mTOR signalling to facilitate myogenic differentiation and improve aged muscle performance. Bone 2021; 146: 115886. doi: 10.1016/j.bone.2021.115886
  47. Chen WD, Wen MS, Shie SS, Lo YL, Wo HT, Wang CC, et al. The circadian rhythm controls telomeres and telomerase activity. Biochem Biophys Res Commun 2014; 451(3): 408–14. doi: 10.1016/j.bbrc.2014.07.138
  48. Liu M, Dudley SC, Jr., de Baaij JH, Arjona FJ, van den Brand M, Lavrijsen M, et al. Magnesium, oxidative stress, inflammation, and cardiovascular disease. Antioxidants 2020; 9(10): 28565. doi: 10.3390/antiox9100907
  49. Yamanaka R, Tabata S, Shindo Y, Hotta K, Suzuki K, Soga T, et al. Mitochondrial Mg(2+) homeostasis decides cellular energy metabolism and vulnerability to stress. Sci Rep 2016; 6: 30027. doi: 10.1038/srep30027
  50. Merolle L, Sponder G, Sargenti A, Mastrototaro L, Cappadone C, Farruggia G, et al. Overexpression of the mitochondrial Mg channel MRS2 increases total cellular Mg concentration and influences sensitivity to apoptosis. Metallomics 2018; 10(7): 917–28. doi: 10.1039/C8MT00050F
  51. Goytain A, Quamme GA. Identification and characterization of a novel family of membrane magnesium transporters, MMgT1 and MMgT2. Am J Physiol Cell Physiol 2008; 294(2): C495–502. doi: 10.1152/ajpcell.00238.2007
  52. Mastrototaro L, Smorodchenko A, Aschenbach JR, Kolisek M, Sponder G. Solute carrier 41A3 encodes for a mitochondrial Mg(2+) efflux system. Sci Rep 2016; 6: 27999. doi: 10.1038/srep27999
  53. Su LT, Chen HC, González-Pagán O, Overton JD, Xie J, Yue L, et al. TRPM7 activates m-calpain by stress-dependent stimulation of p38 MAPK and c-Jun N-terminal kinase. J Mol Biol 2010; 396(4): 858–69. doi: 10.1016/j.jmb.2010.01.014
  54. Inoue H, Murayama T, Tashiro M, Sakurai T, Konishi M. Mg(2+)- and ATP-dependent inhibition of transient receptor potential melastatin 7 by oxidative stress. Free Radic Biol Med 2014; 72: 257–66. doi: 10.1016/j.freeradbiomed.2014.04.015
  55. Morais JB, Severo JS, Santos LR, de Sousa Melo SR, de Oliveira Santos R, de Oliveira AR, et al. Role of magnesium in oxidative stress in individuals with obesity. Biol Trace Elem Res 2017; 176(1): 20–6. doi: 10.1007/s12011-016-0793-1
  56. Villa-Bellosta R. Dietary magnesium supplementation improves lifespan in a mouse model of progeria. EMBO Mol Med 2020; 12(10): e12423. doi: 10.15252/emmm.202012423
  57. Sirover MA, Loeb LA. Metal activation of DNA synthesis. Biochem Biophys Res Commun 1976; 70(3): 812–7. doi: 10.1016/0006-291X(76)90664-1
  58. Ohyama T. New aspects of magnesium function: a key regulator in nucleosome self-assembly, chromatin folding and phase separation. Int J Mol Sci 2019; 20(17): 4232. doi: 10.3390/ijms20174232
  59. Krzywoszynska K, Witkowska D, Swiatek-Kozlowska J, Szebesczyk A, Kozlowski H. General aspects of metal ions as signaling agents in health and disease. Biomolecules 2020; 10(10): 1417. doi: 10.3390/biom10101417
  60. Giannakis C, Forbes IJ, Zalewski PD. Ca2+/Mg(2+)-dependent nuclease: tissue distribution, relationship to inter-nucleosomal DNA fragmentation and inhibition by Zn2+. Biochem Biophys Res Commun 1991; 181(2): 915–20. doi: 10.1016/0006-291X(91)91278-K
  61. Hruby A, McKeown NM, Song Y, Djousse L. Dietary magnesium and genetic interactions in diabetes and related risk factors: a brief overview of current knowledge. Nutrients 2013; 5(12): 4990–5011. doi: 10.3390/nu5124990
  62. Takaya J, Iharada A, Okihana H, Kaneko K. Magnesium deficiency in pregnant rats alters methylation of specific cytosines in the hepatic hydroxysteroid dehydrogenase-2 promoter of the offspring. Epigenetics 2011; 6(5): 573–8. doi: 10.4161/epi.6.5.15220
  63. Shah NC, Shah GJ, Li Z, Jiang XC, Altura BT, Altura BM. Short-term magnesium deficiency downregulates telomerase, upregulates neutral sphingomyelinase and induces oxidative DNA damage in cardiovascular tissues: relevance to atherogenesis, cardiovascular diseases and aging. Int J Clin Exp Med 2014; 7(3): 497–514.
  64. Houtkooper RH, Argmann C, Houten SM, Cantó C, Jeninga EH, Andreux PA, et al. The metabolic footprint of aging in mice. Sci Rep 2011; 1: 134. doi: 10.1038/srep00134
  65. Garfinkel L, Garfinkel D. Magnesium regulation of the glycolytic pathway and the enzymes involved. Magnesium 1985; 4(2–3): 60–72.
  66. Thomas AP, Diggle TA, Denton RM. Sensitivity of pyruvate dehydrogenase phosphate phosphatase to magnesium ions. Similar effects of spermine and insulin. Biochem J 1986; 238(1): 83–91. doi: 10.1042/bj2380083
  67. Tretter L, Adam-Vizi V. Alpha-ketoglutarate dehydrogenase: a target and generator of oxidative stress. Philos Trans R Soc Lond B Biol Sci 2005; 360(1464): 2335–45. doi: 10.1098/rstb.2005.1764
  68. McLain AL, Szweda PA, Szweda LI. α-Ketoglutarate dehydrogenase: a mitochondrial redox sensor. Free Radic Res 2011; 45(1): 29–36. doi: 10.3109/10715762.2010.534163
  69. Mooren FC. Magnesium and disturbances in carbohydrate metabolism. Diabetes Obes Metab 2015; 17(9): 813–23. doi: 10.1111/dom.12492
  70. Rodríguez-Morán M, Guerrero-Romero F. Insulin secretion is decreased in non-diabetic individuals with hypomagnesaemia. Diabetes Metab Res Rev 2011; 27(6): 590–6. doi: 10.1002/dmrr.1206
  71. Kamran M, Kharazmi F, Malekzadeh K, Talebi A, Khosravi F, Soltani N. Effect of long-term administration of oral magnesium sulfate and insulin to reduce streptozotocin-induced hyperglycemia in rats: the role of Akt2 and IRS1 gene expressions. Biol Trace Elem Res 2019; 190(2): 396–404. doi: 10.1007/s12011-018-1555-z
  72. Balasubramanian P, Howell PR, Anderson RM. Aging and caloric restriction research: a biological perspective with translational potential. EBioMedicine 2017; 21: 37–44. doi: 10.1016/j.ebiom.2017.06.015
  73. Abraham KJ, Chan JN, Salvi JS, Ho B, Hall A, Vidya E, et al. Intersection of calorie restriction and magnesium in the suppression of genome-destabilizing RNA-DNA hybrids. Nucleic Acids Res 2016; 44(18): 8870–84. doi: 10.1093/nar/gkw752
  74. Veronese N, Zurlo A, Solmi M, Luchini C, Trevisan C, Bano G, et al. Magnesium status in Alzheimer’s disease: a systematic review. Am J Alzheimers Dis Other Demen 2016; 31(3): 208–13. doi: 10.1177/1533317515602674
  75. Yu J, Sun M, Chen Z, Lu J, Liu Y, Zhou L, et al. Magnesium modulates amyloid-beta protein precursor trafficking and processing. J Alzheimers Dis 2010; 20(4): 1091–106. doi: 10.3233/JAD-2010-091444
  76. Yu X, Guan PP, Zhu D, Liang YY, Wang T, Wang ZY, et al. Magnesium ions inhibit the expression of tumor necrosis factor α and the activity of γ-secretase in a β-amyloid protein-dependent mechanism in APP/PS1 transgenic mice. Front Mol Neurosci 2018; 11(5): 172. doi: 10.3389/fnmol.2018.00172
  77. Zhu D, Su Y, Fu B, Xu H. Magnesium reduces blood-brain barrier permeability and regulates amyloid-β transcytosis. Mol Neurobiol 2018; 55(9): 7118–31. doi: 10.1007/s12035-018-0896-0
  78. Hansen KB, Yi F, Perszyk RE, Furukawa H, Wollmuth LP, Gibb AJ, et al. Structure, function, and allosteric modulation of NMDA receptors. J Gen Physiol 2018; 150(8): 1081–105. doi: 10.1085/jgp.201812032
  79. Nechifor M. Magnesium in addiction – a general view. Magnes Res 2018; 31(3): 90–8. doi: 10.1684/mrh.2018.0443
  80. Yogi A, Callera GE, Antunes TT, Tostes RC, Touyz RM. Transient receptor potential melastatin 7 (TRPM7) cation channels, magnesium and the vascular system in hypertension. Circ J 2011; 75(2): 237–45. doi: 10.1253/circj.CJ-10-1021
  81. Schlingmann KP, Waldegger S, Konrad M, Chubanov V, Gudermann T. TRPM6 and TRPM7 – gatekeepers of human magnesium metabolism. Biochim Biophys Acta 2007; 1772(8): 813–21. doi: 10.1016/j.bbadis.2007.03.009
  82. Trzeciakiewicz A, Opolski A, Mazur A. [TRPM7: a protein responsible for magnesium homeostasis in a cell]. Postepy Hig Med Dosw 2005; 59: 496–502.
  83. He Y, Yao G, Savoia C, Touyz RM. Transient receptor potential melastatin 7 ion channels regulate magnesium homeostasis in vascular smooth muscle cells: role of angiotensin II. Circ Res 2005; 96(2): 207–15. doi: 10.1161/01.RES.0000152967.88472.3e
  84. Sahni J, Tamura R, Sweet IR, Scharenberg AM. TRPM7 regulates quiescent/proliferative metabolic transitions in lymphocytes. Cell Cycle 2010; 9(17): 3565–74. doi: 10.4161/cc.9.17.12798
  85. Sponder G, Abdulhanan N, Fröhlich N, Mastrototaro L, Aschenbach JR, Röntgen M, et al. Overexpression of Na(+)/Mg(2+) exchanger SLC41A1 attenuates pro-survival signaling. Oncotarget 2018; 9(4): 5084–104. doi: 10.18632/oncotarget.23598
  86. Li Z, Zhang Z, Ren Y, Wang Y, Fang J, Yue H, et al. Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology 2021; 22(2): 165–87. doi: 10.1007/s10522-021-09910-5
  87. Lloyd-Jones D, Adams R, Carnethon M, De Simone G, Ferguson TB, Flegal K, et al. Heart disease and stroke statistics – 2009 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation 2009; 119(3): e21–181. doi: 10.1161/CIRCULATIONAHA.108.191261
  88. Mayrbäurl B, Thaler J. [Cancer in the elderly]. Acta Med Austriaca 2004; 31(2): 40–4.
  89. Bringuier M, Tazaro-Martinez C, Bonhomme S, Geiss R. [Epidemiology of cancer in the elderly in France]. Soins Gerontol 2019; 24(135): 12–4. doi: 10.1016/j.sger.2018.11.003
  90. Castiglioni S, Maier JA. Magnesium and cancer: a dangerous liason. Magnes Res 2011; 24(3): S92–100. doi: 10.1684/mrh.2011.0285
  91. Covacci V, Bruzzese N, Sgambato A, Di Francesco A, Russo MA, Wolf FI, et al. Magnesium restriction induces granulocytic differentiation and expression of p27Kip1 in human leukemic HL-60 cells. J Cell Biochem 1998; 70(3): 313–22. doi: 10.1002/(SICI)1097-4644(19980901)70:3<313::AID-JCB4>3.0.CO;2-Q
  92. Sgambato A, Wolf FI, Faraglia B, Cittadini A. Magnesium depletion causes growth inhibition, reduced expression of cyclin D1, and increased expression of P27Kip1 in normal but not in transformed mammary epithelial cells. J Cell Physiol 1999; 180(2): 245–54. doi: 10.1002/(SICI)1097-4652(199908)180:2<245::AID-JCP12>3.0.CO;2-R
  93. Yan YY, Lin J, Ou TM, Tan JH, Li D, Gu LQ, et al. Selective recognition of oncogene promoter G-quadruplexes by Mg2+. Biochem Biophys Res Commun 2010; 402(4): 614–8. doi: 10.1016/j.bbrc.2010.10.065
  94. Yee NS, Kazi AA, Li Q, Yang Z, Berg A, Yee RK. Aberrant over-expression of TRPM7 ion channels in pancreatic cancer: required for cancer cell invasion and implicated in tumor growth and metastasis. Biology Open 2015; 4(4): 507–14. doi: 10.1242/bio.20137088
  95. Yee NS. Role of TRPM7 in cancer: potential as molecular biomarker and therapeutic target. Pharmaceuticals 2017; 10(2): 39. doi: 10.3390/ph10020039
  96. Hardy S, Uetani N, Wong N, Kostantin E, Labbé DP, Bégin LR, et al. The protein tyrosine phosphatase PRL-2 interacts with the magnesium transporter CNNM3 to promote oncogenesis. Oncogene 2015; 34(8): 986–95. doi: 10.1038/onc.2014.33
  97. Tangvoraphonkchai K, Davenport A. Magnesium and cardiovascular disease. Adv Chronic Kidney Dis 2018; 25(3): 251–60. doi: 10.1053/j.ackd.2018.02.010
  98. Kolte D, Vijayaraghavan K, Khera S, Sica DA, Frishman WH. Role of magnesium in cardiovascular diseases. Cardiol Rev 2014; 22(4): 182–92. doi: 10.1097/CRD.0000000000000003
  99. Bibawy JN, Parikh V, Wahba J, Barsoum EA, Lafferty J, Kowalski M, et al. Pantoprazole (proton pump inhibitor) contributing to Torsades de Pointes storm. Circ Arrhythm Electrophysiol 2013; 6(2): e17–9. doi: 10.1161/CIRCEP.112.000101
  100. Khan N, Gray IP, Obejero-Paz CA, Jones SW. Permeation and gating in CaV3.1 (alpha1G) T-type calcium channels effects of Ca2+, Ba2+, Mg2+, and Na+. J Gen Physiol 2008; 132(2): 223–38. doi: 10.1085/jgp.200809986
  101. Romani AM. Beneficial role of Mg2+ in prevention and treatment of hypertension. Int J Hypertens 2018; 2018: 9013721. doi: 10.1155/2018/9013721
  102. Lin J, Zhou S, Zhao T, Ju T, Zhang L. TRPM7 channel regulates ox-LDL-induced proliferation and migration of vascular smooth muscle cells via MEK-ERK pathways. FEBS Lett 2016; 590(4): 520–32. doi: 10.1002/1873-3468.12088
  103. Antunes TT, Callera GE, He Y, Yogi A, Ryazanov AG, Ryazanova LV, et al. Transient receptor potential melastatin 7 cation channel kinase: new player in angiotensin II-induced hypertension. Hypertension 2016; 67(4): 763–73. doi: 10.1161/HYPERTENSIONAHA.115.07021
  104. Satake K, Lee JD, Shimizu H, Uzui H, Mitsuke Y, Yue H, et al. Effects of magnesium on prostacyclin synthesis and intracellular free calcium concentration in vascular cells. Magnes Res 2004; 17(1): 20–7.
  105. White RE, Hartzell HC. Effects of intracellular free magnesium on calcium current in isolated cardiac myocytes. Science 1988; 239(4841 Pt 1): 778–80. doi: 10.1126/science.2448878
  106. Maier JA, Bernardini D, Rayssiguier Y, Mazur A. High concentrations of magnesium modulate vascular endothelial cell behaviour in vitro. Biochim Biophys Acta 2004; 1689(1): 6–12. doi: 10.1016/j.bbadis.2004.02.004
  107. Sobhani AR, Farshidi H, Azarkish F, Eslami M, Eftekhar E, Keshavarz M, et al. Magnesium sulfate improves some risk factors for atherosclerosis in patients suffering from one or two coronary artery diseases: a double-blind clinical trial study. Clin Pharmacol 2020; 12: 159–69. doi: 10.2147/CPAA.S261264
  108. Chistiakov DA, Myasoedova VA, Melnichenko AA, Grechko AV, Orekhov AN. Calcifying matrix vesicles and atherosclerosis. Biomed Res Int 2017; 2017: 7463590. doi: 10.1155/2017/7463590
  109. Montezano AC, Zimmerman D, Yusuf H, Burger D, Chignalia AZ, Wadhera V, et al. Vascular smooth muscle cell differentiation to an osteogenic phenotype involves TRPM7 modulation by magnesium. Hypertension 2010; 56(3): 453–62. doi: 10.1161/HYPERTENSIONAHA.110.152058
  110. Liu M, Liu H, Feng F, Xie A, Kang GJ, Zhao Y, et al. Magnesium deficiency causes a reversible, metabolic, diastolic cardiomyopathy. J Am Heart Assoc 2021; 10(12): e020205. doi: 10.1161/JAHA.120.020205
  111. Slutsky I, Abumaria N, Wu LJ, Huang C, Zhang L, Li B, et al. Enhancement of learning and memory by elevating brain magnesium. Neuron 2010; 65(2): 165–77. doi: 10.1016/j.neuron.2009.12.026
  112. Wilson C, González-Billault C. Regulation of cytoskeletal dynamics by redox signaling and oxidative stress: implications for neuronal development and trafficking. Front Cell Neurosci 2015; 9: 381. doi: 10.3389/fncel.2015.00381
  113. Turlova E, Bae CYJ, Deurloo M, Chen W, Barszczyk A, Horgen FD, et al. TRPM7 regulates axonal outgrowth and maturation of primary hippocampal neurons. Mol Neurobiol 2016; 53(1): 595–610. doi: 10.1007/s12035-014-9032-y
  114. Zeng Z, Leng T, Feng X, Sun H, Inoue K, Zhu L, et al. Silencing TRPM7 in mouse cortical astrocytes impairs cell proliferation and migration via ERK and JNK signaling pathways. PLoS One 2015; 10(3): e0119912. doi: 10.1371/journal.pone.0119912
  115. Cilliler AE, Ozturk S, Ozbakir S. Serum magnesium level and clinical deterioration in Alzheimer’s disease. Gerontology 2007; 53(6): 419–22. doi: 10.1159/000110873
  116. Lin L, Yan M, Wu B, Lin R, Zheng Z. Expression of magnesium transporter SLC41A1 in the striatum of 6-hydroxydopamine-induced parkinsonian rats. Brain Res Bull 2018; 142: 338–43. doi: 10.1016/j.brainresbull.2018.08.019
  117. Kuramoto T, Kuwamura M, Tokuda S, Izawa T, Nakane Y, Kitada K, et al. A mutation in the gene encoding mitochondrial Mg²+ channel MRS2 results in demyelination in the rat. PLoS Genet 2011; 7(1): e1001262. doi: 10.1371/journal.pgen.1001262
  118. Schoenen J, Sianard-Gainko J, Lenaerts M. Blood magnesium levels in migraine. Cephalalgia 1991; 11(2): 97–9. doi: 10.1046/j.1468-2982.1991.1102097.x
  119. Altura BT, Memon ZI, Zhang A, Cheng TP, Silverman R, Cracco RQ, et al. Low levels of serum ionized magnesium are found in patients early after stroke which result in rapid elevation in cytosolic free calcium and spasm in cerebral vascular muscle cells. Neurosci Lett 1997; 230(1): 37–40. doi: 10.1016/S0304-3940(97)00471-0
  120. Sendur OF, Tastaban E, Turan Y, Ulman C. The relationship between serum trace element levels and clinical parameters in patients with fibromyalgia. Rheumatol Int 2008; 28(11): 1117–21. doi: 10.1007/s00296-008-0593-9
  121. Veronese N, Berton L, Carraro S, Bolzetta F, De Rui M, Perissinotto E, et al. Effect of oral magnesium supplementation on physical performance in healthy elderly women involved in a weekly exercise program: a randomized controlled trial. Am J Clin Nutr 2014; 100(3): 974–81. doi: 10.3945/ajcn.113.080168
  122. Uwitonze AM, Razzaque MS. Role of magnesium in vitamin D activation and function. J Am Osteopath Assoc 2018; 118(3): 181–9. doi: 10.7556/jaoa.2018.037
  123. Dos Santos LR, Melo SRS, Severo JS, Morais JBS, da Silva LD, de Paiva Sousa M, et al. Cardiovascular diseases in obesity: what is the role of magnesium? Biol Trace Elem Res 2021; 199(11): 4020–7. doi: 10.1007/s12011-020-02528-7
  124. Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M, et al. PPARγ signaling and metabolism: the good, the bad and the future. Nat Med 2013; 19(5): 557–66. doi: 10.1038/nm.3159
  125. Wei CC, Wu K, Gao Y, Zhang LH, Li DD, Luo Z. Magnesium reduces hepatic lipid accumulation in yellow catfish (Pelteobagrus fulvidraco) and modulates lipogenesis and lipolysis via PPARA, JAK-STAT, and AMPK pathways in hepatocytes. J Nutr 2017; 147(6): 1070–8. doi: 10.3945/jn.116.245852
  126. Volpe SL. Magnesium, the metabolic syndrome, insulin resistance, and type 2 diabetes mellitus. Crit Rev Food Sci Nutr 2008; 48(3): 293–300. doi: 10.1080/10408390701326235
  127. Guerrero-Romero F, Jaquez-Chairez FO, Rodríguez-Morán M. Magnesium in metabolic syndrome: a review based on randomized, double-blind clinical trials. Magnes Res 2016; 29(4): 146–53. doi: 10.1684/mrh.2016.0404
  128. Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, et al. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med 2020; 382(8): 727–33. doi: 10.1056/NEJMoa2001017
  129. Cooper ID, Crofts CAP, DiNicolantonio JJ, Malhotra A, Elliott B, Kyriakidou Y, et al. Relationships between hyperinsulinaemia, magnesium, vitamin D, thrombosis and COVID-19: rationale for clinical management. Open Heart 2020; 7(2): e001356. doi: 10.1136/openhrt-2020-001356
  130. Zeng F, Deng G, Cui Y, Zhang Y, Dai M, Chen L, et al. A predictive model for the severity of COVID-19 in elderly patients. Aging 2020; 12(21): 20982–96. doi: 10.18632/aging.103980
  131. Iotti S, Wolf F, Mazur A, Maier JA. The COVID-19 pandemic: is there a role for magnesium? Hypotheses and perspectives. Magnes Res 2020;33(2):21–27.
  132. Hansen BA, Bruserud Ø, Tang CF, Ding H, Jiao RQ, Wu XX, et al. Hypomagnesemia in critically ill patients. J Intensive Care 2018; 6: 21. doi: 10.1186/s40560-018-0291-y
  133. Clerkin KJ, Fried JA, Raikhelkar J, Sayer G, Griffin JM, Masoumi A, et al. COVID-19 and cardiovascular disease. Circulation 2020; 141(20): 1648–55. doi: 10.1161/CIRCULATIONAHA.120.046941
  134. Çiçek G, Açıkgoz SK, Yayla Ç, Kundi H, İleri M. Magnesium as a predictor of acute stent thrombosis in patients with ST-segment elevation myocardial infarction who underwent primary angioplasty. Coron Artery Dis 2016; 27(1): 47–51. doi: 10.1097/MCA.0000000000000318
  135. Zhu D, You J, Zhao N, Xu H. Magnesium regulates endothelial barrier functions through TRPM7, MagT1, and S1P1. Adv Sci 2019; 6(18): 1901166. doi: 10.1002/advs.201901166
  136. Liu PP, Blet A, Smyth D, Li H. The science underlying COVID-19: implications for the cardiovascular system. Circulation 2020; 142(1): 68–78. doi: 10.1161/CIRCULATIONAHA.120.047549
  137. Tang CF, Ding H, Jiao RQ, Wu XX, Kong LD. Possibility of magnesium supplementation for supportive treatment in patients with COVID-19. Eur J Pharmacol 2020; 886: 173546. doi: 10.1016/j.ejphar.2020.173546