Magnesium and the Hallmarks of Aging
Abstract
:1. Introduction
2. Cellular Magnesium Homeostasis
3. Biology of Aging
4. Magnesium and Aging
5. Magnesium Connections with the Hallmarks of Aging
5.1. Genomic Instability
5.2. Telomere Attrition
5.3. Epigenetic Alterations
5.4. Mitochondrial Dysfunction
5.5. Loss of Proteostasis
5.6. Deregulated Nutrient Sensing
5.7. Cellular Senescence
5.8. Stem Cell Exhaustion
5.9. Altered Intercellular Communication
5.10. Compromised Autophagy
5.11. Dysbiosis
5.12. Inflammation
6. Concluding Remarks
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Beard, J.R.; Officer, A.; de Carvalho, I.A.; Sadana, R.; Pot, A.M.; Michel, J.P.; Lloyd-Sherlock, P.; Epping-Jordan, J.E.; Peeters, G.M.E.E.; Mahanani, W.R.; et al. The World report on ageing and health: A policy framework for healthy ageing. Lancet 2016, 387, 2145–2154. [Google Scholar] [CrossRef] [PubMed]
- Foreman, K.J.; Marquez, N.; Dolgert, A.; Fukutaki, K.; Fullman, N.; McGaughey, M.; Pletcher, M.A.; Smith, A.E.; Tang, K.; Yuan, C.-W.; et al. Forecasting life expectancy, years of life lost, and all-cause and cause-specific mortality for 250 causes of death: Reference and alternative scenarios for 2016-40 for 195 countries and territories. Lancet 2018, 392, 2052–2090. [Google Scholar] [CrossRef] [PubMed]
- Kuo, P.-L.; Schrack, J.A.; Levine, M.E.; Shardell, M.D.; Simonsick, E.M.; Chia, C.W.; Moore, A.Z.; Tanaka, T.; An, Y.; Karikkineth, A.; et al. Longitudinal phenotypic aging metrics in the Baltimore Longitudinal Study of Aging. Nat. Aging 2022, 2, 635–643. [Google Scholar] [CrossRef] [PubMed]
- Fabbri, E.; Zoli, M.; Gonzalez-Freire, M.; Salive, M.E.; Studenski, S.A.; Ferrucci, L. Aging and Multimorbidity: New Tasks, Priorities, and Frontiers for Integrated Gerontological and Clinical Research. J. Am. Med. Dir. Assoc. 2015, 16, 640–647. [Google Scholar] [CrossRef] [PubMed]
- Caspi, R.; Altman, T.; Dreher, K.; Fulcher, C.A.; Subhraveti, P.; Keseler, I.M.; Kothari, A.; Krummenacker, M.; Latendresse, M.; Mueller, L.A.; et al. The MetaCyc database of metabolic pathways and enzymes and the BioCyc collection of pathway/genome databases. Nucleic Acids Res. 2012, 40, D742–D753. [Google Scholar] [CrossRef] [PubMed]
- Saris, N.-E.L.; Mervaala, E.; Karppanen, H.; Khawaja, J.A.; Lewenstam, A. Magnesium: An update on physiological, clinical and analytical aspects. Clin. Chim. Acta 2000, 294, 1–26. [Google Scholar] [CrossRef]
- Barbagallo, M.; Gupta, R.K.; Dominguez, L.J.; Resnick, L.M. Cellular ionic alterations with age: Relation to hypertension and diabetes. J. Am. Geriatr. Soc. 2000, 48, 1111–1116. [Google Scholar] [CrossRef]
- Barbagallo, M.; Veronese, N.; Dominguez, L.J. Magnesium in Aging, Health and Diseases. Nutrients 2021, 13, 463. [Google Scholar] [CrossRef]
- Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef]
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. Hallmarks of aging: An expanding universe. Cell 2023, 186, 243–278. [Google Scholar] [CrossRef]
- Schmauck-Medina, T.; Molière, A.; Lautrup, S.; Zhang, J.; Chlopicki, S.; Madsen, H.B.; Cao, S.; Soendenbroe, C.; Mansell, E.; Vestergaard, M.B.; et al. New hallmarks of ageing: A 2022 Copenhagen ageing meeting summary. Aging 2022, 14, 6829–6839. [Google Scholar] [CrossRef]
- Barbagallo, M.; Veronese, N.; Dominguez, L.J. Magnesium in Type 2 Diabetes Mellitus, Obesity, and Metabolic Syndrome. Nutrients 2022, 14, 714. [Google Scholar] [CrossRef]
- Gröber, U.; Schmidt, J.; Kisters, K. Magnesium in Prevention and Therapy. Nutrients 2015, 7, 8199–8226. [Google Scholar] [CrossRef]
- Barbagallo, M.; Dominguez, L.J.; Galioto, A.; Ferlisi, A.; Cani, C.; Malfa, L.; Pineo, A.; Busardo’, A.; Paolisso, G. Role of magnesium in insulin action, diabetes and cardio-metabolic syndrome X. Mol. Asp. Med. 2003, 24, 39–52. [Google Scholar] [CrossRef]
- Gupta, R.; Benovic, J.; Rose, Z. The determination of the free magnesium level in the human red blood cell by 31P NMR. J. Biol. Chem. 1978, 253, 6172–6176. [Google Scholar] [CrossRef] [PubMed]
- Pan, M.-R.; Li, K.; Lin, S.-Y.; Hung, W.-C. Connecting the Dots: From DNA Damage and Repair to Aging. Int. J. Mol. Sci. 2016, 17, 685. [Google Scholar] [CrossRef]
- Ford, E.S.; Mokdad, A.H. Dietary magnesium intake in a national sample of US adults. J. Nutr. 2003, 133, 2879–2882. [Google Scholar] [CrossRef] [PubMed]
- Resnick, L.M.; Gupta, R.K.; Laragh, J.H. Intracellular free magnesium in erythrocytes of essential hypertension: Relation to blood pressure and serum divalent cations. Proc. Natl. Acad. Sci. USA 1984, 81, 6511–6515. [Google Scholar] [CrossRef] [PubMed]
- Resnick, L.M.; Gupta, R.K.; Laragh, J.H.; Alderman, M.H.; Altura, B.M. Intracellular and extracellular magnesium depletion in Type 2 (non-insulin-dependent) diabetes mellitus. Diabetologia 1993, 36, 767–770. [Google Scholar] [CrossRef] [PubMed]
- Mathers, J.C. Impact of nutrition on the ageing process. Br. J. Nutr. 2015, 113, S18–S22. [Google Scholar] [CrossRef] [PubMed]
- Aguilera, A.; Gómez-González, B. Genome instability: A mechanistic view of its causes and consequences. Nat. Rev. Genet. 2008, 9, 204–217. [Google Scholar] [CrossRef] [PubMed]
- Jackson, S.P.; Bartek, J. The DNA-damage response in human biology and disease. Nature 2009, 461, 1071–1078. [Google Scholar] [CrossRef] [PubMed]
- Hoeijmakers, J.H. DNA damage, aging, and cancer. N. Engl. J. Med. 2009, 361, 1475–1485. [Google Scholar] [CrossRef] [PubMed]
- Laconi, E.; Marongiu, F.; DeGregori, J. Cancer as a disease of old age: Changing mutational and microenvironmental landscapes. Br. J. Cancer 2020, 122, 943–952. [Google Scholar] [CrossRef]
- Chiu, T.K.; Dickerson, R.E. 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, 915–945. [Google Scholar] [CrossRef]
- Sirover, M.A.; Loeb, L.A. Metal activation of DNA synthesis. Biochem. Biophys. Res. Commun. 1976, 70, 812–817. [Google Scholar] [CrossRef]
- 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, 4232. [Google Scholar] [CrossRef]
- Price, M.A.; Tullius, T.D. Using hydroxyl radical to probe DNA structure. Methods Enzymol. 1992, 212, 194–219. [Google Scholar]
- SantaLucia, J., Jr.; Hicks, D. The thermodynamics of DNA structural motifs. Annu. Rev. Biophys. Biomol. Struct. 2004, 33, 415–440. [Google Scholar] [CrossRef]
- Bui, V.; Nguyen, T. DNA aggregation induced by Mg2+ ions under different conditions. J. Mol. Recognit. 2018, 31, e2721. [Google Scholar] [CrossRef]
- 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, 79–91. [Google Scholar] [CrossRef] [PubMed]
- Kielar, C.; Xin, Y.; Shen, B.; Kostiainen, M.A.; Grundmeier, G.; Linko, V.; Keller, A. On the Stability of DNA Origami Nanostructures in Low-Magnesium Buffers. Angew. Chem. Int. Ed Engl. 2018, 57, 9470–9474. [Google Scholar] [CrossRef] [PubMed]
- Calsou, P.; Salles, B. Properties of damage-dependent DNA incision by nucleotide excision repair in human cell-free extracts. Nucleic Acids Res. 1994, 22, 4937–4942. [Google Scholar] [CrossRef] [PubMed]
- Ban, C.; Junop, M.; Yang, W. Transformation of MutL by ATP binding and hydrolysis: A switch in DNA mismatch repair. Cell 1999, 97, 85–97. [Google Scholar] [CrossRef]
- Hartwig, A. Role of magnesium in genomic stability. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2001, 475, 113–121. [Google Scholar] [CrossRef]
- Blackburn, E.H.; Epel, E.S.; Lin, J. Human telomere biology: A contributory and interactive factor in aging, disease risks, and protection. Science 2015, 350, 1193–1198. [Google Scholar] [CrossRef]
- Saretzki, G. Telomeres, Telomerase and Ageing. Subcell. Biochem. 2018, 90, 221–308. [Google Scholar]
- Herrmann, M.; Pusceddu, I.; März, W.; Herrmann, W. Telomere biology and age-related diseases. CCLM 2018, 56, 1210–1222. [Google Scholar] [CrossRef]
- von Zglinicki, T. Oxidative stress shortens telomeres. Trends Biochem. Sci. 2002, 27, 339–344. [Google Scholar] [CrossRef]
- Yadav, S.; Maurya, P.K. Correlation Between Telomere Length and Biomarkers of Oxidative Stress in Human Aging. Rejuvenation Res. 2022, 25, 25–29. [Google Scholar] [CrossRef]
- Calado, R.T.; Young, N.S. Telomere diseases. N. Engl. J. Med. 2009, 361, 2353–2365. [Google Scholar] [CrossRef]
- Boonekamp, J.J.; Simons, M.J.P.; Hemerik, L.; Verhulst, S. Telomere length behaves as biomarker of somatic redundancy rather than biological age. Aging Cell 2013, 12, 330–332. [Google Scholar] [CrossRef]
- Astuti, Y.; Wardhana, A.; Watkins, J.; Wulaningsih, W. Cigarette smoking and telomere length: A systematic review of 84 studies and meta-analysis. Environ. Res. 2017, 158, 480–489. [Google Scholar] [CrossRef]
- Arsenis, N.C.; You, T.; Ogawa, E.F.; Tinsley, G.M.; Zuo, L. Physical activity and telomere length: Impact of aging and potential mechanisms of action. Oncotarget 2017, 8, 45008–45019. [Google Scholar] [CrossRef]
- Ganesh, S.; Qin, Z.; Spagnol, S.T.; Biegler, M.T.; Coffey, K.A.; Kalinowski, A.; Buehler, M.J.; Dahl, K.N. The tail domain of lamin B1 is more strongly modulated by divalent cations than lamin A. Nucleus 2015, 6, 203–211. [Google Scholar] [CrossRef]
- Zvereva, M.I.; Shcherbakova, D.M.; Dontsova, O.A. Telomerase: Structure, functions, and activity regulation. Biochemistry 2010, 75, 1563–1583. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Ding, D.; Wang, M.; Cong, Y.-S. Telomerase reverse transcriptase in the regulation of gene expression. BMB Rep. 2014, 47, 8–14. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Wang, Q.; Zhang, Z.; Fu, R.; Zhou, T.; Long, C.; He, T.; Yang, D.; Li, Z.; Peng, S. Magnesium supplementation enhances mTOR signalling to facilitate myogenic differentiation and improve aged muscle performance. Bone 2021, 146, 115886. [Google Scholar] [CrossRef] [PubMed]
- Feeney, K.A.; Hansen, L.L.; Putker, M.; Olivares-Yañez, C.; Day, J.; Eades, L.J.; Larrondo, L.F.; Hoyle, N.P.; O’neill, J.S.; van Ooijen, G. Daily magnesium fluxes regulate cellular timekeeping and energy balance. Nature 2016, 532, 375–379. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.-D.; Wen, M.-S.; Shie, S.-S.; Lo, Y.-L.; Wo, H.-T.; Wang, C.-C.; Hsieh, I.-C.; Lee, T.-H.; Wang, C.-Y. The circadian rhythm controls telomeres and telomerase activity. Biochem. Biophys. Res. Commun. 2014, 451, 408–414. [Google Scholar] [CrossRef]
- Pal, S.; Tyler, J.K. Epigenetics and aging. Sci. Adv. 2016, 2, e1600584. [Google Scholar] [CrossRef] [PubMed]
- Yu, M.; Hazelton, W.D.; Luebeck, G.E.; Grady, W.M. Epigenetic Aging: More Than Just a Clock When it Comes to Cancer. Cancer Res. 2020, 80, 367–374. [Google Scholar] [CrossRef] [PubMed]
- Feinberg, A.P.; Levchenko, A. Epigenetics as a mediator of plasticity in cancer. Science 2023, 379, eaaw3835. [Google Scholar] [CrossRef] [PubMed]
- Cora, D.; Re, A.; Caselle, M.; Bussolino, F. MicroRNA-mediated regulatory circuits: Outlook and perspectives. Phys. Biol. 2017, 14, 045001. [Google Scholar] [CrossRef] [PubMed]
- 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, 573–578. [Google Scholar] [CrossRef] [PubMed]
- Newell-Price, J.; Clark, A.J.; King, P. DNA methylation and silencing of gene expression. Trends Endocrinol. Metab. 2000, 11, 142–148. [Google Scholar] [CrossRef]
- Chacko, S.A.; Sul, J.; Song, Y.; Li, X.; LeBlanc, J.; You, Y.; Butch, A.; Liu, S. Magnesium supplementation, metabolic and inflammatory markers, and global genomic and proteomic profiling: A randomized, double-blind, controlled, crossover trial in overweight individuals. Am. J. Clin. Nutr. 2011, 93, 463–473. [Google Scholar] [CrossRef]
- Takaya, J.; Iharada, A.; Okihana, H.; Kaneko, K. A calcium-deficient diet in pregnant, nursing rats induces hypomethylation of specific cytosines in the 11beta-hydroxysteroid dehydrogenase-1 promoter in pup liver. Nutr. Res. 2013, 33, 961–970. [Google Scholar] [CrossRef]
- Hruby, A.; McKeown, N.M.; Song, Y.; Djoussé, L. Dietary magnesium and genetic interactions in diabetes and related risk factors: A brief overview of current knowledge. Nutrients 2013, 5, 4990–5011. [Google Scholar] [CrossRef] [PubMed]
- Shah, N.C.; Shah, G.J.; Li, Z.; Jiang, X.C.; Altura, B.T.; Altura, B.M. Short-term magnesium deficiency downregulates telomerase, up-regulates neutral sphingomyelinase and induces oxidative DNA damage in cardiovascular tissues: Relevance to atherogenesis, cardiovascular diseases and aging. Int. J. Clin. Exp. Med. 2014, 7, 497–514. [Google Scholar] [PubMed]
- McBride, H.M.; Neuspiel, M.; Wasiak, S. Mitochondria: More than just a powerhouse. Curr. Biol. 2006, 16, R551–R560. [Google Scholar] [CrossRef]
- Hekimi, S.; Lapointe, J.; Wen, Y. Taking a “good” look at free radicals in the aging process. Trends Cell Biol. 2011, 21, 569–576. [Google Scholar] [CrossRef] [PubMed]
- Park, J.-S.; Davis, R.L.; Sue, C.M. Mitochondrial Dysfunction in Parkinson’s Disease: New Mechanistic Insights and Therapeutic Perspectives. Curr. Neurol. Neurosci. Rep. 2018, 18, 21. [Google Scholar] [CrossRef] [PubMed]
- Birnbaum, J.H.; Wanner, D.; Gietl, A.F.; Saake, A.; Kundig, T.M.; Hock, C.; Nitsch, R.M.; Tackenberg, C. Oxidative stress and altered mitochondrial protein expression in the absence of amyloid-beta and tau pathology in iPSC-derived neurons from sporadic Alzheimer’s disease pa-tients. Stem Cell Res. 2018, 27, 121–130. [Google Scholar] [CrossRef] [PubMed]
- Whitehall, J.C.; Greaves, L.C. Aberrant mitochondrial function in ageing and cancer. Biogerontology 2020, 21, 445–459. [Google Scholar] [CrossRef] [PubMed]
- Romani, A.; Marfella, C.; Scarpa, A. Cell magnesium transport and homeostasis: Role of intracellular compartments. Miner. Electrolyte Metab. 1993, 19, 282–289. [Google Scholar] [PubMed]
- Zsurka, G.; Gregáň, J.; Schweyen, R.J. The human mitochondrial Mrs2 protein functionally substitutes for its yeast homologue, a candidate magnesium transporter. Genomics 2001, 72, 158–168. [Google Scholar] [CrossRef] [PubMed]
- Garfinkel, L.; Garfinkel, D. Magnesium regulation of the glycolytic pathway and the enzymes involved. Magnesium 1985, 4, 60–72. [Google Scholar] [PubMed]
- Soulimane, T.; Buse, G. Integral cytochrome-c oxidase. Preparation and progress towards a three-dimensional crystallization. Eur. J. Biochem. 1995, 227, 588–595. [Google Scholar] [CrossRef]
- Yamanaka, R.; Tabata, S.; Shindo, Y.; Hotta, K.; Suzuki, K.; Soga, T.; Oka, K. Mitochondrial Mg2+ homeostasis decides cellular energy metabolism and vulnerability to stress. Sci. Rep. 2016, 6, 30027. [Google Scholar] [CrossRef]
- Pilchova, I.; Klacanova, K.; Tatarkova, Z.; Kaplan, P.; Racay, P. The Involvement of Mg2+ in Regulation of Cellular and Mitochondrial Functions. Oxidative Med. Cell. Longev. 2017, 2017, 6797460. [Google Scholar] [CrossRef] [PubMed]
- Resnick, L.M.; Barbagallo, M.; Dominguez, L.J.; Veniero, J.M.; Nicholson, J.P.; Gupta, R.K. Relation of cellular potassium to other mineral ions in hypertension and diabetes. Hypertension 2001, 38 Pt 2, 709–712. [Google Scholar] [CrossRef]
- Dominguez, L.J.; Veronese, N.; Barbagallo, M. Magnesium and Hypertension in Old Age. Nutrients 2020, 13, 139. [Google Scholar] [CrossRef] [PubMed]
- Gout, E.; Rebeille, F.; Douce, R.; Bligny, R. Interplay of Mg2+, ADP, and ATP in the cytosol and mitochondria: Unravelling the role of Mg2+ in cell respiration. Proc. Natl. Acad. Sci. USA 2014, 111, E4560–E4567. [Google Scholar] [CrossRef]
- Panov, A.; Scarpa, A. Mg2+ control of respiration in isolated rat liver mitochondria. Biochemistry. Biochemistry 1996, 35, 12849–12856. [Google Scholar] [CrossRef]
- Rodríguez-Zavala, J.S.; Moreno-Sánchez, R. Modulation of oxidative phosphorylation by Mg2+ in rat heart mitochondria. J. Biol. Chem. 1998, 273, 7850–7855. [Google Scholar] [CrossRef] [PubMed]
- Morais, J.B.; Severo, J.S.; Santos, L.R.; de Sousa Melo, S.R.; de Oliveira Santos, R.; de Oliveira, A.R.; Cruz, K.J.C.; Marreiro, D.D.N. Role of Magnesium in Oxidative Stress in Individuals with Obesity. Biol. Trace Elem. Res. 2017, 176, 20–26. [Google Scholar] [CrossRef]
- Calviello, G.; Ricci, P.; Lauro, L.; Palozza, P.; Cittadini, A. Mg deficiency induces mineral content changes and oxidative stress in rats. Biochem. Mol. Biol. Int. 1994, 32, 903–911. [Google Scholar]
- Shah, N.C.; Liu, J.-P.; Iqbal, J.; Hussain, M.; Jiang, X.-C.; Li, Z.; Li, Y.; Zheng, T.; Li, W.; Sica, A.C.; et al. Mg deficiency results in modulation of serum lipids, glutathione, and NO synthase isozyme activation in cardiovascular tissues: Relevance to de novo synthesis of ceramide, serum Mg and atherogenesis. Int. J. Clin. Exp. Med. 2011, 4, 103–118. [Google Scholar]
- Kumar, B.P.; Shivakumar, K. Depressed antioxidant defense in rat heart in experimental magnesium deficiency implications for the pathogenesis of myocardial lesions. Biol. Trace Element Res. 1997, 60, 139–144. [Google Scholar] [CrossRef]
- Liu, M.; Dudley, S.C., Jr. Magnesium, Oxidative Stress, Inflammation, and Cardiovascular Disease. Antioxidants 2020, 9, 907. [Google Scholar] [CrossRef]
- Liu, M.; Jeong, E.-M.; Liu, H.; Xie, A.; So, E.Y.; Shi, G.; Jeong, G.E.; Zhou, A.; Dudley, S.C. Magnesium supplementation improves diabetic mitochondrial and cardiac diastolic function. J. Clin. Investig. 2019, 4, e123182. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Liu, H.; Feng, F.; Xie, A.; Kang, G.; Zhao, Y.; Hou, C.R.; Zhou, X.; Dudley, S.C., Jr. Magnesium Deficiency Causes a Reversible, Metabolic, Diastolic Cardiomyopathy. J. Am. Heart Assoc. 2021, 10, e020205. [Google Scholar] [CrossRef]
- Kramer, J.H.; Mišík, V.; Weglicki, W.B. Magnesium-deficiency potentiates free radical production associated with postischemic injury to rat hearts: Vitamin E affords protection. Free Radic. Biol. Med. 1994, 16, 713–723. [Google Scholar] [CrossRef]
- Chen, Y.; Wei, X.; Yan, P.; Han, Y.; Sun, S.; Wu, K.; Fan, D. Human mitochondrial Mrs2 protein promotes multidrug resistance in gastric cancer cells by regulating p27, cyclin D1 expression and cytochrome C release. Cancer Biol. Ther. 2009, 8, 607–614. [Google Scholar] [CrossRef]
- Salvi, M.; Bozac, A.; Toninello, A. Gliotoxin induces Mg2+ efflux from intact brain mitochondria. Neurochem. Int. 2004, 45, 759–764. [Google Scholar] [CrossRef]
- Sponder, G.; Abdulhanan, N.; Frohlich, N.; Mastrototaro, L.; Aschenbach, J.R.; Rontgen, M.; Pilchova, I.; Cibulka, M.; Racay, P.; Kolisek, M. Overexpression of Na(+)/Mg2+exchanger SLC41A1 attenuates pro-survival signaling. Oncotarget 2018, 9, 5084–5104. [Google Scholar] [CrossRef] [PubMed]
- Racay, P. Effect of magnesium on calcium-induced depolarisation of mitochondrial transmembrane potential. Cell Biol. Int. 2008, 32, 136–145. [Google Scholar] [CrossRef] [PubMed]
- Blomeyer, C.A.; Bazil, J.N.; Stowe, D.F.; Dash, R.K.; Camara, A.K.S. Mg2+ differentially regulates two modes of mitochondrial Ca2+ uptake in isolated cardiac mitochondria: Implications for mitochondrial Ca2+ sequestration. J. Bioenerg. Biomembr. 2016, 48, 175–188. [Google Scholar] [CrossRef]
- Bednarczyk, P.; Dolowy, K.; Szewczyk, A. Matrix Mg2+ regulates mitochondrial ATP-dependent potassium channel from heart. FEBS Lett. 2005, 579, 1625–1632. [Google Scholar] [CrossRef]
- Zoratti, M.; Szabo, I. The mitochondrial permeability transition. Biochim. Biophys Acta. 1995, 1241, 139–176. [Google Scholar] [CrossRef]
- Beavis, A.D.; Powers, M.F. On the regulation of the mitochondrial inner membrane anion channel by magnesium and protons. J. Biol. Chem. 1989, 264, 17148–17155. [Google Scholar] [CrossRef] [PubMed]
- Sharikabad, M.N.; Ostbye, K.M.; Brors, O. Increased [Mg2+]o reduces Ca2+ influx and disruption of mitochondrial membrane potential during reoxygenation. Am. J. Physiol. Heart Circ. Physiol. 2001, 281, H2113–H2123. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.Y.; Hsieh, Y.L.; Ju, D.T.; Lin, C.C.; Kuo, C.H.; Liou, Y.F.; Ho, T.-J.; Tsai, C.-H.; Tsai, F.-J.; Lin, J.-Y. Attenuation of Magnesium Sulfate on CoCl(2)-Induced Cell Death by Activating ERK1/2/MAPK and Inhibiting HIF-1alpha via Mitochondrial Apoptotic Signaling Suppression in a Neuronal Cell Line. Chin. J. Physiol. 2015, 58, 244–253. [Google Scholar] [CrossRef] [PubMed]
- Gorgoglione, V.; Laraspata, D.; La Piana, G.; Marzulli, D.; Lofrumento, N.E. Protective effect of magnesium and potassium ions on the permeability of the external mitochondrial membrane. Arch. Biochem. Biophys. 2007, 461, 13–23. [Google Scholar] [CrossRef]
- La Piana, G.; Gorgoglione, V.; Laraspata, D.; Marzulli, D.; Lofrumento, N.E. Effect of magnesium ions on the activity of the cytosolic NADH/cytochrome c electron transport system. FEBS J. 2008, 275, 6168–6179. [Google Scholar] [CrossRef]
- Seo, Y.-W.; Na Shin, J.; Ko, K.H.; Cha, J.H.; Park, J.Y.; Lee, B.R.; Yun, C.-W.; Kim, Y.M.; Seol, D.-W.; Kim, D.-W.; et al. The molecular mechanism of Noxa-induced mitochondrial dysfunction in p53-mediated cell death. J. Biol. Chem. 2003, 278, 48292–48299. [Google Scholar] [CrossRef]
- Ferrari, R.; Albertini, A.; Curello, S.; Ceconi, C.; Dilisa, F.; Raddino, R.; Visioli, O. Myocardial recovery during post-ischaemic reperfusion: Effects of nifedipine, calcium and magnesium. J. Mol. Cell. Cardiol. 1986, 18, 487–498. [Google Scholar] [CrossRef]
- Boelens, A.D.; Pradhan, R.K.; Blomeyer, C.A.; Camara, A.K.S.; Dash, R.K.; Stowe, D.F. Extra-matrix Mg2+ limits Ca2+ uptake and modulates Ca2+ uptake–independent respiration and redox state in cardiac isolated mitochondria. J. Bioenerg. Biomembr. 2013, 45, 203–218. [Google Scholar] [CrossRef]
- Li, Y.; Wang, J.; Yue, J.; Wang, Y.; Yang, C.; Cui, Q. High magnesium prevents matrix vesicle-mediated mineralization in human bone marrow-derived mesenchymal stem cells via mitochondrial pathway and autophagy. Cell Biol. Int. 2018, 42, 205–215. [Google Scholar] [CrossRef]
- Mastrototaro, L.; Smorodchenko, A.; Aschenbach, J.R.; Kolisek, M.; Sponder, G. Solute carrier 41A3 encodes for a mitochondrial Mg2+efflux system. Sci. Rep. 2016, 6, 27999. [Google Scholar] [CrossRef]
- Su, L.-T.; Chen, H.-C.; González-Pagán, O.; Overton, J.D.; Xie, J.; Yue, L.; Runnels, L.W. TRPM7 activates m-calpain by stress-dependent stimulation of p38 MAPK and c-Jun N-terminal kinase. J. Mol. Biol. 2010, 396, 858–869. [Google Scholar] [CrossRef]
- Inoue, H.; Murayama, T.; Tashiro, M.; Sakurai, T.; Konishi, M. Mg2+- and ATP-dependent inhibition of transient receptor potential melastatin 7 by oxidative stress. Free. Radic. Biol. Med. 2014, 72, 257–266. [Google Scholar] [CrossRef]
- Villa-Bellosta, R. Dietary magnesium supplementation improves lifespan in a mouse model of progeria. EMBO Mol. Med. 2020, 12, e12423. [Google Scholar] [CrossRef]
- Dominguez, L.J.; Barbagallo, M.; Lauretani, F.; Bandinelli, S.; Bos, A.; Corsi, A.M.; Simonsick, E.M.; Ferrucci, L. Magnesium and muscle performance in older persons: The InCHIANTI study. Am. J. Clin. Nutr. 2006, 84, 419–426. [Google Scholar] [CrossRef] [PubMed]
- Welch, A.A.; Kelaiditi, E.; Jennings, A.; Steves, C.J.; Spector, T.D.; MacGregor, A. Dietary Magnesium Is Positively Associated With Skeletal Muscle Power and Indices of Muscle Mass and May Attenuate the Association Between Circulating C-Reactive Protein and Muscle Mass in Women. J. Bone Miner. Res. 2016, 31, 317–325. [Google Scholar] [CrossRef]
- Hayhoe, R.P.; Lentjes, M.A.; Mulligan, A.A.; Luben, R.N.; Khaw, K.-T.; Welch, A.A. Cross-sectional associations of dietary and circulating magnesium with skeletal muscle mass in the EPIC-Norfolk cohort. Clin. Nutr. 2019, 38, 317–323. [Google Scholar] [CrossRef] [PubMed]
- Verlaan, S.; Aspray, T.J.; Bauer, J.M.; Cederholm, T.; Hemsworth, J.; Hill, T.R.; McPhee, J.S.; Piasecki, M.; Seal, C.; Sieber, C.C.; et al. Nutritional status, body composition, and quality of life in community-dwelling sarcopenic and non-sarcopenic older adults: A case-control study. Clin. Nutr. 2017, 36, 267–274. [Google Scholar] [CrossRef] [PubMed]
- Veronese, N.; Berton, L.; Carraro, S.; Bolzetta, F.; De Rui, M.; Perissinotto, E.; Toffanello, E.D.; Bano, G.; Pizzato, S.; Miotto, F.; 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, 974–981. [Google Scholar] [CrossRef]
- Lukaski, H.C.; Nielsen, F.H. Dietary magnesium depletion affects metabolic responses during submaximal exercise in postmen-opausal women. J. Nutr. 2002, 132, 930–935. [Google Scholar] [CrossRef]
- Cameron, D.; Welch, A.A.; Adelnia, F.; Bergeron, C.M.; Reiter, D.A.; Dominguez, L.J.; Brennan, N.A.; Fishbein, K.W.; Spencer, R.G.; Ferrucci, L. Age and Muscle Function Are More Closely Associated With Intracellular Magnesium, as Assessed by 31P Magnetic Resonance Spectroscopy, Than With Serum Magnesium. Front. Physiol. 2019, 10, 1454. [Google Scholar] [CrossRef]
- van Dronkelaar, C.; van Velzen, A.; Abdelrazek, M.; van der Steen, A.; Weijs, P.J.M.; Tieland, M. Minerals and Sarcopenia; The Role of Calcium, Iron, Magnesium, Phosphorus, Potassium, Selenium, Sodium, and Zinc on Muscle Mass, Muscle Strength, and Physical Performance in Older Adults: A Systematic Review. J. Am. Med. Dir. Assoc. 2018, 19, 6–11.e3. [Google Scholar] [CrossRef]
- Labbadia, J.; Morimoto, R.I. The biology of proteostasis in aging and disease. Annu. Rev. Biochem. 2015, 84, 435–464. [Google Scholar] [CrossRef]
- Yerbury, J.J.; Ooi, L.; Dillin, A.; Saunders, D.N.; Hatters, D.M.; Beart, P.M.; Cashman, N.R.; Wilson, M.R.; Ecroyd, H. Walking the tightrope: Proteostasis and neurodegenerative disease. J. Neurochem. 2016, 137, 489–505. [Google Scholar] [CrossRef]
- Henning, R.H.; Brundel, B.J.J.M. Proteostasis in cardiac health and disease. Nat. Rev. Cardiol. 2017, 14, 637–653. [Google Scholar] [CrossRef]
- Dugger, B.N.; Dickson, D.W. Pathology of Neurodegenerative Diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef]
- Veronese, N.; Zurlo, A.; Solmi, M.; Luchini, C.; Trevisan, C.; Bano, G.; Manzato, E.; Sergi, G.; Rylander, R. Magnesium Status in Alzheimer’s Disease: A Systematic Review. Am. J. Alzheimers Dis. Other Demen. 2016, 31, 208–213. [Google Scholar] [CrossRef] [PubMed]
- Yu, J.; Sun, M.; Chen, Z.; Lu, J.; Liu, Y.; Zhou, L.; Xu, X.; Fan, D.; Chui, D. Magnesium modulates amyloid-beta protein precursor trafficking and processing. J. Alzheimers Dis. 2010, 20, 1091–1106. [Google Scholar] [CrossRef] [PubMed]
- Yu, X.; Guan, P.P.; Zhu, D.; Liang, Y.Y.; Wang, T.; Wang, Z.Y.; Wang, P. Magnesium Ions Inhibit the Expression of Tumor Necrosis Factor alpha and the Activity of gamma-Secretase in a beta-Amyloid Protein-Dependent Mechanism in APP/PS1 Transgenic Mice. Front. Mol. Neurosci. 2018, 11, 172. [Google Scholar] [CrossRef] [PubMed]
- Zhu, D.; Su, Y.; Fu, B.; Xu, H. Magnesium Reduces Blood-Brain Barrier Permeability and Regulates Amyloid-beta Transcytosis. Mol. Neurobiol. 2018, 55, 7118–7131. [Google Scholar] [CrossRef] [PubMed]
- Hansen, K.B.; Yi, F.; Perszyk, R.E.; Furukawa, H.; Wollmuth, L.P.; Gibb, A.J.; Traynelis, S.F. Structure, function, and allosteric modulation of NMDA receptors. J. Gen. Physiol. 2018, 150, 1081–1105. [Google Scholar] [CrossRef]
- Nechifor, M. Magnesium in addiction—A general view. Magnes. Res. 2018, 31, 90–98. [Google Scholar] [CrossRef]
- Hou, H.; Wang, L.; Fu, T.; Papasergi, M.; Yule, D.I.; Xia, H. Magnesium Acts as a Second Messenger in the Regulation of NMDA Re-ceptor-Mediated CREB Signaling in Neurons. Mol. Neurobiol. 2020, 57, 2539–2550. [Google Scholar] [CrossRef]
- Templeman, N.M.; Murphy, C.T. Regulation of reproduction and longevity by nutrient-sensing pathways. J. Cell Biol. 2018, 217, 93–106. [Google Scholar] [CrossRef] [PubMed]
- de Lucia, C.; Murphy, T.; Steves, C.J.; Dobson, R.J.B.; Proitsi, P.; Thuret, S. Lifestyle mediates the role of nutrient-sensing pathways in cognitive aging: Cellular and epidemiological evidence. Commun. Biol. 2020, 3, 157. [Google Scholar] [CrossRef] [PubMed]
- Kenyon, C.; Chang, J.; Gensch, E.; Rudner, A.; Tabtiang, R. A C. elegans mutant that lives twice as long as wild type. Nature 1993, 366, 461–464. [Google Scholar] [CrossRef] [PubMed]
- Puig, O.; Tjian, R. Transcriptional feedback control of insulin receptor by dFOXO/FOXO1. Minerva Anestesiol. 2005, 19, 2435–2446. [Google Scholar] [CrossRef] [PubMed]
- van der Horst, A.; Burgering, B.M. Stressing the role of FoxO proteins in lifespan and disease. Nat. Rev. Mol. Cell Biol. 2007, 8, 440–450. [Google Scholar] [CrossRef] [PubMed]
- van Heemst, D. Insulin, IGF-1 and longevity. Aging Dis. 2010, 1, 147–157. [Google Scholar] [PubMed]
- Calnan, D.R.; Brunet, A. The FoxO code. Oncogene 2008, 27, 2276–2288. [Google Scholar] [CrossRef] [PubMed]
- Johnson, S.C.; Rabinovitch, P.S.; Kaeberlein, M. mTOR is a key modulator of ageing and age-related disease. Nature 2013, 493, 338–345. [Google Scholar] [CrossRef]
- Alers, S.; Loffler, A.S.; Wesselborg, S.; Stork, B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: Cross talk, shortcuts, and feedbacks. Mol. Cell Biol. 2012, 32, 2–11. [Google Scholar] [CrossRef] [PubMed]
- Rodgers, J.T.; Lerin, C.; Haas, W.; Gygi, S.P.; Spiegelman, B.M.; Puigserver, P. Nutrient control of glucose homeostasis through a complex of PGC-1α and SIRT1. Nature 2005, 434, 113–118. [Google Scholar] [CrossRef] [PubMed]
- Barbagallo, M.; Dominguez, L.J. Magnesium metabolism in type 2 diabetes mellitus, metabolic syndrome and insulin resistance. Arch. Biochem. Biophys. 2007, 458, 40–47. [Google Scholar] [CrossRef] [PubMed]
- Barbagallo, M.; Dominguez, L.J. Magnesium and type 2 diabetes. World J. Diabetes 2015, 6, 1152–1157. [Google Scholar] [CrossRef] [PubMed]
- Mather, H.M.; Levin, G.E. Magnesium status in diabetes. Lancet 1979, 1, 924. [Google Scholar] [CrossRef] [PubMed]
- Schnack, C.; Bauer, I.; Pregant, P.; Hopmeier, P.; Schernthaner, G. Hypomagnesaemia in Type 2 (non-insulin-dependent) diabetes mellitus is not corrected by improvement of long-term metabolic control. Diabetologia 1992, 35, 77–79. [Google Scholar] [CrossRef] [PubMed]
- Barbagallo, M.; Di Bella, G.; Brucato, V.; D’angelo, D.; Damiani, P.; Monteverde, A.; Belvedere, M.; Dominguez, L.J. Serum ionized magnesium in diabetic older persons. Metabolism 2014, 63, 502–509. [Google Scholar] [CrossRef] [PubMed]
- Wälti, M.K.; Zimmermann, M.B.; Walczyk, T.; Spinas, G.A.; Hurrell, R.F. Measurement of magnesium absorption and retention in type 2 diabetic patients with the use of stable isotopes. Am. J. Clin. Nutr. 2003, 78, 448–453. [Google Scholar] [CrossRef]
- Larsson, S.C.; Wolk, A. Magnesium intake and risk of type 2 diabetes: A meta-analysis. J. Intern. Med. 2007, 262, 208–214. [Google Scholar] [CrossRef]
- Dong, J.Y.; Xun, P.; He, K.; Qin, L.Q. Magnesium intake and risk of type 2 diabetes: Meta-analysis of prospective cohort studies. Diabetes Care 2011, 34, 2116–2122. [Google Scholar] [CrossRef]
- Guerrero-Romero, F.; Rascón-Pacheco, R.A.; Rodríguez-Morán, M.; De La Peña, J.E.; Wacher, N. Hypomagnesaemia and risk for metabolic glucose disorders: A 10-year follow-up study. Eur. J. Clin. Investig. 2008, 38, 389–396. [Google Scholar] [CrossRef] [PubMed]
- Ma, B.; Lawson, A.B.; Liese, A.D.; Bell, R.A.; Mayer-Davis, E.J. Dairy, magnesium, and calcium intake in relation to insulin sensitivity: Approaches to modeling a dose-dependent association. Am. J. Epidemiol. 2006, 164, 449–458. [Google Scholar] [CrossRef]
- Song, Y.; Manson, J.E.; Buring, J.E.; Liu, S. Dietary magnesium intake in relation to plasma insulin levels and risk of type 2 diabetes in women. Diabetes Care 2004, 27, 59–65. [Google Scholar] [CrossRef]
- Lopez-Ridaura, R.; Willett, W.C.; Rimm, E.B.; Liu, S.; Stampfer, M.J.; Manson, J.E.; Hu, F.B. Magnesium intake and risk of type 2 diabetes in men and women. Diabetes Care 2004, 27, 134–140. [Google Scholar] [CrossRef]
- Kim, D.J.; Xun, P.; Liu, K.; Loria, C.; Yokota, K.; Jr, D.R.J.; He, K. Magnesium intake in relation to systemic inflammation, insulin resistance, and the incidence of diabetes. Diabetes Care 2010, 33, 2604–2610. [Google Scholar] [CrossRef] [PubMed]
- McNAIR, P.; Christensen, M.S.; Christiansen, C.; Madsbad, S.; Transbøl, I. Renal hypomagnesaemia in human diabetes mellitus: Its relation to glucose homeostasis. Eur. J. Clin. Investig. 1982, 12, 81–85. [Google Scholar] [CrossRef]
- Djurhuus, M.; Skøtt, P.; Hother-Nielsen, O.; Klitgaard, N.; Beck-Nielsen, H. Insulin increases renal magnesium excretion: A possible cause of magnesium depletion in hyperinsulinaemic states. Diabet. Med. 1995, 12, 664–669. [Google Scholar] [CrossRef]
- Veronese, N.; Dominguez, L.J.; Pizzol, D.; Demurtas, J.; Smith, L.; Barbagallo, M. Oral Magnesium Supplementation for Treating Glucose Metabolism Parameters in People with or at Risk of Diabetes: A Systematic Review and Meta-Analysis of Double-Blind Ran-domized Controlled Trials. Nutrients 2021, 13, 4074. [Google Scholar] [CrossRef]
- Veronese, N.; Demurtas, J.; Pesolillo, G.; Celotto, S.; Barnini, T.; Calusi, G.; Caruso, M.G.; Notarnicola, M.; Reddavide, R.; Stubbs, B.; et al. Magnesium and health outcomes: An umbrella review of systematic reviews and meta-analyses of observational and intervention studies. Eur. J. Nutr. 2020, 59, 263–272. [Google Scholar] [CrossRef]
- He, K.; Liu, K.; Daviglus, M.L.; Morris, S.J.; Loria, C.M.; Van Horn, L.; Jacobs, D.R.; Savage, P.J. Magnesium intake and incidence of metabolic syndrome among young adults. Circulation 2006, 113, 1675–1682. [Google Scholar] [CrossRef]
- Fung, T.T.; Manson, J.E.; Solomon, C.G.; Liu, S.; Willett, W.C.; Hu, F.B. The association between magnesium intake and fasting insulin concentration in healthy middle-aged women. J. Am. Coll. Nutr. 2003, 22, 533–538. [Google Scholar] [CrossRef]
- Humphries, S.; Kushner, H.; Falkner, B. Low dietary magnesium is associated with insulin resistance in a sample of young, nondiabetic Black Americans. Am. J. Hypertens. 1999, 12 Pt 1, 747–756. [Google Scholar] [CrossRef] [PubMed]
- Houtkooper, R.H.; Argmann, C.; Houten, S.M.; Cantó, C.; Jeninga, E.H.; Andreux, P.A.; Thomas, C.; Doenlen, R.; Schoonjans, K.; Auwerx, J. The metabolic footprint of aging in mice. Sci. Rep. 2011, 1, 134. [Google Scholar] [CrossRef] [PubMed]
- Matsunobu, S.; Terashima, Y.; Senshu, T.; Sano, H.; Itoh, H. Insulin secretion and glucose uptake in hypomagnesemic sheep fed a low magnesium, high potassium diet. J. Nutr. Biochem. 1990, 1, 167–171. [Google Scholar] [CrossRef] [PubMed]
- Balon, T.W.; Gu, J.L.; Tokuyama, Y.; Jasman, A.P.; Nadler, J.L. Magnesium supplementation reduces development of diabetes in a rat model of spontaneous NIDDM. Am. J. Physiol. Metab. 1995, 269, E745–E752. [Google Scholar] [CrossRef] [PubMed]
- Thomas, A.P.; Diggle, T.; Denton, R.M. Sensitivity of pyruvate dehydrogenase phosphate phosphatase to magnesium ions. Similar effects of spermine and insulin. Biochem. J. 1986, 238, 83–91. [Google Scholar] [CrossRef] [PubMed]
- Tretter, L.; Adam-Vizi, V. Alpha-ketoglutarate dehydrogenase: A target and generator of oxidative stress. Philos. Trans. R. Soc. B Biol. Sci. 2005, 360, 2335–2345. [Google Scholar] [CrossRef]
- McLain, A.L.; Szweda, P.A.; Szweda, L.I. alpha-Ketoglutarate dehydrogenase: A mitochondrial redox sensor. Free Radic. Res. 2011, 45, 29–36. [Google Scholar] [CrossRef]
- Mooren, F.C. Magnesium and disturbances in carbohydrate metabolism. Diabetes Obes. Metab. 2015, 17, 813–823. [Google Scholar] [CrossRef]
- Rodríguez-Morán, M.; Guerrero-Romero, F. Insulin secretion is decreased in non-diabetic individuals with hypomagnesaemia. Diabetes/Metab. Res. Rev. 2011, 27, 590–596. [Google Scholar] [CrossRef] [PubMed]
- 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 Element Res. 2019, 190, 396–404. [Google Scholar] [CrossRef]
- Le Couteur, D.G.; Simpson, S.J. 90th Anniversary Commentary: Caloric Restriction Effects on Aging. J. Nutr. 2018, 148, 1656–1659. [Google Scholar] [CrossRef] [PubMed]
- Abraham, K.J.; Chan, J.N.; Salvi, J.S.; Ho, B.; Hall, A.; Vidya, E.; Guo, R.; Killackey, S.A.; Liu, N.; Lee, J.E.; et al. Intersection of calorie restriction and magnesium in the suppression of genome-destabilizing RNA–DNA hybrids. Nucleic Acids Res. 2016, 44, 8870–8884. [Google Scholar] [CrossRef]
- Collado, M.; Blasco, M.A.; Serrano, M. Cellular senescence in cancer and aging. Cell 2007, 130, 223–233. [Google Scholar] [CrossRef]
- Tchkonia, T.; Zhu, Y.; van Deursen, J.; Campisi, J.; Kirkland, J.L. Cellular senescence and the senescent secretory phenotype: Therapeutic opportunities. J. Clin. Investig. 2013, 123, 966–972. [Google Scholar] [CrossRef]
- Song, S.; Lam, E.W.-F.; Tchkonia, T.; Kirkland, J.L.; Sun, Y. Senescent Cells: Emerging Targets for Human Aging and Age-Related Diseases. Trends Biochem. Sci. 2020, 45, 578–592. [Google Scholar] [CrossRef] [PubMed]
- Baker, D.J.; Childs, B.G.; Durik, M.; Wijers, M.E.; Sieben, C.J.; Zhong, J.; Saltness, R.A.; Jeganathan, K.B.; Verzosa, G.C.; Pezeshki, A.; et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature 2016, 530, 184–189. [Google Scholar] [CrossRef]
- Xu, M.; Pirtskhalava, T.; Farr, J.N.; Weigand, B.M.; Palmer, A.K.; Weivoda, M.M.; Inman, C.L.; Ogrodnik, M.B.; Hachfeld, C.M.; Fraser, D.G.; et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 2018, 24, 1246–1256. [Google Scholar] [CrossRef]
- Blackburn, E.H. Telomere states and cell fates. Nature 2000, 408, 53–56. [Google Scholar] [CrossRef]
- Sgambato, A.; Wolf, F.I.; 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, 245–254. [Google Scholar] [CrossRef]
- Killilea, D.W.; Ames, B.N. Magnesium deficiency accelerates cellular senescence in cultured human fibroblasts. Proc. Natl. Acad. Sci. USA 2008, 105, 5768–5773. [Google Scholar] [CrossRef] [PubMed]
- Ferrè, S.; Mazur, A.; Maier, J.A.M. Low-magnesium induces senescent features in cultured human endothelial cells. Magnes. Res. 2007, 20, 66–71. [Google Scholar] [PubMed]
- Mihaylova, M.M.; Sabatini, D.M.; Yilmaz, H. Dietary and metabolic control of stem cell function in physiology and cancer. Cell Stem Cell 2014, 14, 292–305. [Google Scholar] [CrossRef] [PubMed]
- Gruver, A.; Hudson, L.; Sempowski, G. Immunosenescence of ageing. J. Pathol. 2007, 211, 144–156. [Google Scholar] [CrossRef] [PubMed]
- Fulop, T.; Larbi, A.; Dupuis, G.; Le Page, A.; Frost, E.H.; Cohen, A.A.; Witkowski, J.M.; Franceschi, C. Immunosenescence and Inflamm-Aging as Two Sides of the Same Coin: Friends or Foes? Front. Immunol. 2017, 8, 1960. [Google Scholar] [CrossRef] [PubMed]
- Elias, R.; Hartshorn, K.; Rahma, O.; Lin, N.; Snyder-Cappione, J.E. Aging, immune senescence, and immunotherapy: A comprehensive review. Semin. Oncol. 2018, 45, 187–200. [Google Scholar] [CrossRef]
- Tannou, T.; Koeberle, S.; Manckoundia, P.; Aubry, R. Multifactorial immunodeficiency in frail elderly patients: Contributing factors and management. Med. Mal. Infect. 2019, 49, 167–172. [Google Scholar] [CrossRef]
- Dominguez, L.J.; Veronese, N.; Guerrero-Romero, F.; Barbagallo, M. Magnesium in Infectious Diseases in Older People. Nutrients 2021, 13, 180. [Google Scholar] [CrossRef]
- Tam, M.; Gómez, S.; González-Gross, M.; Marcos, A. Possible roles of magnesium on the immune system. Eur. J. Clin. Nutr. 2003, 57, 1193–1197. [Google Scholar] [CrossRef]
- Galland, L. Magnesium and immune function: An overview. Magnesium 1988, 7, 290–299. [Google Scholar]
- Sugimoto, J.; Romani, A.M.; Valentin-Torres, A.M.; Luciano, A.A.; Kitchen, C.M.R.; Funderburg, N.; Mesiano, S.; Bernstein, H.B. Magnesium decreases inflammatory cytokine production: A novel innate immunomodulatory mechanism. J. Immunol. 2012, 188, 6338–6346. [Google Scholar] [CrossRef]
- Feske, S.; Skolnik, E.Y.; Prakriya, M. Ion channels and transporters in lymphocyte function and immunity. Nat. Rev. Immunol. 2012, 12, 532–547. [Google Scholar] [CrossRef]
- Bussière, F.; Tridon, A.; Zimowska, W.; Mazur, A.; Rayssiguier, Y. Increase in complement component C3 is an early response to experimental magnesium deficiency in rats. Life Sci. 2003, 73, 499–507. [Google Scholar] [CrossRef]
- Kraeuter, S.L.; Schwartz, R. Blood and mast cell histamine levels in magnesium-deficient rats. J. Nutr. 1980, 110, 851–858. [Google Scholar] [CrossRef]
- Takemoto, S.; Yamamoto, A.; Tomonaga, S.; Funaba, M.; Matsui, T. Magnesium deficiency induces the emergence of mast cells in the liver of rats. J. Nutr. Sci. Vitaminol. 2013, 59, 560–563. [Google Scholar] [CrossRef]
- Chien, M.M.; Zahradka, K.E.; Newell, M.K.; Freed, J. HFas-induced B cell apoptosis requires an increase in free cytosolic magnesium as an early event. J. Biol. Chem. 1999, 274, 7059–7066. [Google Scholar] [CrossRef] [PubMed]
- Malpuech-Brugere, C.; Nowacki, W.; Daveau, M.; Gueux, E.; Linard, C.; Rock, E.; Lebreton, J.-P.; Mazur, A.; Rayssiguier, Y. Inflammatory response following acute mag-nesium deficiency in the rat. Biochim. Biophys. Acta. 2000, 1501, 91–98. [Google Scholar] [CrossRef]
- Petrault, I.; Zimowska, W.; Mathieu, J.; Bayle, D.; Rock, E.; Favier, A.; Rayssiguier, Y.; Mazur, A. Changes in gene expression in rat thymocytes identified by cDNA array support the occurrence of oxidative stress in early magnesium deficiency. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2002, 1586, 92–98. [Google Scholar] [CrossRef]
- Zimowska, W.; Girardeau, J.P.; Kuryszko, J.; Bayle, D.; Rayssiguier, Y.; Mazur, A. Morphological and immune response alterations in the intestinal mucosa of the mouse after short periods on a low-magnesium diet. Br. J. Nutr. 2002, 88, 515–522. [Google Scholar] [CrossRef] [PubMed]
- Li, F.-Y.; Chaigne-Delalande, B.; Kanellopoulou, C.; Davis, J.C.; Matthews, H.F.; Douek, D.C.; Cohen, J.I.; Uzel, G.; Su, H.C.; Lenardo, M.J. Second messenger role for Mg2+ revealed by human T-cell immunodeficiency. Nature 2011, 475, 471–476. [Google Scholar] [CrossRef]
- Li, F.-Y.; Chaigne-Delalande, B.; Su, H.; Uzel, G.; Matthews, H.; Lenardo, M.J. XMEN disease: A new primary immunodeficiency affecting Mg2+ regulation of immunity against Epstein-Barr virus. Blood 2014, 123, 2148–2152. [Google Scholar] [CrossRef]
- Chaigne-Delalande, B.; Li, F.-Y.; O’connor, G.M.; Lukacs, M.J.; Jiang, P.; Zheng, L.; Shatzer, A.; Biancalana, M.; Pittaluga, S.; Matthews, H.F.; et al. Mg2+ regulates cytotoxic functions of NK and CD8 T cells in chronic EBV infection through NKG2D. Science 2013, 341, 186–191. [Google Scholar] [CrossRef] [PubMed]
- Lima, F.D.S.; Fock, R.A. A Review of the Action of Magnesium on Several Processes Involved in the Modulation of Hematopoiesis. Int. J. Mol. Sci. 2020, 21, 7084. [Google Scholar] [CrossRef]
- Naveiras, O.; Nardi, V.; Wenzel, P.L.; Hauschka, P.V.; Fahey, F.; Daley, G.Q. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature 2009, 460, 259–263. [Google Scholar] [CrossRef] [PubMed]
- Sugimura, R.; Li, L. Shifting in balance between osteogenesis and adipogenesis substantially influences hematopoiesis. J. Mol. Cell Biol. 2010, 2, 61–62. [Google Scholar] [CrossRef]
- Rude, R.K.; Gruber, H.E. Magnesium deficiency and osteoporosis: Animal and human observations. J. Nutr. Biochem. 2004, 15, 710–716. [Google Scholar] [CrossRef] [PubMed]
- Castiglioni, S.; Cazzaniga, A.; Albisetti, W.; Maier, J.A.M. Magnesium and osteoporosis: Current state of knowledge and future research directions. Nutrients 2013, 5, 3022–3033. [Google Scholar] [CrossRef]
- Sargenti, A.; Castiglioni, S.; Olivi, E.; Bianchi, F.; Cazzaniga, A.; Farruggia, G.; Cappadone, C.; Merolle, L.; Malucelli, E.; Ventura, C.; et al. Magnesium Deprivation Potentiates Human Mesenchymal Stem Cell Transcriptional Remodeling. Int. J. Mol. Sci. 2018, 19, 1410. [Google Scholar] [CrossRef]
- Mittelbrunn, M.; Sánchez-Madrid, F. Intercellular communication: Diverse structures for exchange of genetic information. Nat. Rev. Mol. Cell Biol. 2012, 13, 328–335. [Google Scholar] [CrossRef]
- Franceschi, C.; Bonafe, M.; Valensin, S.; Olivieri, F.; De Luca, M.; Ottaviani, E.; De Benedictis, G. Inflamm-aging: An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef] [PubMed]
- Cohen, H.J.; Pieper, C.F.; Harris, T.; Rao, K.M.; Currie, M.S. The association of plasma IL-6 levels with functional disability in commu-nity-dwelling elderly. J. Gerontol. A Biol. Sci. Med. Sci. 1997, 52, M201–M208. [Google Scholar] [CrossRef] [PubMed]
- Franceschi, C.; Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. A Ser. Biol. Sci. Med. Sci. 2014, 69 (Suppl. S1), S4–S9. [Google Scholar] [CrossRef] [PubMed]
- Rosano, C.; Marsland, A.L.; Gianaros, P.J. Maintaining brain health by monitoring inflammatory processes: A mechanism to promote successful aging. Aging Dis. 2012, 3, 16–33. [Google Scholar] [PubMed]
- Harris, T.B.; Ferrucci, L.; Tracy, R.P.; Corti, M.C.; Wacholder, S.; Ettinger, W.H., Jr.; Heimovitz, H.; Cohen, H.J.; Wallace, R. Associations of elevated Interleukin-6 and C-Reactive protein levels with mortality in the elderly. Am. J. Med. 1999, 106, 506–512. [Google Scholar] [CrossRef] [PubMed]
- Michaud, M.; Balardy, L.; Moulis, G.; Gaudin, C.; Peyrot, C.; Vellas, B.; Vellas, B.; Cesari, M.; Nourhashemi, F. Proinflammatory cytokines, aging, and age-related diseases. J. Am. Med. Dir. Assoc. 2013, 14, 877–882. [Google Scholar] [CrossRef] [PubMed]
- Szarcvel Szic, K.; Declerck, K.; Vidakovic, M.; Vanden Berghe, W. From inflammaging to healthy aging by dietary lifestyle choices: Is epigenetics the key to personalized nutrition? Clin. Epigenetics 2015, 7, 33. [Google Scholar] [CrossRef] [PubMed]
- Xia, S.; Zhang, X.; Zheng, S.; Khanabdali, R.; Kalionis, B.; Wu, J.; Wan, W.; Tai, X. An Update on Inflamm-Aging: Mechanisms, Prevention, and Treatment. J. Immunol. Res. 2016, 2016, 8426874. [Google Scholar] [CrossRef]
- Fransen, F.; Van Beek, A.A.; Borghuis, T.; El Aidy, S.; Hugenholtz, F.; van der Gaast–de Jongh, C.; Savelkoul, H.F.J.; De Jonge, M.I.; Boekschoten, M.V.; Smidt, H.; et al. Aged Gut Microbiota Contributes to Systemical Inflammaging after Transfer to Germ-Free Mice. Front. Immunol. 2017, 8, 1385. [Google Scholar] [CrossRef]
- Leonardi, G.C.; Accardi, G.; Monastero, R.; Nicoletti, F.; Libra, M. Ageing: From inflammation to cancer. Immun. Ageing 2018, 15, 1. [Google Scholar] [CrossRef]
- Shapiro, H.A.; Thaiss, C.; Levy, M.; Elinav, E. The cross talk between microbiota and the immune system: Metabolites take center stage. Curr. Opin. Immunol. 2014, 30, 54–62. [Google Scholar] [CrossRef]
- Yogi, A.; Callera, G.E.; Antunes, T.T.; Tostes, R.C.; Touyz, R.M. Transient receptor potential melastatin 7 (TRPM7) cation channels, magnesium and the vascular system in hypertension. Circ. J. 2011, 75, 237–245. [Google Scholar] [CrossRef]
- Schlingmann, K.P.; Waldegger, S.; Konrad, M.; Chubanov, V.; Gudermann, T. TRPM6 and TRPM7—Gatekeepers of human magnesium metabolism. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2007, 1772, 813–821. [Google Scholar] [CrossRef] [PubMed]
- Trzeciakiewicz, A.; Opolski, A.; Mazur, A. TRPM7: A protein responsible for magnesium homeostasis in a cell. Adv. Hyg. Exp. Med. 2005, 59, 496–502. [Google Scholar]
- Sahni, J.; Tamura, R.; Sweet, I.R.; Scharenberg, A.M. TRPM7 regulates quiescent/proliferative metabolic transitions in lymphocytes. Cell Cycle 2010, 9, 3565–3574. [Google Scholar] [CrossRef] [PubMed]
- Wong, S.Q.; Kumar, A.V.; Mills, J.; Lapierre, L.R. Autophagy in aging and longevity. Hum. Genet. 2020, 139, 277–290. [Google Scholar] [CrossRef] [PubMed]
- Aman, Y.; Schmauck-Medina, T.; Hansen, M.; Morimoto, R.I.; Simon, A.K.; Bjedov, I.; Palikaras, K.; Simonsen, A.; Johansen, T.; Tavernarakis, N.; et al. Autophagy in healthy aging and disease. Nat. Aging 2021, 1, 634–650. [Google Scholar] [CrossRef] [PubMed]
- Fernández, Á.F.; Sebti, S.; Wei, Y.; Zou, Z.; Shi, M.; McMillan, K.L.; He, C.; Ting, T.; Liu, Y.; Chiang, W.-C.; et al. Disruption of the beclin 1–BCL2 autophagy regulatory complex promotes longevity in mice. Nature 2018, 558, 136–140. [Google Scholar] [CrossRef]
- Alsaleh, G.; Panse, I.; Swadling, L.; Zhang, H.; Richter, F.C.; Meyer, A.; Lord, J.; Barnes, E.; Klenerman, P.; Green, C.; et al. Autophagy in T cells from aged donors is maintained by spermidine and correlates with function and vaccine responses. Elife 2020, 9, e57950. [Google Scholar] [CrossRef]
- Kaushik, S.; Cuervo, A.M. The coming of age of chaperone-mediated autophagy. Nat. Rev. Mol. Cell Biol. 2018, 19, 365–381. [Google Scholar] [CrossRef]
- Wolf, F.I.; Trapani, V. Cell (patho)physiology of magnesium. Clin. Sci. 2008, 114, 27–35. [Google Scholar] [CrossRef] [PubMed]
- 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, 3563–3572. [Google Scholar] [CrossRef] [PubMed]
- Martin, H.; Richert, L.; Berthelot, A. Magnesium deficiency induces apoptosis in primary cultures of rat hepatocytes. J. Nutr. 2003, 133, 2505–2511. [Google Scholar] [CrossRef] [PubMed]
- Feng, H.; Guo, L.; Gao, H.; Li, X.-A. Deficiency of calcium and magnesium induces apoptosis via scavenger receptor BI. Life Sci. 2011, 88, 606–612. [Google Scholar] [CrossRef]
- Krzywoszyńska, K.; Witkowska, D.; Świątek-Kozłowska, J.; Szebesczyk, A.; Kozłowski, H. General Aspects of Metal Ions as Signaling Agents in Health and Disease. Biomolecules 2020, 10, 1417. [Google Scholar] [CrossRef]
- Giannakis, C.; Forbes, I.J.; Zalewski, P.D. Ca2+ Mg2+-dependent nuclease: Tissue distribution, relationship to inter-nucleosomal DNA fragmentation and inhibition by Zn2+. Biochem. Biophys. Res. Commun. 1991, 181, 915–920. [Google Scholar] [CrossRef]
- Wilmanski, T.; Diener, C.; Rappaport, N.; Patwardhan, S.; Wiedrick, J.; Lapidus, J.; Earls, J.C.; Zimmer, A.; Glusman, G.; Robinson, M.; et al. Gut microbiome pattern reflects healthy ageing and predicts survival in humans. Nat. Metab. 2021, 3, 274–286. [Google Scholar] [CrossRef]
- Agirman, G.; Yu, K.B.; Hsiao, E.Y. Signaling inflammation across the gut-brain axis. Science 2021, 374, 1087–1092. [Google Scholar] [CrossRef]
- Laragione, T.; Harris, C.; Azizgolshani, N.; Beeton, C.; Bongers, G.; Gulko, P.S. Magnesium increases numbers of Foxp3+ Treg cells and reduces arthritis severity and joint damage in an IL-10-dependent manner mediated by the intestinal microbiome. EBioMedicine 2023, 92, 104603. [Google Scholar] [CrossRef] [PubMed]
- Gommers, L.M.M.; Hoenderop, J.G.J.; de Baaij, J.H.F. Mechanisms of proton pump inhibitor-induced hypomagnesemia. Acta Physiol. 2022, 235, e13846. [Google Scholar] [CrossRef] [PubMed]
- Xia, Y.; Shi, H.; Qian, C.; Han, H.; Lu, K.; Tao, R.; Gu, R.; Zhao, Y.; Wei, Z.; Lu, Y.; et al. Modulation of Gut Microbiota by Magnesium Isoglycyrrhizinate Mediates En-hancement of Intestinal Barrier Function and Amelioration of Methotrexate-Induced Liver Injury. Front Immunol. 2022, 13, 874878. [Google Scholar] [CrossRef]
- Del Chierico, F.; Trapani, V.; Petito, V.; Reddel, S.; Pietropaolo, G.; Graziani, C.; Masi, L.; Gasbarrini, A.; Putignani, L.; Scaldaferri, F.; et al. Dietary Magnesium Alleviates Experimental Murine Colitis through Modulation of Gut Microbiota. Nutrients 2021, 13, 4188. [Google Scholar] [CrossRef]
- Cao, S.; Huang, K.; Wen, X.; Gao, J.; Cui, B.; Yao, K.; Zhan, X.; Hu, S.; Wu, Q.; Xiao, H.; et al. Dietary supplementation with potassium-magnesium sulfate modulates the antioxidant capacity, immunity, and gut microbiota in weaned piglets. Front. Microbiol. 2022, 13, 961989. [Google Scholar] [CrossRef]
- Jørgensen, B.P.; Winther, G.; Kihl, P.; Nielsen, D.S.; Wegener, G.; Hansen, A.K.; Sørensen, D.B. Dietary magnesium deficiency affects gut microbiota and anxiety-like behaviour in C57BL/6N mice. Acta Neuropsychiatr. 2015, 27, 307–311. [Google Scholar] [CrossRef] [PubMed]
- Winther, G.; Jørgensen, B.M.P.; Elfving, B.; Nielsen, D.S.; Kihl, P.; Lund, S.; Sørensen, D.B.; Wegener, G. Dietary magnesium deficiency alters gut microbiota and leads to depressive-like behaviour. Acta Neuropsychiatr. 2015, 27, 168–176. [Google Scholar] [CrossRef] [PubMed]
- Pachikian, B.D.; Neyrinck, A.M.; Deldicque, L.; De Backer, F.C.; Catry, E.; Dewulf, E.M.; Sohet, F.M.; Bindels, L.B.; Everard, A.; Francaux, M.; et al. Changes in intestinal bifidobacteria levels are associated with the inflammatory response in magnesium-deficient mice. J. Nutr. 2010, 140, 509–514. [Google Scholar] [CrossRef] [PubMed]
- Ferrucci, L.; Fabbri, E. Inflammageing: Chronic inflammation in ageing, cardiovascular disease, and frailty. Nat. Rev. Cardiol. 2018, 15, 505–522. [Google Scholar] [CrossRef] [PubMed]
- Kolisek, M.; Touyz, R.M.; Romani, A.; Barbagallo, M. Magnesium and Other Biometals in Oxidative Medicine and Redox Biology. Oxidative Med. Cell. Longev. 2017, 2017, 7428796. [Google Scholar] [CrossRef] [PubMed]
- Maier, J.A.; Castiglioni, S.; Locatelli, L.; Zocchi, M.; Mazur, A. Magnesium and inflammation: Advances and perspectives. Semin. Cell Dev. Biol. 2021, 115, 37–44. [Google Scholar] [CrossRef]
- Mazur, A.; Maier, J.A.; Rock, E.; Gueux, E.; Nowacki, W.; Rayssiguier, Y. Magnesium and the inflammatory response: Potential physiopathological implications. Arch. Biochem. Biophys. 2007, 458, 48–56. [Google Scholar] [CrossRef]
- Kramer, J.H.; Mak, I.T.; Phillips, T.M.; Weglicki, W.B. Dietary magnesium intake influences circulating pro-inflammatory neuropeptide levels and loss of myocardial tolerance to postischemic stress. Exp. Biol. Med. 2003, 228, 665–673. [Google Scholar] [CrossRef] [PubMed]
- Maier, J.A.; Malpuech-Brugere, C.; Zimowska, W.; Rayssiguier, Y.; Mazur, A. Low magnesium promotes endothelial cell dysfunction: Implications for atherosclerosis, inflammation and thrombosis. Biochim. Biophys Acta. 2004, 1689, 13–21. [Google Scholar] [CrossRef] [PubMed]
- Locatelli, L.; Fedele, G.; Castiglioni, S.; Maier, J.A. 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, 1050. [Google Scholar] [CrossRef] [PubMed]
- Stankovic, M.S.; Janjetovic, K.; Velimirovic, M.; Milenkovic, M.; Stojkovic, T.; Puskas, N.; Zaletel, I.; De Luka, S.R.; Jankovic, S.; Stefanovic, S.; 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, 31–37. [Google Scholar] [CrossRef] [PubMed]
- Su, N.-Y.; Peng, T.-C.; Tsai, P.-S.; Huang, C.-J. Phosphoinositide 3-kinase/Akt pathway is involved in mediating the anti-inflammation effects of magnesium sulfate. J. Surg. Res. 2013, 185, 726–732. [Google Scholar] [CrossRef]
- Lin, C.Y.; Tsai, P.S.; Hung, Y.C.; Huang, C.J. L-type calcium channels are involved in mediating the anti-inflammatory effects of magnesium sulphate. Br. J. Anaesth. 2010, 104, 44–51. [Google Scholar] [CrossRef] [PubMed]
- King, D.E.; Mainous, A.G., 3rd; Geesey, M.E.; Woolson, R.F. Dietary magnesium and C-reactive protein levels. J. Am. Coll. Nutr. 2005, 24, 166–171. [Google Scholar] [CrossRef]
- Song, Y.; Li, T.Y.; van Dam, R.M.; Manson, J.E.; Hu, F.B. Magnesium intake and plasma concentrations of markers of systemic in-flammation and endothelial dysfunction in women. Am. J. Clin. Nutr. 2007, 85, 1068–1074. [Google Scholar] [CrossRef]
- Guerrero-Romero, F.; Bermudez-Peña, C.; Rodríguez-Morán, M. Severe hypomagnesemia and low-grade inflammation in metabolic syndrome. Magnes. Res. 2011, 24, 45–53. [Google Scholar] [CrossRef]
- Song, Y.; Ridker, P.M.; Manson, J.E.; Cook, N.R.; Buring, J.E.; Liu, S. Magnesium intake, C-reactive protein, and the prevalence of met-abolic syndrome in middle-aged and older U.S. women. Diabetes Care 2005, 28, 1438–1444. [Google Scholar] [CrossRef]
- Mazidi, M.; Kengne, A.P.; Mikhailidis, D.P.; Cicero, A.F.; Banach, M. Effects of selected dietary constituents on high-sensitivity C-reactive protein levels in U.S. adults. Ann. Med. 2018, 50, 1–6. [Google Scholar] [CrossRef]
- Konstari, S.; Sares-Jaske, L.; Heliovaara, M.; Rissanen, H.; Knekt, P.; Arokoski, J.; Sundvall, J.; Karppinen, J. Dietary magnesium intake, serum high sensitivity C-reactive protein and the risk of incident knee osteoarthritis leading to hospitalization-A cohort study of 4,953 Finns. PLoS ONE 2019, 14, e0214064. [Google Scholar] [CrossRef] [PubMed]
- Mazidi, M.; Rezaie, P.; Banach, M. Effect of magnesium supplements on serum C-reactive protein: A systematic review and me-ta-analysis. Arch. Med. Sci. 2018, 14, 707–716. [Google Scholar] [CrossRef] [PubMed]
- Veronese, N.; Pizzol, D.; Smith, L.; Dominguez, L.J.; Barbagallo, M. Effect of Magnesium Supplementation on Inflammatory Param-eters: A Meta-Analysis of Randomized Controlled Trials. Nutrients 2022, 14, 679. [Google Scholar] [CrossRef] [PubMed]
Genetic disorders of renal magnesium wasting
|
Medications
|
Neuromuscular and central nervous system
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Dominguez, L.J.; Veronese, N.; Barbagallo, M. Magnesium and the Hallmarks of Aging. Nutrients 2024, 16, 496. https://doi.org/10.3390/nu16040496
Dominguez LJ, Veronese N, Barbagallo M. Magnesium and the Hallmarks of Aging. Nutrients. 2024; 16(4):496. https://doi.org/10.3390/nu16040496
Chicago/Turabian StyleDominguez, Ligia J., Nicola Veronese, and Mario Barbagallo. 2024. "Magnesium and the Hallmarks of Aging" Nutrients 16, no. 4: 496. https://doi.org/10.3390/nu16040496
APA StyleDominguez, L. J., Veronese, N., & Barbagallo, M. (2024). Magnesium and the Hallmarks of Aging. Nutrients, 16(4), 496. https://doi.org/10.3390/nu16040496