The Role of Taurine in Mitochondria Health: More Than Just an Antioxidant
Abstract
:1. Introduction
2. Mitochondria Biology
3. Mitochondria in Pathologies
4. Taurine Biology
5. Taurine as a Therapeutic Agent in Mitochondrial Dysfunction
5.1. Taurine Forms a Complex with Mitochondrial tRNA
5.2. Taurine Reduces Superoxide Generation in the Mitochondria
5.3. Taurine Regulates Intracellular Calcium Homeostasis
5.4. Taurine Inhibits Mitochondria-Mediated Apoptosis
6. Clinical Application of Taurine in Mitochondria-Targeted Pathologies
6.1. Cardiovascular Diseases
6.2. Metabolic Syndrome
6.3. Mitochondrial Diseases
6.4. Neurological Disorders
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Harman, D. Aging: A theory based on free radical and radiation chemistry. J. Gerontol. 1956, 11, 298–300. [Google Scholar] [CrossRef] [Green Version]
- Harman, D. The biologic clock: The mitochondria? J. Am. Geriatr. Soc. 1972, 20, 145–147. [Google Scholar] [CrossRef]
- Scheubel, R.J.; Tostlebe, M.; Simm, A.; Rohrbach, S.; Prondzinsky, R.; Gellerich, F.N.; Silber, R.E.; Holtz, J. Dysfunction of mitochondrial respiratory chain complex I in human failing myocardium is not due to disturbed mitochondrial gene expression. J. Am. Coll. Cardiol. 2002, 40, 2174–2181. [Google Scholar] [CrossRef] [Green Version]
- Marin-Garcia, J.; Goldenthal, M.J.; Moe, G.W. Mitochondrial pathology in cardiac failure. Cardiovasc. Res. 2001, 49, 17–26. [Google Scholar] [CrossRef] [Green Version]
- Shapira, Y.; Cederbaum, S.D.; Cancilla, P.A.; Nielsen, D.; Lippe, B.M. Familial poliodystrophy, mitochondrial myopathy, and lactate acidemia. Neurology 1975, 25, 614–621. [Google Scholar] [CrossRef]
- Hayashi, G.; Cortopassi, G. Oxidative stress in inherited mitochondrial diseases. Free Radic. Biol. Med. 2015, 88, 10–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bournat, J.C.; Brown, C.W. Mitochondrial dysfunction in obesity. Curr. Opin. Endocrinol. Diabetes Obes. 2010, 17, 446–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prasun, P. Role of mitochondria in pathogenesis of type 2 diabetes mellitus. J. Diabetes Metab. Disord. 2020, 19, 2017–2022. [Google Scholar] [CrossRef] [PubMed]
- Zong, W.X.; Rabinowitz, J.D.; White, E. Mitochondria and Cancer. Mol. Cell 2016, 61, 667–676. [Google Scholar] [CrossRef] [Green Version]
- Modica-Napolitano, J.S.; Singh, K.K. Mitochondrial dysfunction in cancer. Mitochondrion 2004, 4, 755–762. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Zhao, F.; Ma, X.; Perry, G.; Zhu, X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: Recent advances. Mol. Neurodegener. 2020, 15, 30. [Google Scholar] [CrossRef]
- Pallardo, F.V.; Lloret, A.; Lebel, M.; D’Ischia, M.; Cogger, V.C.; Le Couteur, D.G.; Gadaleta, M.N.; Castello, G.; Pagano, G. Mitochondrial dysfunction in some oxidative stress-related genetic diseases: Ataxia-Telangiectasia, Down Syndrome, Fanconi Anaemia and Werner Syndrome. Biogerontology 2010, 11, 401–419. [Google Scholar] [CrossRef]
- Griffiths, K.K.; Levy, R.J. Evidence of Mitochondrial Dysfunction in Autism: Biochemical Links, Genetic-Based Associations, and Non-Energy-Related Mechanisms. Oxid. Med. Cell. Longev. 2017, 2017, 4314025. [Google Scholar] [CrossRef]
- Haas, R.H. Autism and mitochondrial disease. Dev. Disabil. Res. Rev. 2010, 16, 144–153. [Google Scholar] [CrossRef]
- Negida, A.; Menshawy, A.; El Ashal, G.; Elfouly, Y.; Hani, Y.; Hegazy, Y.; El Ghonimy, S.; Fouda, S.; Rashad, Y. Coenzyme Q10 for Patients with Parkinson’s Disease: A Systematic Review and Meta-Analysis. CNS Neurol. Disord. Drug Targets 2016, 15, 45–53. [Google Scholar] [CrossRef]
- Rossman, M.J.; Santos-Parker, J.R.; Steward, C.A.C.; Bispham, N.Z.; Cuevas, L.M.; Rosenberg, H.L.; Woodward, K.A.; Chonchol, M.; Gioscia-Ryan, R.A.; Murphy, M.P.; et al. Chronic Supplementation With a Mitochondrial Antioxidant (MitoQ) Improves Vascular Function in Healthy Older Adults. Hypertension 2018, 71, 1056–1063. [Google Scholar] [CrossRef] [PubMed]
- Snow, B.J.; Rolfe, F.L.; Lockhart, M.M.; Frampton, C.M.; O’Sullivan, J.D.; Fung, V.; Smith, R.A.; Murphy, M.P.; Taylor, K.M.; Protect Study, G. A double-blind, placebo-controlled study to assess the mitochondria-targeted antioxidant MitoQ as a disease-modifying therapy in Parkinson’s disease. Mov. Disord. 2010, 25, 1670–1674. [Google Scholar] [CrossRef] [PubMed]
- Sozen, E.; Demirel, T.; Ozer, N.K. Vitamin E: Regulatory role in the cardiovascular system. IUBMB Life 2019, 71, 507–515. [Google Scholar] [CrossRef] [PubMed]
- Le Bars, P.L.; Katz, M.M.; Berman, N.; Itil, T.M.; Freedman, A.M.; Schatzberg, A.F. A placebo-controlled, double-blind, randomized trial of an extract of Ginkgo biloba for dementia. North American EGb Study Group. JAMA 1997, 278, 1327–1332. [Google Scholar] [CrossRef] [PubMed]
- Yamaguchi, T.; Sano, K.; Takakura, K.; Saito, I.; Shinohara, Y.; Asano, T.; Yasuhara, H. Ebselen in acute ischemic stroke: A placebo-controlled, double-blind clinical trial. Ebselen Study Group. Stroke 1998, 29, 12–17. [Google Scholar] [CrossRef] [PubMed]
- Tarnopolsky, M.A.; Roy, B.D.; MacDonald, J.R. A randomized, controlled trial of creatine monohydrate in patients with mitochondrial cytopathies. Muscle Nerve 1997, 20, 1502–1509. [Google Scholar] [CrossRef]
- Hager, K.; Kenklies, M.; McAfoose, J.; Engel, J.; Munch, G. Alpha-lipoic acid as a new treatment option for Alzheimer’s disease—A 48 months follow-up analysis. J. Neural Transm. Suppl. 2007, 72, 189–193. [Google Scholar] [CrossRef]
- Chahbouni, M.; Escames, G.; Venegas, C.; Sevilla, B.; Garcia, J.A.; Lopez, L.C.; Munoz-Hoyos, A.; Molina-Carballo, A.; Acuna-Castroviejo, D. Melatonin treatment normalizes plasma pro-inflammatory cytokines and nitrosative/oxidative stress in patients suffering from Duchenne muscular dystrophy. J. Pineal Res. 2010, 48, 282–289. [Google Scholar] [CrossRef]
- Weishaupt, J.H.; Bartels, C.; Polking, E.; Dietrich, J.; Rohde, G.; Poeggeler, B.; Mertens, N.; Sperling, S.; Bohn, M.; Huther, G.; et al. Reduced oxidative damage in ALS by high-dose enteral melatonin treatment. J. Pineal Res. 2006, 41, 313–323. [Google Scholar] [CrossRef]
- Koga, Y.; Akita, Y.; Nishioka, J.; Yatsuga, S.; Povalko, N.; Tanabe, Y.; Fujimoto, S.; Matsuishi, T. L-arginine improves the symptoms of strokelike episodes in MELAS. Neurology 2005, 64, 710–712. [Google Scholar] [CrossRef]
- Koga, Y.; Ishibashi, M.; Ueki, I.; Yatsuga, S.; Fukiyama, R.; Akita, Y.; Matsuishi, T. Effects of L-arginine on the acute phase of strokes in three patients with MELAS. Neurology 2002, 58, 827–828. [Google Scholar] [CrossRef] [Green Version]
- Ohsawa, Y.; Hagiwara, H.; Nishimatsu, S.I.; Hirakawa, A.; Kamimura, N.; Ohtsubo, H.; Fukai, Y.; Murakami, T.; Koga, Y.; Goto, Y.I.; et al. Taurine supplementation for prevention of stroke-like episodes in MELAS: A multicentre, open-label, 52-week phase III trial. J. Neurol. Neurosurg. Psychiatry 2019, 90, 529–536. [Google Scholar] [CrossRef] [PubMed]
- Rikimaru, M.; Ohsawa, Y.; Wolf, A.M.; Nishimaki, K.; Ichimiya, H.; Kamimura, N.; Nishimatsu, S.; Ohta, S.; Sunada, Y. Taurine ameliorates impaired the mitochondrial function and prevents stroke-like episodes in patients with MELAS. Intern. Med. 2012, 51, 3351–3357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Azuma, J.; Sawamura, A.; Awata, N.; Ohta, H.; Hamaguchi, T.; Harada, H.; Takihara, K.; Hasegawa, H.; Yamagami, T.; Ishiyama, T.; et al. Therapeutic effect of taurine in congestive heart failure: A double-blind crossover trial. Clin. Cardiol. 1985, 8, 276–282. [Google Scholar] [CrossRef] [PubMed]
- Azuma, J.; Hasegawa, H.; Sawamura, A.; Awata, N.; Ogura, K.; Harada, H.; Yamamura, Y.; Kishimoto, S. Therapy of congestive heart failure with orally administered taurine. Clin. Ther. 1983, 5, 398–408. [Google Scholar]
- Beyranvand, M.R.; Khalafi, M.K.; Roshan, V.D.; Choobineh, S.; Parsa, S.A.; Piranfar, M.A. Effect of taurine supplementation on exercise capacity of patients with heart failure. J. Cardiol. 2011, 57, 333–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jacobsen, J.G.; Smith, L.H. Biochemistry and physiology of taurine and taurine derivatives. Physiol. Rev. 1968, 48, 424–511. [Google Scholar] [CrossRef]
- Detmer, S.A.; Chan, D.C. Functions and dysfunctions of mitochondrial dynamics. Nat. Rev. Mol. Cell Biol. 2007, 8, 870–879. [Google Scholar] [CrossRef]
- Murphy, E.; Ardehali, H.; Balaban, R.S.; DiLisa, F.; Dorn, G.W., 2nd; Kitsis, R.N.; Otsu, K.; Ping, P.; Rizzuto, R.; Sack, M.N.; et al. Mitochondrial Function, Biology, and Role in Disease: A Scientific Statement From the American Heart Association. Circ. Res. 2016, 118, 1960–1991. [Google Scholar] [CrossRef] [PubMed]
- Herst, P.M.; Rowe, M.R.; Carson, G.M.; Berridge, M.V. Functional Mitochondria in Health and Disease. Front. Endocrinol. 2017, 8, 296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Romero-Garcia, S.; Prado-Garcia, H. Mitochondrial calcium: Transport and modulation of cellular processes in homeostasis and cancer (Review). Int. J. Oncol. 2019, 54, 1155–1167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tait, S.W.; Green, D.R. Mitochondria and cell signalling. J. Cell Sci. 2012, 125, 807–815. [Google Scholar] [CrossRef] [Green Version]
- Kuhlbrandt, W. Structure and function of mitochondrial membrane protein complexes. BMC Biol. 2015, 13, 89. [Google Scholar] [CrossRef] [Green Version]
- Alexeyev, M.F.; Ledoux, S.P.; Wilson, G.L. Mitochondrial DNA and aging. Clin. Sci. 2004, 107, 355–364. [Google Scholar] [CrossRef] [Green Version]
- Xing, G.; Chen, Z.; Cao, X. Mitochondrial rRNA and tRNA and hearing function. Cell Res. 2007, 17, 227–239. [Google Scholar] [CrossRef] [PubMed]
- Zhao, R.Z.; Jiang, S.; Zhang, L.; Yu, Z.B. Mitochondrial electron transport chain, ROS generation and uncoupling (Review). Int. J. Mol. Med. 2019, 44, 3–15. [Google Scholar] [CrossRef] [Green Version]
- Turrens, J.F. Mitochondrial formation of reactive oxygen species. J. Physiol. 2003, 552, 335–344. [Google Scholar] [CrossRef] [PubMed]
- Chen, Q.; Vazquez, E.J.; Moghaddas, S.; Hoppel, C.L.; Lesnefsky, E.J. Production of reactive oxygen species by mitochondria: Central role of complex III. J. Biol. Chem. 2003, 278, 36027–36031. [Google Scholar] [CrossRef] [Green Version]
- Hirst, J.; King, M.S.; Pryde, K.R. The production of reactive oxygen species by complex I. Biochem. Soc. Trans. 2008, 36, 976–980. [Google Scholar] [CrossRef]
- Cho, Y.M.; Kwon, S.; Pak, Y.K.; Seol, H.W.; Choi, Y.M.; Park, D.J.; Park, K.S.; Lee, H.K. Dynamic changes in mitochondrial biogenesis and antioxidant enzymes during the spontaneous differentiation of human embryonic stem cells. Biochem. Biophys. Res. Commun. 2006, 348, 1472–1478. [Google Scholar] [CrossRef]
- Tormos, K.V.; Anso, E.; Hamanaka, R.B.; Eisenbart, J.; Joseph, J.; Kalyanaraman, B.; Chandel, N.S. Mitochondrial complex III ROS regulate adipocyte differentiation. Cell Metab. 2011, 14, 537–544. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; McMillan-Ward, E.; Kong, J.; Israels, S.J.; Gibson, S.B. Mitochondrial electron-transport-chain inhibitors of complexes I and II induce autophagic cell death mediated by reactive oxygen species. J. Cell Sci. 2007, 120, 4155–4166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scherz-Shouval, R.; Shvets, E.; Fass, E.; Shorer, H.; Gil, L.; Elazar, Z. Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J. 2007, 26, 1749–1760. [Google Scholar] [CrossRef]
- Nemoto, S.; Takeda, K.; Yu, Z.X.; Ferrans, V.J.; Finkel, T. Role for mitochondrial oxidants as regulators of cellular metabolism. Mol. Cell. Biol. 2000, 20, 7311–7318. [Google Scholar] [CrossRef] [Green Version]
- Liemburg-Apers, D.C.; Willems, P.H.; Koopman, W.J.; Grefte, S. Interactions between mitochondrial reactive oxygen species and cellular glucose metabolism. Arch. Toxicol. 2015, 89, 1209–1226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- West, A.P.; Shadel, G.S.; Ghosh, S. Mitochondria in innate immune responses. Nat. Rev. Immunol. 2011, 11, 389–402. [Google Scholar] [CrossRef] [Green Version]
- Andreyev, A.Y.; Kushnareva, Y.E.; Starkova, N.N.; Starkov, A.A. Metabolic ROS Signaling: To Immunity and Beyond. Biochemistry 2020, 85, 1650–1667. [Google Scholar] [CrossRef]
- Cui, H.; Kong, Y.; Zhang, H. Oxidative stress, mitochondrial dysfunction, and aging. J. Signal Transduct. 2012, 2012, 646354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kowalska, M.; Piekut, T.; Prendecki, M.; Sodel, A.; Kozubski, W.; Dorszewska, J. Mitochondrial and Nuclear DNA Oxidative Damage in Physiological and Pathological Aging. DNA Cell Biol. 2020, 39, 1410–1420. [Google Scholar] [CrossRef]
- Cai, Z.; Yan, L.J. Protein Oxidative Modifications: Beneficial Roles in Disease and Health. J. Biochem. Pharmacol. Res. 2013, 1, 15–26. [Google Scholar] [PubMed]
- Nystrom, T. Role of oxidative carbonylation in protein quality control and senescence. EMBO J. 2005, 24, 1311–1317. [Google Scholar] [CrossRef]
- Ramana, K.V.; Srivastava, S.; Singhal, S.S. Lipid Peroxidation Products in Human Health and Disease 2019. Oxid. Med. Cell. Longev. 2019, 2019, 7147235. [Google Scholar] [CrossRef] [Green Version]
- Haines, T.H.; Dencher, N.A. Cardiolipin: A proton trap for oxidative phosphorylation. FEBS Lett. 2002, 528, 35–39. [Google Scholar] [CrossRef] [Green Version]
- Houtkooper, R.H.; Vaz, F.M. Cardiolipin, the heart of mitochondrial metabolism. Cell. Mol. Life Sci. 2008, 65, 2493–2506. [Google Scholar] [CrossRef] [PubMed]
- Osman, C.; Voelker, D.R.; Langer, T. Making heads or tails of phospholipids in mitochondria. J. Cell Biol. 2011, 192, 7–16. [Google Scholar] [CrossRef] [Green Version]
- Vahaheikkila, M.; Peltomaa, T.; Rog, T.; Vazdar, M.; Poyry, S.; Vattulainen, I. How cardiolipin peroxidation alters the properties of the inner mitochondrial membrane? Chem. Phys. Lipids 2018, 214, 15–23. [Google Scholar] [CrossRef] [PubMed]
- Wong-Ekkabut, J.; Xu, Z.; Triampo, W.; Tang, I.M.; Tieleman, D.P.; Monticelli, L. Effect of lipid peroxidation on the properties of lipid bilayers: A molecular dynamics study. Biophys. J. 2007, 93, 4225–4236. [Google Scholar] [CrossRef] [Green Version]
- Oemer, G.; Koch, J.; Wohlfarter, Y.; Alam, M.T.; Lackner, K.; Sailer, S.; Neumann, L.; Lindner, H.H.; Watschinger, K.; Haltmeier, M.; et al. Phospholipid Acyl Chain Diversity Controls the Tissue-Specific Assembly of Mitochondrial Cardiolipins. Cell Rep. 2020, 30, 4281–4291.e4. [Google Scholar] [CrossRef] [PubMed]
- Paradies, G.; Petrosillo, G.; Pistolese, M.; Di Venosa, N.; Federici, A.; Ruggiero, F.M. Decrease in mitochondrial complex I activity in ischemic/reperfused rat heart: Involvement of reactive oxygen species and cardiolipin. Circ. Res. 2004, 94, 53–59. [Google Scholar] [CrossRef] [Green Version]
- Mileykovskaya, E.; Dowhan, W. Cardiolipin-dependent formation of mitochondrial respiratory supercomplexes. Chem. Phys. Lipids 2014, 179, 42–48. [Google Scholar] [CrossRef] [Green Version]
- Paradies, G.; Petrosillo, G.; Paradies, V.; Ruggiero, F.M. Role of cardiolipin peroxidation and Ca2+ in mitochondrial dysfunction and disease. Cell Calcium 2009, 45, 643–650. [Google Scholar] [CrossRef] [PubMed]
- Raja, V.; Greenberg, M.L. The functions of cardiolipin in cellular metabolism-potential modifiers of the Barth syndrome phenotype. Chem. Phys. Lipids 2014, 179, 49–56. [Google Scholar] [CrossRef] [Green Version]
- Orrenius, S.; Zhivotovsky, B. Cardiolipin oxidation sets cytochrome c free. Nat. Chem. Biol. 2005, 1, 188–189. [Google Scholar] [CrossRef]
- Li, X.X.; Tsoi, B.; Li, Y.F.; Kurihara, H.; He, R.R. Cardiolipin and its different properties in mitophagy and apoptosis. J. Histochem. Cytochem. 2015, 63, 301–311. [Google Scholar] [CrossRef] [Green Version]
- Manoharan, S.; Kolanjiappan, K.; Suresh, K.; Panjamurthy, K. Lipid peroxidation & antioxidants status in patients with oral squamous cell carcinoma. Indian J. Med. Res. 2005, 122, 529–534. [Google Scholar] [PubMed]
- Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef] [PubMed]
- Lechuga-Sancho, A.M.; Gallego-Andujar, D.; Ruiz-Ocana, P.; Visiedo, F.M.; Saez-Benito, A.; Schwarz, M.; Segundo, C.; Mateos, R.M. Obesity induced alterations in redox homeostasis and oxidative stress are present from an early age. PLoS ONE 2018, 13, e0191547. [Google Scholar] [CrossRef] [PubMed]
- Scudamore, O.; Ciossek, T. Increased Oxidative Stress Exacerbates alpha-Synuclein Aggregation In Vivo. J. Neuropathol. Exp. Neurol. 2018, 77, 443–453. [Google Scholar] [CrossRef] [PubMed]
- Keller, J.N.; Schmitt, F.A.; Scheff, S.W.; Ding, Q.; Chen, Q.; Butterfield, D.A.; Markesbery, W.R. Evidence of increased oxidative damage in subjects with mild cognitive impairment. Neurology 2005, 64, 1152–1156. [Google Scholar] [CrossRef] [PubMed]
- Narula, J.; Pandey, P.; Arbustini, E.; Haider, N.; Narula, N.; Kolodgie, F.D.; Dal Bello, B.; Semigran, M.J.; Bielsa-Masdeu, A.; Dec, G.W.; et al. Apoptosis in heart failure: Release of cytochrome c from mitochondria and activation of caspase-3 in human cardiomyopathy. Proc. Natl. Acad. Sci. USA 1999, 96, 8144–8149. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.; Gong, Q.; Stice, J.P.; Knowlton, A.A. Mitochondrial OPA1, apoptosis, and heart failure. Cardiovasc. Res. 2009, 84, 91–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hanna, M.G.; Nelson, I.P.; Morgan-Hughes, J.A.; Wood, N.W. MELAS: A new disease associated mitochondrial DNA mutation and evidence for further genetic heterogeneity. J. Neurol. Neurosurg. Psychiatry 1998, 65, 512–517. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Zhang, Y.; Liu, X.; Zuo, J.; Wang, K.; Liu, W.; Ge, J. Exogenous taurine attenuates mitochondrial oxidative stress and endoplasmic reticulum stress in rat cardiomyocytes. Acta Biochim. Biophys. Sin. 2013, 45, 359–367. [Google Scholar] [CrossRef] [Green Version]
- Takatani, T.; Takahashi, K.; Uozumi, Y.; Shikata, E.; Yamamoto, Y.; Ito, T.; Matsuda, T.; Schaffer, S.W.; Fujio, Y.; Azuma, J. Taurine inhibits apoptosis by preventing formation of the Apaf-1/caspase-9 apoptosome. Am. J. Physiol. Cell Physiol. 2004, 287, C949–C953. [Google Scholar] [CrossRef] [Green Version]
- Niu, X.; Zheng, S.; Liu, H.; Li, S. Protective effects of taurine against inflammation, apoptosis, and oxidative stress in brain injury. Mol. Med. Rep. 2018, 18, 4516–4522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, R.; Wang, X.; Gao, Q.; Jiang, H.; Zhang, S.; Lu, M.; Liu, F.; Xue, X. Taurine Supplementation Reverses Diabetes-Induced Podocytes Injury via Modulation of the CSE/TRPC6 Axis and Improvement of Mitochondrial Function. Nephron 2020, 144, 84–95. [Google Scholar] [CrossRef] [PubMed]
- Homma, K.; Toda, E.; Osada, H.; Nagai, N.; Era, T.; Tsubota, K.; Okano, H.; Ozawa, Y. Taurine rescues mitochondria-related metabolic impairments in the patient-derived induced pluripotent stem cells and epithelial-mesenchymal transition in the retinal pigment epithelium. Redox Biol. 2021, 41, 101921. [Google Scholar] [CrossRef]
- Shetewy, A.; Shimada-Takaura, K.; Warner, D.; Jong, C.J.; Mehdi, A.B.; Alexeyev, M.; Takahashi, K.; Schaffer, S.W. Mitochondrial defects associated with beta-alanine toxicity: Relevance to hyper-beta-alaninemia. Mol. Cell. Biochem. 2016, 416, 11–22. [Google Scholar] [CrossRef] [Green Version]
- Jong, C.J.; Azuma, J.; Schaffer, S. Mechanism underlying the antioxidant activity of taurine: Prevention of mitochondrial oxidant production. Amino Acids 2012, 42, 2223–2232. [Google Scholar] [CrossRef]
- Ommati, M.M.; Heidari, R.; Ghanbarinejad, V.; Abdoli, N.; Niknahad, H. Taurine Treatment Provides Neuroprotection in a Mouse Model of Manganism. Biol. Trace Elem. Res. 2019, 190, 384–395. [Google Scholar] [CrossRef] [PubMed]
- Thirupathi, A.; Freitas, S.; Sorato, H.R.; Pedroso, G.S.; Effting, P.S.; Damiani, A.P.; Andrade, V.M.; Nesi, R.T.; Gupta, R.C.; Muller, A.P.; et al. Modulatory effects of taurine on metabolic and oxidative stress parameters in a mice model of muscle overuse. Nutrition 2018, 54, 158–164. [Google Scholar] [CrossRef]
- Oudit, G.Y.; Trivieri, M.G.; Khaper, N.; Husain, T.; Wilson, G.J.; Liu, P.; Sole, M.J.; Backx, P.H. Taurine supplementation reduces oxidative stress and improves cardiovascular function in an iron-overload murine model. Circulation 2004, 109, 1877–1885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Q.; Fan, W.; Cai, Y.; Wu, Q.; Mo, L.; Huang, Z.; Huang, H. Protective effects of taurine in traumatic brain injury via mitochondria and cerebral blood flow. Amino Acids 2016, 48, 2169–2177. [Google Scholar] [CrossRef] [PubMed]
- Jamshidzadeh, A.; Heidari, R.; Abasvali, M.; Zarei, M.; Ommati, M.M.; Abdoli, N.; Khodaei, F.; Yeganeh, Y.; Jafari, F.; Zarei, A.; et al. Taurine treatment preserves brain and liver mitochondrial function in a rat model of fulminant hepatic failure and hyperammonemia. Biomed. Pharmacother. 2017, 86, 514–520. [Google Scholar] [CrossRef] [PubMed]
- Huxtable, R.J. Physiological actions of taurine. Physiol. Rev. 1992, 72, 101–163. [Google Scholar] [CrossRef] [Green Version]
- Stipanuk, M.H. Role of the liver in regulation of body cysteine and taurine levels: A brief review. Neurochem. Res. 2004, 29, 105–110. [Google Scholar] [CrossRef]
- Heird, W.C. Taurine in neonatal nutrition—Revisited. Arch. Dis. Child. Fetal Neonatal Ed. 2004, 89, F473–F474. [Google Scholar] [CrossRef] [Green Version]
- Wojcik, O.P.; Koenig, K.L.; Zeleniuch-Jacquotte, A.; Costa, M.; Chen, Y. The potential protective effects of taurine on coronary heart disease. Atherosclerosis 2010, 208, 19–25. [Google Scholar] [CrossRef] [Green Version]
- Yamori, Y.; Taguchi, T.; Hamada, A.; Kunimasa, K.; Mori, H.; Mori, M. Taurine in health and diseases: Consistent evidence from experimental and epidemiological studies. J. Biomed. Sci. 2010, 17 (Suppl. 1), S6. [Google Scholar] [CrossRef] [Green Version]
- Galeano, N.F.; Darling, P.; Lepage, G.; Leroy, C.; Collet, S.; Giguere, R.; Roy, C.C. Taurine supplementation of a premature formula improves fat absorption in preterm infants. Pediatr. Res. 1987, 22, 67–71. [Google Scholar] [CrossRef] [Green Version]
- Taurine deficiency in a child on total parenteral nutrition. Nutr. Rev. 1985, 43, 81–83. [CrossRef]
- Chesney, R.W.; Helms, R.A.; Christensen, M.; Budreau, A.M.; Han, X.; Sturman, J.A. The role of taurine in infant nutrition. Adv. Exp. Med. Biol. 1998, 442, 463–476. [Google Scholar] [CrossRef] [PubMed]
- Lourenco, R.; Camilo, M.E. Taurine: A conditionally essential amino acid in humans? An overview in health and disease. Nutr. Hosp. 2002, 17, 262–270. [Google Scholar]
- Verner, A.; Craig, S.; McGuire, W. Effect of taurine supplementation on growth and development in preterm or low birth weight infants. Cochrane Database Syst. Rev. 2007, 4, CD006072. [Google Scholar] [CrossRef]
- Gaull, G.E. Taurine in pediatric nutrition: Review and update. Pediatrics 1989, 83, 433–442. [Google Scholar] [PubMed]
- Backus, R.C.; Ko, K.S.; Fascetti, A.J.; Kittleson, M.D.; Macdonald, K.A.; Maggs, D.J.; Berg, J.R.; Rogers, Q.R. Low plasma taurine concentration in Newfoundland dogs is associated with low plasma methionine and cyst(e)ine concentrations and low taurine synthesis. J. Nutr. 2006, 136, 2525–2533. [Google Scholar] [CrossRef] [Green Version]
- Hayes, K.C.; Trautwein, E.A. Taurine deficiency syndrome in cats. Vet. Clin. N. Am. Small Anim. Pract. 1989, 19, 403–413. [Google Scholar] [CrossRef]
- Novotny, M.J.; Hogan, P.M.; Flannigan, G. Echocardiographic evidence for myocardial failure induced by taurine deficiency in domestic cats. Can. J. Vet. Res. 1994, 58, 6–12. [Google Scholar]
- Pion, P.D.; Kittleson, M.D.; Skiles, M.L.; Rogers, Q.R.; Morris, J.G. Dilated cardiomyopathy associated with taurine deficiency in the domestic cat: Relationship to diet and myocardial taurine content. Adv. Exp. Med. Biol. 1992, 315, 63–73. [Google Scholar] [CrossRef]
- Barnett, K.C.; Burger, I.H. Taurine deficiency retinopathy in the cat. J. Small Anim. Pract. 1980, 21, 521–534. [Google Scholar] [CrossRef]
- Leon, A.; Levick, W.R.; Sarossy, M.G. Lesion topography and new histological features in feline taurine deficiency retinopathy. Exp. Eye Res. 1995, 61, 731–741. [Google Scholar] [CrossRef]
- Madl, J.E.; McIlnay, T.R.; Powell, C.C.; Gionfriddo, J.R. Depletion of taurine and glutamate from damaged photoreceptors in the retinas of dogs with primary glaucoma. Am. J. Vet. Res. 2005, 66, 791–799. [Google Scholar] [CrossRef]
- Fariello, R.G.; Lloyd, K.G.; Hornykiewicz, O. Cortical and subcortical projected foci in cats: Inhibitory action of taurine. Neurology 1975, 25, 1077–1083. [Google Scholar] [CrossRef] [PubMed]
- Sturman, J.A.; Moretz, R.C.; French, J.H.; Wisniewski, H.M. Taurine deficiency in the developing cat: Persistence of the cerebellar external granule cell layer. J. Neurosci. Res. 1985, 13, 405–416. [Google Scholar] [CrossRef] [PubMed]
- Schuller-Levis, G.; Mehta, P.D.; Rudelli, R.; Sturman, J. Immunologic consequences of taurine deficiency in cats. J. Leukoc. Biol. 1990, 47, 321–331. [Google Scholar] [CrossRef] [PubMed]
- Dieter, J.A.; Stewart, D.R.; Haggarty, M.A.; Stabenfeldt, G.H.; Lasley, B.L. Pregnancy failure in cats associated with long-term dietary taurine insufficiency. J. Reprod. Fertil. Suppl. 1993, 47, 457–463. [Google Scholar]
- Sturman, J.A.; Gargano, A.D.; Messing, J.M.; Imaki, H. Feline maternal taurine deficiency: Effect on mother and offspring. J. Nutr. 1986, 116, 655–667. [Google Scholar] [CrossRef]
- Backus, R.C.; Rogers, Q.R.; Rosenquist, G.L.; Calam, J.; Morris, J.G. Diets causing taurine depletion in cats substantially elevate postprandial plasma cholecystokinin concentration. J. Nutr. 1995, 125, 2650–2657. [Google Scholar] [CrossRef]
- Rabin, B.; Nicolosi, R.J.; Hayes, K.C. Dietary influence on bile acid conjugation in the cat. J. Nutr. 1976, 106, 1241–1246. [Google Scholar] [CrossRef] [Green Version]
- Backus, R.C.; Cohen, G.; Pion, P.D.; Good, K.L.; Rogers, Q.R.; Fascetti, A.J. Taurine deficiency in Newfoundlands fed commercially available complete and balanced diets. J. Am. Vet. Med. Assoc. 2003, 223, 1130–1136. [Google Scholar] [CrossRef]
- Pion, P.D.; Kittleson, M.D.; Thomas, W.P.; Delellis, L.A.; Rogers, Q.R. Response of cats with dilated cardiomyopathy to taurine supplementation. J. Am. Vet. Med. Assoc. 1992, 201, 275–284. [Google Scholar] [PubMed]
- van Gelder, N.M.; Koyama, I.; Jasper, H.H. Taurine treatment of spontaneous chronic epilepsy in a cat. Epilepsia 1977, 18, 45–54. [Google Scholar] [CrossRef]
- Berson, E.L.; Hayes, K.C.; Rabin, A.R.; Schmidt, S.Y.; Watson, G. Retinal degeneration in cats fed casein. II. Supplementation with methionine, cysteine, or taurine. Investig. Ophthalmol. 1976, 15, 52–58. [Google Scholar]
- Sturman, J.A.; Messing, J.M. Dietary taurine content and feline reproduction and outcome. J. Nutr. 1991, 121, 1195–1203. [Google Scholar] [CrossRef] [PubMed]
- Caine, J.J.; Geracioti, T.D. Taurine, energy drinks, and neuroendocrine effects. Clevel. Clin. J. Med. 2016, 83, 895–904. [Google Scholar] [CrossRef] [Green Version]
- Higgins, J.P.; Tuttle, T.D.; Higgins, C.L. Energy beverages: Content and safety. Mayo Clin. Proc. 2010, 85, 1033–1041. [Google Scholar] [CrossRef] [Green Version]
- Kurtz, J.A.; VanDusseldorp, T.A.; Doyle, J.A.; Otis, J.S. Taurine in sports and exercise. J. Int. Soc. Sports Nutr. 2021, 18, 39. [Google Scholar] [CrossRef]
- Seidel, U.; Huebbe, P.; Rimbach, G. Taurine: A Regulator of Cellular Redox Homeostasis and Skeletal Muscle Function. Mol. Nutr. Food Res. 2019, 63, e1800569. [Google Scholar] [CrossRef] [PubMed]
- Ghandforoush-Sattari, M.; Mashayekhi, S.; Krishna, C.V.; Thompson, J.P.; Routledge, P.A. Pharmacokinetics of oral taurine in healthy volunteers. J. Amino Acids 2010, 2010, 346237. [Google Scholar] [CrossRef] [Green Version]
- Sturman, J.A.; Hepner, G.W.; Hofmann, A.F.; Thomas, P.J. Metabolism of [35S]taurine in man. J. Nutr. 1975, 105, 1206–1214. [Google Scholar] [CrossRef] [PubMed]
- Ito, T.; Oishi, S.; Takai, M.; Kimura, Y.; Uozumi, Y.; Fujio, Y.; Schaffer, S.W.; Azuma, J. Cardiac and skeletal muscle abnormality in taurine transporter-knockout mice. J. Biomed. Sci. 2010, 17 (Suppl. 1), S20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Warskulat, U.; Flogel, U.; Jacoby, C.; Hartwig, H.G.; Thewissen, M.; Merx, M.W.; Molojavyi, A.; Heller-Stilb, B.; Schrader, J.; Haussinger, D. Taurine transporter knockout depletes muscle taurine levels and results in severe skeletal muscle impairment but leaves cardiac function uncompromised. FASEB J. 2004, 18, 577–579. [Google Scholar] [CrossRef] [Green Version]
- Garcia-Ayuso, D.; Di Pierdomenico, J.; Valiente-Soriano, F.J.; Martinez-Vacas, A.; Agudo-Barriuso, M.; Vidal-Sanz, M.; Picaud, S.; Villegas-Perez, M.P. beta-alanine supplementation induces taurine depletion and causes alterations of the retinal nerve fiber layer and axonal transport by retinal ganglion cells. Exp. Eye Res. 2019, 188, 107781. [Google Scholar] [CrossRef] [PubMed]
- Jong, C.J.; Ito, T.; Mozaffari, M.; Azuma, J.; Schaffer, S. Effect of beta-alanine treatment on mitochondrial taurine level and 5-taurinomethyluridine content. J. Biomed. Sci. 2010, 17 (Suppl. 1), S25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lake, N. Depletion of taurine in the adult rat retina. Neurochem. Res. 1982, 7, 1385–1390. [Google Scholar] [CrossRef]
- Pasantes-Morales, H.; Quesada, O.; Carabez, A.; Huxtable, R.J. Effects of the taurine transport antagonist, guanidinoethane sulfonate, and beta-alanine on the morphology of rat retina. J. Neurosci. Res. 1983, 9, 135–143. [Google Scholar] [CrossRef]
- Han, X.; Patters, A.B.; Ito, T.; Azuma, J.; Schaffer, S.W.; Chesney, R.W. Knockout of the TauT gene predisposes C57BL/6 mice to streptozotocin-induced diabetic nephropathy. PLoS ONE 2015, 10, e0117718. [Google Scholar] [CrossRef]
- Rascher, K.; Servos, G.; Berthold, G.; Hartwig, H.G.; Warskulat, U.; Heller-Stilb, B.; Haussinger, D. Light deprivation slows but does not prevent the loss of photoreceptors in taurine transporter knockout mice. Vision Res. 2004, 44, 2091–2100. [Google Scholar] [CrossRef] [PubMed]
- Warskulat, U.; Borsch, E.; Reinehr, R.; Heller-Stilb, B.; Monnighoff, I.; Buchczyk, D.; Donner, M.; Flogel, U.; Kappert, G.; Soboll, S.; et al. Chronic liver disease is triggered by taurine transporter knockout in the mouse. FASEB J. 2006, 20, 574–576. [Google Scholar] [CrossRef]
- Jong, C.J.; Ito, T.; Prentice, H.; Wu, J.Y.; Schaffer, S.W. Role of Mitochondria and Endoplasmic Reticulum in Taurine-Deficiency-Mediated Apoptosis. Nutrients 2017, 9, 795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jong, C.J.; Ito, T.; Azuma, J.; Schaffer, S. Taurine Depletion Decreases GRP78 Expression and Downregulates Perk-Dependent Activation of the Unfolded Protein Response. Adv. Exp. Med. Biol. 2015, 803, 571–579. [Google Scholar] [CrossRef] [PubMed]
- Jong, C.J.; Ito, T.; Schaffer, S.W. The ubiquitin-proteasome system and autophagy are defective in the taurine-deficient heart. Amino Acids 2015, 47, 2609–2622. [Google Scholar] [CrossRef]
- Ito, T.; Yoshikawa, N.; Inui, T.; Miyazaki, N.; Schaffer, S.W.; Azuma, J. Tissue depletion of taurine accelerates skeletal muscle senescence and leads to early death in mice. PLoS ONE 2014, 9, e107409. [Google Scholar] [CrossRef] [Green Version]
- Azari, J.; Bahl, J.; Huxtable, R. Guanidinoethyl sulfonate and other inhibitors of the taurine transporting system in the heart. Proc. West. Pharmacol. Soc. 1979, 22, 389–393. [Google Scholar] [PubMed]
- Huxtable, R.J.; Laird, H.E., 2nd; Lippincott, S.E. The transport of taurine in the heart and the rapid depletion of tissue taurine content by guanidinoethyl sulfonate. J. Pharmacol. Exp. Ther. 1979, 211, 465–471. [Google Scholar]
- Pansani, M.C.; Azevedo, P.S.; Rafacho, B.P.; Minicucci, M.F.; Chiuso-Minicucci, F.; Zorzella-Pezavento, S.G.; Marchini, J.S.; Padovan, G.J.; Fernandes, A.A.; Matsubara, B.B.; et al. Atrophic cardiac remodeling induced by taurine deficiency in Wistar rats. PLoS ONE 2012, 7, e41439. [Google Scholar] [CrossRef]
- Parildar, H.; Dogru-Abbasoglu, S.; Mehmetcik, G.; Ozdemirler, G.; Kocak-Toker, N.; Uysal, M. Lipid peroxidation potential and antioxidants in the heart tissue of beta-alanine- or taurine-treated old rats. J. Nutr. Sci. Vitaminol. 2008, 54, 61–65. [Google Scholar] [CrossRef] [Green Version]
- Jong, C.J.; Azuma, J.; Schaffer, S.W. Role of mitochondrial permeability transition in taurine deficiency-induced apoptosis. Exp. Clin. Cardiol. 2011, 16, 125–128. [Google Scholar] [PubMed]
- Schaffer, S.W.; Ballard-Croft, C.; Azuma, J.; Takahashi, K.; Kakhniashvili, D.G.; Jenkins, T.E. Shape and size changes induced by taurine depletion in neonatal cardiomyocytes. Amino Acids 1998, 15, 135–142. [Google Scholar] [CrossRef]
- Suzuki, T.; Suzuki, T.; Wada, T.; Saigo, K.; Watanabe, K. Taurine as a constituent of mitochondrial tRNAs: New insights into the functions of taurine and human mitochondrial diseases. EMBO J. 2002, 21, 6581–6589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wada, T.; Shimazaki, T.; Nakagawa, S.; Otuki, T.; Kurata, S.; Suzuki, T.; Watanabe, K.; Saigo, K. Chemical synthesis of novel taurine-containing uridine derivatives. Nucleic Acids Res. Suppl. 2002, 2, 11–12. [Google Scholar] [CrossRef] [PubMed]
- Fakruddin, M.; Wei, F.Y.; Suzuki, T.; Asano, K.; Kaieda, T.; Omori, A.; Izumi, R.; Fujimura, A.; Kaitsuka, T.; Miyata, K.; et al. Defective Mitochondrial tRNA Taurine Modification Activates Global Proteostress and Leads to Mitochondrial Disease. Cell Rep. 2018, 22, 482–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kirino, Y.; Goto, Y.; Campos, Y.; Arenas, J.; Suzuki, T. Specific correlation between the wobble modification deficiency in mutant tRNAs and the clinical features of a human mitochondrial disease. Proc. Natl. Acad. Sci. USA 2005, 102, 7127–7132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kirino, Y.; Yasukawa, T.; Ohta, S.; Akira, S.; Ishihara, K.; Watanabe, K.; Suzuki, T. Codon-specific translational defect caused by a wobble modification deficiency in mutant tRNA from a human mitochondrial disease. Proc. Natl. Acad. Sci. USA 2004, 101, 15070–15075. [Google Scholar] [CrossRef] [Green Version]
- Asano, K.; Suzuki, T.; Saito, A.; Wei, F.Y.; Ikeuchi, Y.; Numata, T.; Tanaka, R.; Yamane, Y.; Yamamoto, T.; Goto, T.; et al. Metabolic and chemical regulation of tRNA modification associated with taurine deficiency and human disease. Nucleic Acids Res. 2018, 46, 1565–1583. [Google Scholar] [CrossRef] [Green Version]
- Aruoma, O.I.; Halliwell, B.; Hoey, B.M.; Butler, J. The antioxidant action of taurine, hypotaurine and their metabolic precursors. Biochem. J. 1988, 256, 251–255. [Google Scholar] [CrossRef] [PubMed]
- Li, J.X.; Pang, Y.Z.; Tang, C.S.; Li, Z.Q. Protective effect of taurine on hypochlorous acid toxicity to nuclear nucleoside triphosphatase in isolated nuclei from rat liver. World J. Gastroenterol. 2004, 10, 694–698. [Google Scholar] [CrossRef] [PubMed]
- Cheong, S.H.; Lee, D.S. Taurine Chloramine Prevents Neuronal HT22 Cell Damage Through Nrf2-Related Heme Oxygenase-1. Adv. Exp. Med. Biol. 2017, 975 Pt 1, 145–157. [Google Scholar] [CrossRef]
- Kang, I.S.; Kim, C. Taurine chloramine administered in vivo increases NRF2-regulated antioxidant enzyme expression in murine peritoneal macrophages. Adv. Exp. Med. Biol. 2013, 775, 259–267. [Google Scholar] [CrossRef]
- Kim, C.; Cha, Y.N. Taurine chloramine produced from taurine under inflammation provides anti-inflammatory and cytoprotective effects. Amino Acids 2014, 46, 89–100. [Google Scholar] [CrossRef] [PubMed]
- Higuchi, M.; Celino, F.T.; Shimizu-Yamaguchi, S.; Miura, C.; Miura, T. Taurine plays an important role in the protection of spermatogonia from oxidative stress. Amino Acids 2012, 43, 2359–2369. [Google Scholar] [CrossRef]
- Okado-Matsumoto, A.; Fridovich, I. Subcellular distribution of superoxide dismutases (SOD) in rat liver: Cu,Zn-SOD in mitochondria. J. Biol. Chem. 2001, 276, 38388–38393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sturtz, L.A.; Diekert, K.; Jensen, L.T.; Lill, R.; Culotta, V.C. A fraction of yeast Cu,Zn-superoxide dismutase and its metallochaperone, CCS, localize to the intermembrane space of mitochondria. A physiological role for SOD1 in guarding against mitochondrial oxidative damage. J. Biol. Chem. 2001, 276, 38084–38089. [Google Scholar] [CrossRef]
- Tabassum, H.; Rehman, H.; Banerjee, B.D.; Raisuddin, S.; Parvez, S. Attenuation of tamoxifen-induced hepatotoxicity by taurine in mice. Clin. Chim. Acta 2006, 370, 129–136. [Google Scholar] [CrossRef]
- Pasantes-Morales, H.; Cruz, C. Taurine and hypotaurine inhibit light-induced lipid peroxidation and protect rod outer segment structure. Brain Res. 1985, 330, 154–157. [Google Scholar] [CrossRef]
- Pasantes-Morales, H.; Cruz, C. Taurine: A physiological stabilizer of photoreceptor membranes. Prog. Clin. Biol. Res. 1985, 179, 371–381. [Google Scholar] [PubMed]
- Pasantes-Morales, H.; Wright, C.E.; Gaull, G.E. Taurine protection of lymphoblastoid cells from iron-ascorbate induced damage. Biochem. Pharmacol. 1985, 34, 2205–2207. [Google Scholar] [CrossRef]
- Hansen, S.H.; Andersen, M.L.; Cornett, C.; Gradinaru, R.; Grunnet, N. A role for taurine in mitochondrial function. J. Biomed. Sci. 2010, 17 (Suppl. 1), S23. [Google Scholar] [CrossRef] [Green Version]
- El Idrissi, A. Taurine increases mitochondrial buffering of calcium: Role in neuroprotection. Amino Acids 2008, 34, 321–328. [Google Scholar] [CrossRef]
- El Idrissi, A.; Trenkner, E. Growth factors and taurine protect against excitotoxicity by stabilizing calcium homeostasis and energy metabolism. J. Neurosci. 1999, 19, 9459–9468. [Google Scholar] [CrossRef]
- El Idrissi, A.; Trenkner, E. Taurine regulates mitochondrial calcium homeostasis. Adv. Exp. Med. Biol. 2003, 526, 527–536. [Google Scholar] [CrossRef]
- El Idrissi, A.; Trenkner, E. Taurine as a modulator of excitatory and inhibitory neurotransmission. Neurochem. Res. 2004, 29, 189–197. [Google Scholar] [CrossRef]
- Trenkner, E.; el Idrissi, A.; Harris, C. Balanced interaction of growth factors and taurine regulate energy metabolism, neuronal survival, and function of cultured mouse cerebellar cells under depolarizing conditions. Adv. Exp. Med. Biol. 1996, 403, 507–517. [Google Scholar] [CrossRef]
- Bkaily, G.; Jaalouk, D.; Sader, S.; Shbaklo, H.; Pothier, P.; Jacques, D.; D’Orleans-Juste, P.; Cragoe, E.J., Jr.; Bose, R. Taurine indirectly increases [Ca]i by inducing Ca2+ influx through the Na(+)-Ca2+ exchanger. Mol. Cell Biochem. 1998, 188, 187–197. [Google Scholar] [CrossRef]
- Schaffer, S.; Solodushko, V.; Azuma, J. Taurine-deficient cardiomyopathy: Role of phospholipids, calcium and osmotic stress. Adv. Exp. Med. Biol. 2000, 483, 57–69. [Google Scholar] [CrossRef]
- Schaffer, S.W.; Punna, S.; Duan, J.; Harada, H.; Hamaguchi, T.; Azuma, J. Mechanism underlying physiological modulation of myocardial contraction by taurine. Adv. Exp. Med. Biol. 1992, 315, 193–198. [Google Scholar] [CrossRef]
- Takahashi, K.; Harada, H.; Schaffer, S.W.; Azuma, J. Effect of taurine on intracellular calcium dynamics of cultured myocardial cells during the calcium paradox. Adv. Exp. Med. Biol. 1992, 315, 153–161. [Google Scholar] [CrossRef] [PubMed]
- Steele, D.S.; Smith, G.L.; Miller, D.J. The effects of taurine on Ca2+ uptake by the sarcoplasmic reticulum and Ca2+ sensitivity of chemically skinned rat heart. J. Physiol. 1990, 422, 499–511. [Google Scholar] [CrossRef] [Green Version]
- Galler, S.; Hutzler, C.; Haller, T. Effects of taurine on Ca2(+)-dependent force development of skinned muscle fibre preparations. J. Exp. Biol. 1990, 152, 255–264. [Google Scholar] [CrossRef]
- Griffiths, E.J.; Rutter, G.A. Mitochondrial calcium as a key regulator of mitochondrial ATP production in mammalian cells. Biochim. Biophys. Acta 2009, 1787, 1324–1333. [Google Scholar] [CrossRef] [Green Version]
- Chen, M.; Guerrero, A.D.; Huang, L.; Shabier, Z.; Pan, M.; Tan, T.H.; Wang, J. Caspase-9-induced mitochondrial disruption through cleavage of anti-apoptotic BCL-2 family members. J. Biol. Chem. 2007, 282, 33888–33895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leon, R.; Wu, H.; Jin, Y.; Wei, J.; Buddhala, C.; Prentice, H.; Wu, J.Y. Protective function of taurine in glutamate-induced apoptosis in cultured neurons. J. Neurosci. Res. 2009, 87, 1185–1194. [Google Scholar] [CrossRef]
- Menzie, J.; Prentice, H.; Wu, J.Y. Neuroprotective Mechanisms of Taurine against Ischemic Stroke. Brain Sci. 2013, 3, 877–907. [Google Scholar] [CrossRef] [Green Version]
- Wu, J.Y.; Prentice, H. Role of taurine in the central nervous system. J. Biomed. Sci. 2010, 17 (Suppl. 1), S1. [Google Scholar] [CrossRef] [Green Version]
- Taranukhin, A.G.; Taranukhina, E.Y.; Saransaari, P.; Podkletnova, I.M.; Pelto-Huikko, M.; Oja, S.S. Neuroprotection by taurine in ethanol-induced apoptosis in the developing cerebellum. J. Biomed. Sci. 2010, 17 (Suppl. 1), S12. [Google Scholar] [CrossRef] [Green Version]
- Azuma, J.; Sawamura, A.; Awata, N. Usefulness of taurine in chronic congestive heart failure and its prospective application. Jpn. Circ. J. 1992, 56, 95–99. [Google Scholar] [CrossRef] [Green Version]
- Di Lorenzo, A.; Iannuzzo, G.; Parlato, A.; Cuomo, G.; Testa, C.; Coppola, M.; D’Ambrosio, G.; Oliviero, D.A.; Sarullo, S.; Vitale, G.; et al. Clinical Evidence for Q10 Coenzyme Supplementation in Heart Failure: From Energetics to Functional Improvement. J. Clin. Med. 2020, 9, 1266. [Google Scholar] [CrossRef]
- Jafari, M.; Mousavi, S.M.; Asgharzadeh, A.; Yazdani, N. Coenzyme Q10 in the treatment of heart failure: A systematic review of systematic reviews. Indian Heart J. 2018, 70 (Suppl. 1), S111–S117. [Google Scholar] [CrossRef]
- Sharma, A.; Fonarow, G.C.; Butler, J.; Ezekowitz, J.A.; Felker, G.M. Coenzyme Q10 and Heart Failure: A State-of-the-Art Review. Circ. Heart Fail. 2016, 9, e002639. [Google Scholar] [CrossRef]
- Doenst, T.; Nguyen, T.D.; Abel, E.D. Cardiac metabolism in heart failure: Implications beyond ATP production. Circ. Res. 2013, 113, 709–724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sheeran, F.L.; Pepe, S. Energy deficiency in the failing heart: Linking increased reactive oxygen species and disruption of oxidative phosphorylation rate. Biochim. Biophys. Acta 2006, 1757, 543–552. [Google Scholar] [CrossRef] [Green Version]
- Militante, J.D.; Lombardini, J.B. Treatment of hypertension with oral taurine: Experimental and clinical studies. Amino Acids 2002, 23, 381–393. [Google Scholar] [CrossRef] [PubMed]
- Sun, Q.; Wang, B.; Li, Y.; Sun, F.; Li, P.; Xia, W.; Zhou, X.; Li, Q.; Wang, X.; Chen, J.; et al. Taurine Supplementation Lowers Blood Pressure and Improves Vascular Function in Prehypertension: Randomized, Double-Blind, Placebo-Controlled Study. Hypertension 2016, 67, 541–549. [Google Scholar] [CrossRef] [Green Version]
- Sagara, M.; Murakami, S.; Mizushima, S.; Liu, L.; Mori, M.; Ikeda, K.; Nara, Y.; Yamori, Y. Taurine in 24-h Urine Samples Is Inversely Related to Cardiovascular Risks of Middle Aged Subjects in 50 Populations of the World. Adv. Exp. Med. Biol. 2015, 803, 623–636. [Google Scholar] [CrossRef] [PubMed]
- Yamori, Y.; Liu, L.; Mori, M.; Sagara, M.; Murakami, S.; Nara, Y.; Mizushima, S. Taurine as the nutritional factor for the longevity of the Japanese revealed by a world-wide epidemiological survey. Adv. Exp. Med. Biol. 2009, 643, 13–25. [Google Scholar] [CrossRef]
- Yamori, Y.; Murakami, S.; Ikeda, K.; Nara, Y. Fish and lifestyle-related disease prevention: Experimental and epidemiological evidence for anti-atherogenic potential of taurine. Clin. Exp. Pharmacol. Physiol. 2004, 31 (Suppl. 2), S20–S23. [Google Scholar] [CrossRef]
- Yamori, Y.; Taguchi, T.; Mori, H.; Mori, M. Low cardiovascular risks in the middle aged males and females excreting greater 24-hour urinary taurine and magnesium in 41 WHO-CARDIAC study populations in the world. J. Biomed. Sci. 2010, 17 (Suppl. 1), S21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adedara, I.A.; Alake, S.E.; Olajide, L.O.; Adeyemo, M.O.; Ajibade, T.O.; Farombi, E.O. Taurine Ameliorates Thyroid Hypofunction and Renal Injury in L-NAME-Induced Hypertensive Rats. Drug Res. 2019, 69, 83–92. [Google Scholar] [CrossRef]
- Ibrahim, M.A.; Eraqi, M.M.; Alfaiz, F.A. Therapeutic role of taurine as antioxidant in reducing hypertension risks in rats. Heliyon 2020, 6, e03209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rahman, M.M.; Park, H.M.; Kim, S.J.; Go, H.K.; Kim, G.B.; Hong, C.U.; Lee, Y.U.; Kim, S.Z.; Kim, J.S.; Kang, H.S. Taurine prevents hypertension and increases exercise capacity in rats with fructose-induced hypertension. Am. J. Hypertens. 2011, 24, 574–581. [Google Scholar] [CrossRef] [Green Version]
- Zaric, B.L.; Radovanovic, J.N.; Gluvic, Z.; Stewart, A.J.; Essack, M.; Motwalli, O.; Gojobori, T.; Isenovic, E.R. Atherosclerosis Linked to Aberrant Amino Acid Metabolism and Immunosuppressive Amino Acid Catabolizing Enzymes. Front. Immunol. 2020, 11, 551758. [Google Scholar] [CrossRef] [PubMed]
- Dikalov, S.I.; Ungvari, Z. Role of mitochondrial oxidative stress in hypertension. Am. J. Physiol. Heart Circ. Physiol. 2013, 305, H1417–H1427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Esmaeili, F.; Maleki, V.; Kheirouri, S.; Alizadeh, M. The Effects of Taurine Supplementation on Metabolic Profiles, Pentosidine, Soluble Receptor of Advanced Glycation End Products and Methylglyoxal in Adults With Type 2 Diabetes: A Randomized, Double-Blind, Placebo-Controlled Trial. Can. J. Diabetes 2021, 45, 39–46. [Google Scholar] [CrossRef]
- Maleki, V.; Alizadeh, M.; Esmaeili, F.; Mahdavi, R. The effects of taurine supplementation on glycemic control and serum lipid profile in patients with type 2 diabetes: A randomized, double-blind, placebo-controlled trial. Amino Acids 2020, 52, 905–914. [Google Scholar] [CrossRef]
- Maleki, V.; Mahdavi, R.; Hajizadeh-Sharafabad, F.; Alizadeh, M. The effects of taurine supplementation on oxidative stress indices and inflammation biomarkers in patients with type 2 diabetes: A randomized, double-blind, placebo-controlled trial. Diabetol. Metab. Syndr. 2020, 12, 9. [Google Scholar] [CrossRef]
- De Luca, G.; Calpona, P.R.; Caponetti, A.; Romano, G.; Di Benedetto, A.; Cucinotta, D.; Di Giorgio, R.M. Taurine and osmoregulation: Platelet taurine content, uptake, and release in type 2 diabetic patients. Metabolism 2001, 50, 60–64. [Google Scholar] [CrossRef]
- Franconi, F.; Bennardini, F.; Mattana, A.; Miceli, M.; Ciuti, M.; Mian, M.; Gironi, A.; Anichini, R.; Seghieri, G. Plasma and platelet taurine are reduced in subjects with insulin-dependent diabetes mellitus: Effects of taurine supplementation. Am. J. Clin. Nutr. 1995, 61, 1115–1119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sak, D.; Erdenen, F.; Muderrisoglu, C.; Altunoglu, E.; Sozer, V.; Gungel, H.; Guler, P.A.; Sak, T.; Uzun, H. The Relationship between Plasma Taurine Levels and Diabetic Complications in Patients with Type 2 Diabetes Mellitus. Biomolecules 2019, 9, 96. [Google Scholar] [CrossRef] [Green Version]
- Trautwein, E.A.; Hayes, K.C. Plasma and whole blood taurine concentrations respond differently to taurine supplementation (humans) and depletion (cats). Z. Ernahrungswiss. 1995, 34, 137–142. [Google Scholar] [CrossRef] [PubMed]
- Haythorne, E.; Rohm, M.; van de Bunt, M.; Brereton, M.F.; Tarasov, A.I.; Blacker, T.S.; Sachse, G.; Silva Dos Santos, M.; Terron Exposito, R.; Davis, S.; et al. Diabetes causes marked inhibition of mitochondrial metabolism in pancreatic beta-cells. Nat. Commun. 2019, 10, 2474. [Google Scholar] [CrossRef]
- Hyeon, J.S.; Jung, Y.; Lee, G.; Ha, H.; Hwang, G.S. Urinary Metabolomic Profiling in Streptozotocin-Induced Diabetic Mice after Treatment with Losartan. Int. J. Mol. Sci. 2020, 21, 8969. [Google Scholar] [CrossRef] [PubMed]
- Trachtman, H.; Futterweit, S.; Maesaka, J.; Ma, C.; Valderrama, E.; Fuchs, A.; Tarectecan, A.A.; Rao, P.S.; Sturman, J.A.; Boles, T.H.; et al. Taurine ameliorates chronic streptozocin-induced diabetic nephropathy in rats. Am. J. Physiol. 1995, 269, F429–F438. [Google Scholar] [CrossRef] [PubMed]
- Evans, J.L.; Goldfine, I.D.; Maddux, B.A.; Grodsky, G.M. Oxidative stress and stress-activated signaling pathways: A unifying hypothesis of type 2 diabetes. Endocr. Rev. 2002, 23, 599–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haber, C.A.; Lam, T.K.; Yu, Z.; Gupta, N.; Goh, T.; Bogdanovic, E.; Giacca, A.; Fantus, I.G. N-acetylcysteine and taurine prevent hyperglycemia-induced insulin resistance in vivo: Possible role of oxidative stress. Am. J. Physiol. Endocrinol. Metab. 2003, 285, E744–E753. [Google Scholar] [CrossRef] [Green Version]
- Han, J.; Bae, J.H.; Kim, S.Y.; Lee, H.Y.; Jang, B.C.; Lee, I.K.; Cho, C.H.; Lim, J.G.; Suh, S.I.; Kwon, T.K.; et al. Taurine increases glucose sensitivity of UCP2-overexpressing beta-cells by ameliorating mitochondrial metabolism. Am. J. Physiol. Endocrinol. Metab. 2004, 287, E1008–E1018. [Google Scholar] [CrossRef]
- Ito, T.; Schaffer, S.W.; Azuma, J. The potential usefulness of taurine on diabetes mellitus and its complications. Amino Acids 2012, 42, 1529–1539. [Google Scholar] [CrossRef] [Green Version]
- Kim, K.S.; Oh, D.H.; Kim, J.Y.; Lee, B.G.; You, J.S.; Chang, K.J.; Chung, H.J.; Yoo, M.C.; Yang, H.I.; Kang, J.H.; et al. Taurine ameliorates hyperglycemia and dyslipidemia by reducing insulin resistance and leptin level in Otsuka Long-Evans Tokushima fatty (OLETF) rats with long-term diabetes. Exp. Mol. Med. 2012, 44, 665–673. [Google Scholar] [CrossRef] [Green Version]
- Chauncey, K.B.; Tenner, T.E., Jr.; Lombardini, J.B.; Jones, B.G.; Brooks, M.L.; Warner, R.D.; Davis, R.L.; Ragain, R.M. The effect of taurine supplementation on patients with type 2 diabetes mellitus. Adv. Exp. Med. Biol. 2003, 526, 91–96. [Google Scholar] [CrossRef]
- Nakamura, T.; Ushiyama, C.; Suzuki, S.; Shimada, N.; Ohmuro, H.; Ebihara, I.; Koide, H. Effects of taurine and vitamin E on microalbuminuria, plasma metalloproteinase-9, and serum type IV collagen concentrations in patients with diabetic nephropathy. Nephron 1999, 83, 361–362. [Google Scholar] [CrossRef]
- Rosa, F.T.; Freitas, E.C.; Deminice, R.; Jordao, A.A.; Marchini, J.S. Oxidative stress and inflammation in obesity after taurine supplementation: A double-blind, placebo-controlled study. Eur. J. Nutr. 2014, 53, 823–830. [Google Scholar] [CrossRef] [PubMed]
- Mizushima, S.; Nara, Y.; Sawamura, M.; Yamori, Y. Effects of oral taurine supplementation on lipids and sympathetic nerve tone. Adv. Exp. Med. Biol. 1996, 403, 615–622. [Google Scholar] [CrossRef] [PubMed]
- De Carvalho, F.G.; Brandao, C.F.C.; Batitucci, G.; Souza, A.O.; Ferrari, G.D.; Alberici, L.C.; Munoz, V.R.; Pauli, J.R.; De Moura, L.P.; Ropelle, E.R.; et al. Taurine supplementation associated with exercise increases mitochondrial activity and fatty acid oxidation gene expression in the subcutaneous white adipose tissue of obese women. Clin. Nutr. 2021, 40, 2180–2187. [Google Scholar] [CrossRef] [PubMed]
- Yamori, Y. Preliminary report of cardiac study: Cross-sectional multicenter study on dietary factors of cardiovascular diseases. CARDIAC Study Group. Clin. Exp. Hypertens. A 1989, 11, 957–972. [Google Scholar] [CrossRef]
- Harada, H.; Tsujino, T.; Watari, Y.; Nonaka, H.; Emoto, N.; Yokoyama, M. Oral taurine supplementation prevents fructose-induced hypertension in rats. Heart Vessels 2004, 19, 132–136. [Google Scholar] [CrossRef]
- Harada, N.; Ninomiya, C.; Osako, Y.; Morishima, M.; Mawatari, K.; Takahashi, A.; Nakaya, Y. Taurine alters respiratory gas exchange and nutrient metabolism in type 2 diabetic rats. Obes. Res. 2004, 12, 1077–1084. [Google Scholar] [CrossRef] [PubMed]
- Nandhini, A.T.; Thirunavukkarasu, V.; Ravichandran, M.K.; Anuradha, C.V. Effect of taurine on biomarkers of oxidative stress in tissues of fructose-fed insulin-resistant rats. Singap. Med. J. 2005, 46, 82–87. [Google Scholar]
- Nardelli, T.R.; Ribeiro, R.A.; Balbo, S.L.; Vanzela, E.C.; Carneiro, E.M.; Boschero, A.C.; Bonfleur, M.L. Taurine prevents fat deposition and ameliorates plasma lipid profile in monosodium glutamate-obese rats. Amino Acids 2011, 41, 901–908. [Google Scholar] [CrossRef]
- Tsuboyama-Kasaoka, N.; Shozawa, C.; Sano, K.; Kamei, Y.; Kasaoka, S.; Hosokawa, Y.; Ezaki, O. Taurine (2-aminoethanesulfonic acid) deficiency creates a vicious circle promoting obesity. Endocrinology 2006, 147, 3276–3284. [Google Scholar] [CrossRef] [Green Version]
- Fukuda, M.; Nagao, Y. Dynamic derangement in amino acid profile during and after a stroke-like episode in adult-onset mitochondrial disease: A case report. J. Med. Case Rep. 2019, 13, 313. [Google Scholar] [CrossRef]
- Che, Y.; Hou, L.; Sun, F.; Zhang, C.; Liu, X.; Piao, F.; Zhang, D.; Li, H.; Wang, Q. Taurine protects dopaminergic neurons in a mouse Parkinson’s disease model through inhibition of microglial M1 polarization. Cell Death Dis. 2018, 9, 435. [Google Scholar] [CrossRef] [Green Version]
- Hou, L.; Che, Y.; Sun, F.; Wang, Q. Taurine protects noradrenergic locus coeruleus neurons in a mouse Parkinson’s disease model by inhibiting microglial M1 polarization. Amino Acids 2018, 50, 547–556. [Google Scholar] [CrossRef] [PubMed]
- Jang, H.; Lee, S.; Choi, S.L.; Kim, H.Y.; Baek, S.; Kim, Y. Taurine Directly Binds to Oligomeric Amyloid-beta and Recovers Cognitive Deficits in Alzheimer Model Mice. Adv. Exp. Med. Biol. 2017, 975 Pt 1, 233–241. [Google Scholar] [CrossRef]
- Kim, H.Y.; Kim, H.V.; Yoon, J.H.; Kang, B.R.; Cho, S.M.; Lee, S.; Kim, J.Y.; Kim, J.W.; Cho, Y.; Woo, J.; et al. Taurine in drinking water recovers learning and memory in the adult APP/PS1 mouse model of Alzheimer’s disease. Sci. Rep. 2014, 4, 7467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oh, S.J.; Lee, H.J.; Jeong, Y.J.; Nam, K.R.; Kang, K.J.; Han, S.J.; Lee, K.C.; Lee, Y.J.; Choi, J.Y. Evaluation of the neuroprotective effect of taurine in Alzheimer’s disease using functional molecular imaging. Sci. Rep. 2020, 10, 15551. [Google Scholar] [CrossRef] [PubMed]
- Santa-Maria, I.; Hernandez, F.; Moreno, F.J.; Avila, J. Taurine, an inducer for tau polymerization and a weak inhibitor for amyloid-beta-peptide aggregation. Neurosci. Lett. 2007, 429, 91–94. [Google Scholar] [CrossRef]
- Avshalumov, M.V.; Rice, M.E. NMDA receptor activation mediates hydrogen peroxide-induced pathophysiology in rat hippocampal slices. J. Neurophysiol. 2002, 87, 2896–2903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carvajal, F.J.; Mattison, H.A.; Cerpa, W. Role of NMDA Receptor-Mediated Glutamatergic Signaling in Chronic and Acute Neuropathologies. Neural Plast. 2016, 2016, 2701526. [Google Scholar] [CrossRef]
- Esteras, N.; Kopach, O.; Maiolino, M.; Lariccia, V.; Amoroso, S.; Qamar, S.; Wray, S.; Rusakov, D.A.; Jaganjac, M.; Abramov, A.Y. Mitochondrial ROS control neuronal excitability and cell fate in frontotemporal dementia. Alzheimers Dement. 2021. [Google Scholar] [CrossRef]
- Rossi, A.; Pizzo, P.; Filadi, R. Calcium, mitochondria and cell metabolism: A functional triangle in bioenergetics. Biochim. Biophys. Acta Mol. Cell Res. 2019, 1866, 1068–1078. [Google Scholar] [CrossRef] [PubMed]
- Rossi, A.; Rigotto, G.; Valente, G.; Giorgio, V.; Basso, E.; Filadi, R.; Pizzo, P. Defective Mitochondrial Pyruvate Flux Affects Cell Bioenergetics in Alzheimer’s Disease-Related Models. Cell Rep. 2020, 30, 2332–2348.e10. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Wang, F.; Mai, D.; Qu, S. Molecular Mechanisms of Glutamate Toxicity in Parkinson’s Disease. Front. Neurosci. 2020, 14, 585584. [Google Scholar] [CrossRef] [PubMed]
- Johri, A.; Beal, M.F. Mitochondrial dysfunction in neurodegenerative diseases. J. Pharmacol. Exp. Ther. 2012, 342, 619–630. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.; Chen, M.; Jiang, J. Mitochondrial dysfunction in neurodegenerative diseases and drug targets via apoptotic signaling. Mitochondrion 2019, 49, 35–45. [Google Scholar] [CrossRef]
- Erickson, C.A.; Early, M.; Stigler, K.A.; Wink, L.K.; Mullett, J.E.; McDougle, C.J. An open-label naturalistic pilot study of acamprosate in youth with autistic disorder. J. Child. Adolesc. Psychopharmacol. 2011, 21, 565–569. [Google Scholar] [CrossRef] [Green Version]
- Erickson, C.A.; Mullett, J.E.; McDougle, C.J. Brief report: Acamprosate in fragile X syndrome. J. Autism Dev. Disord. 2010, 40, 1412–1416. [Google Scholar] [CrossRef]
- Erickson, C.A.; Ray, B.; Maloney, B.; Wink, L.K.; Bowers, K.; Schaefer, T.L.; McDougle, C.J.; Sokol, D.K.; Lahiri, D.K. Impact of acamprosate on plasma amyloid-beta precursor protein in youth: A pilot analysis in fragile X syndrome-associated and idiopathic autism spectrum disorder suggests a pharmacodynamic protein marker. J. Psychiatr. Res. 2014, 59, 220–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Erickson, C.A.; Wink, L.K.; Early, M.C.; Stiegelmeyer, E.; Mathieu-Frasier, L.; Patrick, V.; McDougle, C.J. Brief report: Pilot single-blind placebo lead-in study of acamprosate in youth with autistic disorder. J. Autism Dev. Disord. 2014, 44, 981–987. [Google Scholar] [CrossRef] [PubMed]
- Erickson, C.A.; Wink, L.K.; Ray, B.; Early, M.C.; Stiegelmeyer, E.; Mathieu-Frasier, L.; Patrick, V.; Lahiri, D.K.; McDougle, C.J. Impact of acamprosate on behavior and brain-derived neurotrophic factor: An open-label study in youth with fragile X syndrome. Psychopharmacology 2013, 228, 75–84. [Google Scholar] [CrossRef] [PubMed]
- Wright, T.M.; Myrick, H. Acamprosate: A new tool in the battle against alcohol dependence. Neuropsychiatr. Dis. Treat. 2006, 2, 445–453. [Google Scholar] [CrossRef] [Green Version]
- McDougle, C.J.; Erickson, C.A.; Stigler, K.A.; Posey, D.J. Neurochemistry in the pathophysiology of autism. J. Clin. Psychiatry 2005, 66 (Suppl. 10), 9–18. [Google Scholar]
- Silverman, J.L.; Tolu, S.S.; Barkan, C.L.; Crawley, J.N. Repetitive self-grooming behavior in the BTBR mouse model of autism is blocked by the mGluR5 antagonist MPEP. Neuropsychopharmacology 2010, 35, 976–989. [Google Scholar] [CrossRef] [PubMed]
- Yizhar, O.; Fenno, L.E.; Prigge, M.; Schneider, F.; Davidson, T.J.; O’Shea, D.J.; Sohal, V.S.; Goshen, I.; Finkelstein, J.; Paz, J.T.; et al. Neocortical excitation/inhibition balance in information processing and social dysfunction. Nature 2011, 477, 171–178. [Google Scholar] [CrossRef]
- Filipek, P.A.; Juranek, J.; Smith, M.; Mays, L.Z.; Ramos, E.R.; Bocian, M.; Masser-Frye, D.; Laulhere, T.M.; Modahl, C.; Spence, M.A.; et al. Mitochondrial dysfunction in autistic patients with 15q inverted duplication. Ann. Neurol. 2003, 53, 801–804. [Google Scholar] [CrossRef]
- Giulivi, C.; Zhang, Y.F.; Omanska-Klusek, A.; Ross-Inta, C.; Wong, S.; Hertz-Picciotto, I.; Tassone, F.; Pessah, I.N. Mitochondrial dysfunction in autism. JAMA 2010, 304, 2389–2396. [Google Scholar] [CrossRef] [Green Version]
- Oliveira, G.; Diogo, L.; Grazina, M.; Garcia, P.; Ataide, A.; Marques, C.; Miguel, T.; Borges, L.; Vicente, A.M.; Oliveira, C.R. Mitochondrial dysfunction in autism spectrum disorders: A population-based study. Dev. Med. Child. Neurol. 2005, 47, 185–189. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Jong, C.J.; Sandal, P.; Schaffer, S.W. The Role of Taurine in Mitochondria Health: More Than Just an Antioxidant. Molecules 2021, 26, 4913. https://doi.org/10.3390/molecules26164913
Jong CJ, Sandal P, Schaffer SW. The Role of Taurine in Mitochondria Health: More Than Just an Antioxidant. Molecules. 2021; 26(16):4913. https://doi.org/10.3390/molecules26164913
Chicago/Turabian StyleJong, Chian Ju, Priyanka Sandal, and Stephen W. Schaffer. 2021. "The Role of Taurine in Mitochondria Health: More Than Just an Antioxidant" Molecules 26, no. 16: 4913. https://doi.org/10.3390/molecules26164913
APA StyleJong, C. J., Sandal, P., & Schaffer, S. W. (2021). The Role of Taurine in Mitochondria Health: More Than Just an Antioxidant. Molecules, 26(16), 4913. https://doi.org/10.3390/molecules26164913