Endocrinol Metab.  2022 Aug;37(4):559-574. 10.3803/EnM.2022.1443.

A Key Metabolic Regulator of Bone and Cartilage Health

Affiliations
  • 1Medical Unit of High Specialty of Traumatology, Orthopedics and Rehabilitation “Dr. Victorio de la Fuente Narváez”, Mexican Social Security Institute, Mexico City, Mexico
  • 2National School of Medicine and Homeopathy, National Polytechnic Institute, Mexico City, Mexico
  • 3School of Agricultural Sciences and Rural Development (SASRD) Nagaland University, Medziphema, India

Abstract

Taurine, a cysteine-derived zwitterionic sulfonic acid, is a common ingredient in energy drinks and is naturally found in fish and other seafood. In humans, taurine is produced mainly in the liver, and it can also be obtained from food. In target tissues, such as the retina, heart, and skeletal muscle, it functions as an essential antioxidant, osmolyte, and antiapoptotic agent. Taurine is also involved in energy metabolism and calcium homeostasis. Taurine plays a considerable role in bone growth and development, and high-profile reports have demonstrated the importance of its metabolism for bone health. However, these reports have not been collated for more than 10 years. Therefore, this review focuses on taurine–bone interactions and covers recently discovered aspects of taurine’s effects on osteoblastogenesis, osteoclastogenesis, bone structure, and bone pathologies (e.g., osteoporosis and fracture healing), with due attention to the taurine–cartilage relationship.

Keyword

Taurine; Osteoblasts; Osteoclasts; Osteoporosis; Cartilage; Osteoarthritis

Figure

  • Fig. 1. Biosynthetic pathways of taurine in humans. Orange and blue arrows indicate synthesis pathways (I) and (II) from cysteine, respectively. Synthesis pathway (III) is indicated in green arrows [13] (created with BioRender.com). CBS, cystathionine β-synthase; CDO, cysteine dioxygenase; CSAD, cysteine sulfinate decarboxylase; HDH, hypotaurine dehydrogenase; CAD, candysteic acid decarboxylase; ADO, 2-aminoethanethiol dioxygenase.

  • Fig. 2. Human taurine tissue content, distribution, and excretion. The plasma level of taurine is the sum of the dietary intake and the endogenous synthesis by the liver, while the kidney is the primary organ for its excretion. Tissues with remarkably high taurine accumulation include the retina, heart, lung, and skeletal muscle, as well as some hematopoietic cells such as erythrocytes, platelets, and white blood cells. Data represents the amount per day. Data taken from [13] (created with BioRender.com).

  • Fig. 3. Cytoprotective roles of taurine against cell damage. Antioxidant activity: taurine facilitates the release of the nuclear factor E2-related factor (Nrf2), a redox-sensitive transcription factor, by the Kelch-like ECH-associated protein 1 (Keap1), and the translocation of Nrf2 to the nucleus and its binding to the antioxidant response element (ARE) activates the gene transcription of antioxidant enzymes. Anti-apoptotic activity: taurine’s contribution to calcium homeostasis blocks the activation of the calpain-dependent apoptotic cascade. Anti-inflammatory activity: taurine represses the degradation of the inhibitor of nuclear factor-κB (NF-κB) alpha (IκBα) and keeps NF-κB inactivated; it also prevents its translocation to the nucleus and reduces the transcription of genes encoding proinflammatory cytokines (created with BioRender.com). APAF1, apoptosis protease-activating factor-1; Bax, Bcl-2-associated X protein; BCl-2, B-cell lymphoma 2; CASP3, caspase-3; Ikβα, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha; Keap1, Kelch-like ECH-associated protein 1; MyD88, innate immune signal transduction adaptor; Nrf2, NEMO, NF-kappa-B essential modulator; sMaF, musculoaponeurotic fibrosarcoma; TLR, Toll-like receptor.

  • Fig. 4. Potential actions of taurine in bone and cartilage. Taurine promotes osteogenesis by upregulating Runt-related transcription factor 2 (RUNX2), as well as increasing alkaline phosphatase (ALP) activity and calcium deposition by osteoblasts; it also plays an antioxidant role in osteoblasts and osteocytes. In osteoclastogenesis, taurine exerts an inhibitory function. In chondrogenic differentiation, taurine promotes the synthesis of extracellular matrix components such as glycosaminoglycans and collagen, in addition to increasing the expression of SRY-box transcription factor 9 (SOX9). In bone and cartilage health, taurine promotes the formation of bone calluses in the consolidation process and promotes osseointegration. In pathological conditions such as osteoporosis, taurine supplementation increases bone mass, while in osteoarthritis, rheumatoid arthritis, and some bone tumors there are alterations in the metabolism of taurine (created with BioRender.com). TauT, taurine transporter.


Reference

1. Lambert IH, Kristensen DM, Holm JB, Mortensen OH. Physiological role of taurine: from organism to organelle. Acta Physiol (Oxf). 2015; 213:191–212.
2. Ribeiro RA, Bonfleur ML, Batista TM, Borck PC, Carneiro EM. Regulation of glucose and lipid metabolism by the pancreatic and extra-pancreatic actions of taurine. Amino Acids. 2018; 50:1511–24.
Article
3. Yu X, Hu L, Li S, Shen J, Wang D, Xu R, et al. Long non-coding RNA taurine upregulated gene 1 promotes osteosarcoma cell metastasis by mediating HIF-1α via miR-143-5p. Cell Death Dis. 2019; 10:280.
Article
4. Tiedemann F, Gmelin L. Einige neue bestandtheile der galle des ochsen. Ann Phys. 1827; 85:326–37.
Article
5. Demarcay H. Ueber die Natur der Galle. Ann Pharm. 1838; 27:270–91.
Article
6. Jacobsen JG, Smith LH. Biochemistry and physiology of taurine and taurine derivatives. Physiol Rev. 1968; 48:424–511.
Article
7. Kataoka H, Ohnishi N. Occurrence of taurine in plants. Agric Biol Chem. 2014; 50:1887–8.
Article
8. Wishart DS, Feunang YD, Marcu A, Guo AC, Liang K, Vazquez-Fresno R, et al. HMDB 4.0: the human metabolome database for 2018. Nucleic Acids Res. 2018; 46(D1):D608–17.
Article
9. Ma CC, Butler D, Milligan V, Hammann BA, Luo H, Brazdil JF, et al. Continuous process for the production of taurine from monoethanolamine. Ind Eng Chem Res. 2020; 59:13007–15.
Article
10. Laidlaw SA, Grosvenor M, Kopple JD. The taurine content of common foodstuffs. JPEN J Parenter Enteral Nutr. 1990; 14:183–8.
Article
11. Chawla D. Taurine and neonatal nutrition. Indian J Pediatr. 2018; 85:829.
Article
12. Tappaz ML. Taurine biosynthetic enzymes and taurine transporter: molecular identification and regulations. Neurochem Res. 2004; 29:83–96.
Article
13. Sarenac TM, Mikov M. Bile acid synthesis: from nature to the chemical modification and synthesis and their applications as drugs and nutrients. Front Pharmacol. 2018; 9:939.
14. Huxtable RJ. Biochemistry of Sulfur. Boston: Springer;1986. Chapter 4, Taurine and the oxidative metabolism of cysteine. p. 121–97.
15. Anderson CM, Howard A, Walters JR, Ganapathy V, Thwaites DT. Taurine uptake across the human intestinal brush-border membrane is via two transporters: H+-coupled PAT1 (SLC36A1) and Na+- and Cl(-)-dependent TauT (SLC6A6). J Physiol. 2009; 587(Pt 4):731–44.
16. Lourenço R, Camilo ME. Taurine: a conditionally essential amino acid in humans?: an overview in health and disease. Nutr Hosp. 2002; 17:262–70.
17. van Stijn MF, Vermeulen MA, Siroen MP, Wong LN, van den Tol MP, Ligthart-Melis GC, et al. Human taurine metabolism: fluxes and fractional extraction rates of the gut, liver, and kidneys. Metabolism. 2012; 61:1036–40.
Article
18. Seidel U, Huebbe P, Rimbach G. Taurine: a regulator of cellular redox homeostasis and skeletal muscle function. Mol Nutr Food Res. 2019; 63:e1800569.
Article
19. Jong CJ, Azuma J, Schaffer S. Mechanism underlying the antioxidant activity of taurine: prevention of mitochondrial oxidant production. Amino Acids. 2012; 42:2223–32.
Article
20. Shimada K, Jong CJ, Takahashi K, Schaffer SW. Role of ROS production and turnover in the antioxidant activity of taurine. Adv Exp Med Biol. 2015; 803:581–96.
Article
21. 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.
Article
22. Weiss SJ, Klein R, Slivka A, Wei M. Chlorination of taurine by human neutrophils: evidence for hypochlorous acid generation. J Clin Invest. 1982; 70:598–607.
23. Kang IS, Kim C. Taurine chloramine administered in vivo increases NRF2-regulated antioxidant enzyme expression in murine peritoneal macrophages. Adv Exp Med Biol. 2013; 775:259–67.
Article
24. Schaffer SW, Shimada K, Jong CJ, Ito T, Azuma J, Takahashi K. Effect of taurine and potential interactions with caffeine on cardiovascular function. Amino Acids. 2014; 46:1147–57.
Article
25. Bkaily G, Jazzar A, Normand A, Simon Y, Al-Khoury J, Jacques D. Taurine and cardiac disease: state of the art and perspectives. Can J Physiol Pharmacol. 2020; 98:67–73.
Article
26. Ramila KC, Jong CJ, Pastukh V, Ito T, Azuma J, Schaffer SW. Role of protein phosphorylation in excitation-contraction coupling in taurine deficient hearts. Am J Physiol Heart Circ Physiol. 2015; 308:H232–9.
Article
27. Dutka TL, Lamboley CR, Murphy RM, Lamb GD. Acute effects of taurine on sarcoplasmic reticulum Ca2+ accumulation and contractility in human type I and type II skeletal muscle fibers. J Appl Physiol (1985). 2014; 117:797–805.
Article
28. Terrill JR, Pinniger GJ, Graves JA, Grounds MD, Arthur PG. Increasing taurine intake and taurine synthesis improves skeletal muscle function in the mdx mouse model for Duchenne muscular dystrophy. J Physiol. 2016; 594:3095–110.
Article
29. Wu H, Jin Y, Wei J, Jin H, Sha D, Wu JY. Mode of action of taurine as a neuroprotector. Brain Res. 2005; 1038:123–31.
Article
30. Jong CJ, Ito T, Prentice H, Wu JY, Schaffer SW. Role of mitochondria and endoplasmic reticulum in taurine-deficiency-mediated apoptosis. Nutrients. 2017; 9:795.
Article
31. Roy A, Sil PC. Taurine protects murine hepatocytes against oxidative stress-induced apoptosis by tert-butyl hydroperoxide via PI3K/Akt and mitochondrial-dependent pathways. Food Chem. 2012; 131:1086–96.
Article
32. Sun G, Wang X, Li T, Qu S, Sun J. Taurine attenuates acrylamide-induced apoptosis via a PI3K/AKT-dependent manner. Hum Exp Toxicol. 2018; 37:1249–57.
Article
33. Li K, Wang D, Zhou X, Shao J, Li Y, Liu X, et al. Taurine protects against arsenic-induced apoptosis via PI3K/Akt pathway in primary cortical neurons. Adv Exp Med Biol. 2019; 1155:747–54.
Article
34. Prideaux M, Kitase Y, Kimble M, O’Connell TM, Bonewald LF. Taurine, an osteocyte metabolite, protects against oxidative stress-induced cell death and decreases inhibitors of the Wnt/β-catenin signaling pathway. Bone. 2020; 137:115374.
Article
35. Sun Q, Jia N, Yang J, Chen G. Nrf2 signaling pathway mediates the antioxidative effects of taurine against corticosterone-induced cell death in human SK-N-SH cells. Neurochem Res. 2018; 43:276–86.
Article
36. Iezhitsa I, Agarwal R. New solutions for old challenges in glaucoma treatment: is taurine an option to consider? Neural Regen Res. 2021; 16:967–71.
Article
37. Qiu T, Pei P, Yao X, Jiang L, Wei S, Wang Z, et al. Taurine attenuates arsenic-induced pyroptosis and nonalcoholic steatohepatitis by inhibiting the autophagic-inflammasomal pathway. Cell Death Dis. 2018; 9:946.
Article
38. Gentile CL, Nivala AM, Gonzales JC, Pfaffenbach KT, Wang D, Wei Y, et al. Experimental evidence for therapeutic potential of taurine in the treatment of nonalcoholic fatty liver disease. Am J Physiol Regul Integr Comp Physiol. 2011; 301:R1710–22.
Article
39. Wu G, Yang J, Lv H, Jing W, Zhou J, Feng Y, et al. Taurine prevents ethanol-induced apoptosis mediated by mitochondrial or death receptor pathways in liver cells. Amino Acids. 2018; 50:863–75.
Article
40. Wen C, Li F, Zhang L, Duan Y, Guo Q, Wang W, et al. Taurine is involved in energy metabolism in muscles, adipose tissue, and the liver. Mol Nutr Food Res. 2019; 63:e1800536.
Article
41. Liu Y, Li F, Zhang L, Wu J, Wang Y, Yu H. Taurine alleviates lipopolysaccharide induced liver injury by anti inflammation and antioxidants in rats. Mol Med Rep. 2017; 16:6512–7.
42. De Luca A, Pierno S, Camerino DC. Taurine: the appeal of a safe amino acid for skeletal muscle disorders. J Transl Med. 2015; 13:243.
Article
43. Kp AD, Martin A. Recent insights into the molecular regulators and mechanisms of taurine to modulate lipid metabolism: a review. Crit Rev Food Sci Nutr. 2022; Jan. 18. [Epub]. https://doi.org/10.1080/10408398.2022.2026873.
Article
44. Midwinter RG, Cheah FC, Moskovitz J, Vissers MC, Winterbourn CC. IkappaB is a sensitive target for oxidation by cell-permeable chloramines: inhibition of NF-kappaB activity by glycine chloramine through methionine oxidation. Biochem J. 2006; 396:71–8.
45. Gomez-Chavez F, Correa D, Navarrete-Meneses P, Cancino-Diaz JC, Cancino-Diaz ME, Rodriguez-Martinez S. NF- κB and its regulators during pregnancy. Front Immunol. 2021; 12:679106.
46. 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; 18:4516–22.
Article
47. Ahmadi S, Wang S, Nagpal R, Wang B, Jain S, Razazan A, et al. A human-origin probiotic cocktail ameliorates aging-related leaky gut and inflammation via modulating the microbiota/taurine/tight junction axis. JCI Insight. 2020; 5:e132055.
Article
48. Kim SH, Yum HW, Kim SH, Kim SJ, Kim K, Kim C, et al. Topically applied taurine chloramine protects against UVB-induced oxidative stress and inflammation in mouse skin. Antioxidants (Basel). 2021; 10:867.
Article
49. Ramos CO, Campos KK, Costa GP, Cangussu SD, Talvani A, Bezerra FS. Taurine treatment decreases inflammation and oxidative stress in lungs of adult mice exposed to cigarette smoke. Regul Toxicol Pharmacol. 2018; 98:50–7.
Article
50. Abd-Elhakim YM, Ghoneim MH, Ebraheim LL, Imam TS. Taurine and hesperidin rescues carbon tetrachloride-triggered testicular and kidney damage in rats via modulating oxidative stress and inflammation. Life Sci. 2020; 254:117782.
Article
51. Wang G, Ma N, He F, Kawanishi S, Kobayashi H, Oikawa S, et al. Taurine attenuates carcinogenicity in ulcerative colitis-colorectal cancer mouse model. Oxid Med Cell Longev. 2020; 2020:7935917.
Article
52. Wu L, Zhang Q, Li Y, Song W, Chen A, Liu J, et al. Collagen sponge prolongs taurine release for improved wound healing through inflammation inhibition and proliferation stimulation. Ann Transl Med. 2021; 9:1010.
Article
53. Faghfouri AH, Seyyed Shoura SM, Fathollahi P, Shadbad MA, Papi S, Ostadrahimi A, et al. Profiling inflammatory and oxidative stress biomarkers following taurine supplementation: a systematic review and dose-response meta-analysis of controlled trials. Eur J Clin Nutr. 2022; 76:647–58.
Article
54. Pasantes-Morales H. Taurine homeostasis and volume control. Adv Neurobiol. 2017; 16:33–53.
Article
55. Shennan DB. Swelling-induced taurine transport: relationship with chloride channels, anion-exchangers and other swelling-activated transport pathways. Cell Physiol Biochem. 2008; 21:15–28.
Article
56. Netti V, Pizzoni A, Perez-Dominguez M, Ford P, Pasantes-Morales H, Ramos-Mandujano G, et al. Release of taurine and glutamate contributes to cell volume regulation in human retinal Muller cells: differences in modulation by calcium. J Neurophysiol. 2018; 120:973–84.
57. Pasantes-Morales H, Quesada O, Moran J. Taurine: an osmolyte in mammalian tissues. Adv Exp Med Biol. 1998; 442:209–17.
Article
58. Morgan EF, Unnikrisnan GU, Hussein AI. Bone mechanical properties in healthy and diseased states. Annu Rev Biomed Eng. 2018; 20:119–43.
Article
59. Allen MR, Burr DB. Basic and applied bone biology. San Diego: Elsevier;2019. Chapter 1, Bone morphology and organization. p. 3–26.
60. Allen MR, Burr DB. Basic and applied bone biology. San Diego: Elsevier;2019. Chapter 5, Bone growth, modeling, and remodeling. p. 85–100.
61. Aghajanian P, Mohan S. The art of building bone: emerging role of chondrocyte-to-osteoblast transdifferentiation in endochondral ossification. Bone Res. 2018; 6:19.
Article
62. Suzuki A, Iwata J. Amino acid metabolism and autophagy in skeletal development and homeostasis. Bone. 2021; 146:115881.
Article
63. Kim SJ, Lee HW, Gupta RC. Taurine, bone growth and bone development. Curr Nutr Food Sci. 2008; 4:135–44.
Article
64. Roman-Garcia P, Quiros-Gonzalez I, Mottram L, Lieben L, Sharan K, Wangwiwatsin A, et al. Vitamin B12-dependent taurine synthesis regulates growth and bone mass. J Clin Invest. 2014; 124:2988–3002.
Article
65. Shari FH, Dawood H, Hassan JK, Aljazeari QA, Najm MA, Salahuddin A, et al. To study the effect of taurine on the effects of vital bones and regulate the level of glucose in type II diabetes. Int J Res Pharm Sci. 2019; 10:2545–51.
Article
66. Moon PD, Kim MH, Lim HS, Oh HA, Nam SY, Han NR, et al. Taurine, a major amino acid of oyster, enhances linear bone growth in a mouse model of protein malnutrition. Biofactors. 2015; 41:190–7.
Article
67. Choi MJ, Seo JN. Effect of taurine feeding on bone mineral density and bone markers in rats. Adv Exp Med Biol. 2013; 776:51–8.
Article
68. Modupe OS, Adijat BR, Salau AK. Immune response and bone marker enzyme activities of broiler birds fed graded level taurine-supplemented-diets. Ann Sci Technol. 2021; 6:16–25.
Article
69. Martiniakova M, Sarocka A, Babosova R, Galbavy D, Kapusta E, Goc Z, et al. Bone microstructure of mice after prolonged taurine treatment. Physiol Res. 2019; 68:519–23.
Article
70. Rutkovskiy A, Stenslokken KO, Vaage IJ. Osteoblast differentiation at a glance. Med Sci Monit Basic Res. 2016; 22:95–106.
Article
71. Long F. Building strong bones: molecular regulation of the osteoblast lineage. Nat Rev Mol Cell Biol. 2011; 13:27–38.
Article
72. Park M, Choi HK, An JH. Taurine activates BMP-2/Wnt3a-mediated osteoblast differentiation and mineralization via Akt and MAPK signaling. Iran J Public Health. 2019; 48:1960–70.
Article
73. Zhou C, Zhang X, Xu L, Wu T, Cui L, Xu D. Taurine promotes human mesenchymal stem cells to differentiate into osteoblast through the ERK pathway. Amino Acids. 2014; 46:1673–80.
Article
74. Lou J, Han D, Yu H, Yu G, Jin M, Kim SJ. Cytoprotective effect of taurine against hydrogen peroxide-induced oxidative stress in UMR-106 cells through the Wnt/β-catenin signaling pathway. Biomol Ther (Seoul). 2018; 26:584–90.
75. Zhang LY, Zhou YY, Chen F, Wang B, Li J, Deng YW, et al. Taurine inhibits serum deprivation-induced osteoblast apoptosis via the taurine transporter/ERK signaling pathway. Braz J Med Biol Res. 2011; 44:618–23.
Article
76. Yuan LQ, Liu W, Cui RR, Wang D, Meng JC, Xie H, et al. Taurine inhibits osteoclastogenesis through the taurine transporter. Amino Acids. 2010; 39:89–99.
Article
77. Kang YS, Kim SJ. The change of taurine transport in osteocytes by oxidative stress, hypertonicity and calcium channel blockers. Biomol Ther. 2008; 16:219–25.
Article
78. Ono T, Nakashima T. Recent advances in osteoclast biology. Histochem Cell Biol. 2018; 149:325–41.
Article
79. Park-Min KH. Mechanisms involved in normal and pathological osteoclastogenesis. Cell Mol Life Sci. 2018; 75:2519–28.
Article
80. Lee NK, Choi YG, Baik JY, Han SY, Jeong DW, Bae YS, et al. A crucial role for reactive oxygen species in RANKL-induced osteoclast differentiation. Blood. 2005; 106:852–9.
Article
81. Jang HJ, Kim SJ. Taurine exerts anti-osteoclastogenesis activity via inhibiting ROS generation, JNK phosphorylation and COX-2 expression in RAW264.7 cells. J Recept Signal Transduct Res. 2013; 33:387–91.
Article
82. Kang IS, Kim C. Taurine chloramine inhibits osteoclastic differentiation and osteoclast marker expression in RAW 264.7 cells. Adv Exp Med Biol. 2019; 1155:61–70.
Article
83. Kim C, Kang IS. Taurine chloramine, a taurine metabolite from activated neutrophils, inhibits osteoclastogenesis by suppressing NFATc1 expression. Adv Exp Med Biol. 2015; 803:99–107.
Article
84. Goldring SR. The osteocyte: key player in regulating bone turnover. RMD Open. 2015; 1(Suppl 1):e000049.
Article
85. Robling AG, Bonewald LF. The osteocyte: new insights. Annu Rev Physiol. 2020; 82:485–506.
Article
86. Einhorn TA, Gerstenfeld LC. Fracture healing: mechanisms and interventions. Nat Rev Rheumatol. 2015; 11:45–54.
Article
87. Kaya Bicer E, Yener N, Doganavsargil B, Argin M, Kapubagli A. Does taurıne improve fracture healing? An experimental study. J Ist Faculty Med. 2018; 81:91–8.
88. Hirata H, Ueda S, Ichiseki T, Shimasaki M, Ueda Y, Kaneuji A, et al. Taurine inhibits glucocorticoid-induced bone mitochondrial injury, preventing osteonecrosis in rabbits and cultured osteocytes. Int J Mol Sci. 2020; 21:6892.
Article
89. Yang SS, Oh JM, Chun S, Kim BS, Kim CS, Lee J. Tauroursodeoxycholic acid induces angiogenic activity in endothelial cells and accelerates bone regeneration. Bone. 2020; 130:115073.
Article
90. Samadian H, Farzamfar S, Vaez A, Ehterami A, Bit A, Alam M, et al. A tailored polylactic acid/polycaprolactone biodegradable and bioactive 3D porous scaffold containing gelatin nanofibers and taurine for bone regeneration. Sci Rep. 2020; 10:13366.
Article
91. Kanis JA. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis: synopsis of a WHO report. WHO Study Group. Osteoporos Int. 1994; 4:368–81.
92. Hershman SH, Razi AE. Vertebral compression fractures in osteoporotic and pathologic bone. Cham: Springer;2020. Chapter 3, The economic burden of osteoporosis. p. 21–9.
Article
93. Pontes TA, Barbosa AD, Silva RD, Melo-Junior MR, Silva RO. Osteopenia-osteoporosis discrimination in postmenopausal women by 1H NMR-based metabonomics. PLoS One. 2019; 14:e0217348.
Article
94. Zhao Q, Shen H, Su KJ, Zhang JG, Tian Q, Zhao LJ, et al. Metabolomic profiles associated with bone mineral density in US Caucasian women. Nutr Metab (Lond). 2018; 15:57.
Article
95. Miyamoto T, Hirayama A, Sato Y, Koboyashi T, Katsuyama E, Kanagawa H, et al. Metabolomics-based profiles predictive of low bone mass in menopausal women. Bone Rep. 2018; 9:11–8.
Article
96. Cheong SH, Chang KJ. The preventive effect of fermented milk supplement containing tomato (lycopersion esculentum) and taurine on bone loss in ovariectomized rats. Adv Exp Med Biol. 2009; 643:333–40.
Article
97. Yu L, Qi H, An G, Bao J, Ma B, Zhu J, et al. Association between metabolic profiles in urine and bone mineral density of pre- and postmenopausal Chinese women. Menopause. 2019; 26:94–102.
Article
98. Berry TM, Moustafa AA. Osteoporosis and the effect of dysregulation of the transsulfuration pathway via taurine on intracellular calcium homeostasis, vitamin D absorption and vitamin K absorption. Clin Nutr ESPEN. 2021; 43:191–6.
Article
99. Ye M, Zhang C, Jia W, Shen Q, Qin X, Zhang H, et al. Metabolomics strategy reveals the osteogenic mechanism of yak (Bos grunniens) bone collagen peptides on ovariectomy-induced osteoporosis in rats. Food Funct. 2020; 11:1498–512.
100. Chen SY, Yu HT, Kao JP, Yang CC, Chiang SS, Mishchuk DO, et al. An NMR metabolomic study on the effect of alendronate in ovariectomized mice. PLoS One. 2014; 9:e106559.
Article
101. Liu S, Yuan X, Ma C, Zhao J, Xiong Z. 1H-NMR-based urinary metabolomic analysis for the preventive effects of gushudan on glucocorticoid-induced osteoporosis rats. Anal Biochem. 2020; 610:113992.
Article
102. Ahmed HH, Hamza AH. Potential role of arginine, glutamine and taurine in ameliorating osteoporotic biomarkers in ovariectomized rats. Rep Opin. 2009; 1:24–35.
103. Paulos J, Poitout DG. Bone tumors. London: Springer;2021. Chapter 2, Osteomas. p. 19–20.
Article
104. Lv D, Zou Y, Zeng Z, Yao H, Ding S, Bian Y, et al. Comprehensive metabolomic profiling of osteosarcoma based on UHPLC-HRMS. Metabolomics. 2020; 16:120.
Article
105. Lopez-Garrido L, Banuelos-Hernandez AE, Perez-Hernandez E, Tecualt-Gomez R, Quiroz-Williams J, Ariza-Castolo A, et al. Metabolic profiling of serum in patients with cartilage tumours using 1 H-NMR spectroscopy: a pilot study. Magn Reson Chem. 2020; 58:65–76.
Article
106. Duarte IF, Lamego I, Marques J, Marques MP, Blaise BJ, Gil AM. Nuclear magnetic resonance (NMR) study of the effect of cisplatin on the metabolic profile of MG-63 osteosarcoma cells. J Proteome Res. 2010; 9:5877–86.
Article
107. Marley K, Helfand SC, Edris WA, Mata JE, Gitelman AI, Medlock J, et al. The effects of taurolidine alone and in combination with doxorubicin or carboplatin in canine osteosarcoma in vitro. BMC Vet Res. 2013; 9:15.
Article
108. Pilz M, Holinka J, Vavken P, Marian B, Krepler P. Taurine chloramine induces apoptosis in human osteosarcoma cell lines. J Orthop Res. 2012; 30:2046–51.
Article
109. Green JD, Tollemar V, Dougherty M, Yan Z, Yin L, Ye J, et al. Multifaceted signaling regulators of chondrogenesis: implications in cartilage regeneration and tissue engineering. Genes Dis. 2015; 2:307–27.
Article
110. Yao X, Huang H, Li Z, Liu X, Fan W, Wang X, et al. Taurine promotes the cartilaginous differentiation of human umbilical cord-derived mesenchymal stem cells in vitro. Neurochem Res. 2017; 42:2344–53.
Article
111. Liu Q, Lu Z, Wu H, Zheng L. Chondroprotective effects of taurine in primary cultures of human articular chondrocytes. Tohoku J Exp Med. 2015; 235:201–13.
Article
112. Karjalainen HM, Qu C, Leskela SS, Rilla K, Lammi MJ. Chondrocytic cells express the taurine transporter on their plasma membrane and regulate its expression under anisotonic conditions. Amino Acids. 2015; 47:561–70.
Article
113. Peat G, Thomas MJ. Osteoarthritis year in review 2020: epidemiology & therapy. Osteoarthritis Cartilage. 2021; 29:180–9.
Article
114. Mobasheri A, Rayman MP, Gualillo O, Sellam J, van der Kraan P, Fearon U. The role of metabolism in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2017; 13:302–11.
Article
115. Chen R, Han S, Liu X, Wang K, Zhou Y, Yang C, et al. Perturbations in amino acids and metabolic pathways in osteoarthritis patients determined by targeted metabolomics analysis. J Chromatogr B Analyt Technol Biomed Life Sci. 2018; 1085:54–62.
Article
116. Yang G, Zhang H, Chen T, Zhu W, Ding S, Xu K, et al. Metabolic analysis of osteoarthritis subchondral bone based on UPLC/Q-TOF-MS. Anal Bioanal Chem. 2016; 408:4275–86.
Article
117. Anderson JR, Chokesuwattanaskul S, Phelan MM, Welting TJM, Lian LY, Peffers MJ, et al. 1H NMR metabolomics identifies underlying inflammatory pathology in osteoarthritis and rheumatoid arthritis synovial joints. J Proteome Res. 2018; 17:3780–90.
118. Xu Z, Chen T, Luo J, Ding S, Gao S, Zhang J. Cartilaginous metabolomic study reveals potential mechanisms of osteophyte formation in osteoarthritis. J Proteome Res. 2017; 16:1425–35.
Article
119. Saraswati AAEW, Sunardi D, Lubis AMT, Heru F, Mudjihartini N. Taurine intakes increase superoxide dismutase activity in knee osteoarthritis. IOP Conf Ser Earth Environ Sci. 2019; 217:012054.
Article
120. Bian Y, Zhang M, Wang K. Taurine protects against knee osteoarthritis development in experimental rat models. Knee. 2018; 25:374–80.
Article
121. Bian Y, Wang H, Sun S. Taurine alleviates endoplasmic reticulum stress in the chondrocytes from patients with osteoarthritis. Redox Rep. 2018; 23:118–24.
Article
122. Almutairi K, Nossent J, Preen D, Keen H, Inderjeeth C. The global prevalence of rheumatoid arthritis: a meta-analysis based on a systematic review. Rheumatol Int. 2021; 41:863–77.
Article
123. Liu Y, Wei M, Yue K, Wang R, Ma Y, Men L, et al. Non-target metabonomic method provided new insights on the therapeutical mechanism of Gancao Fuzi decoction on rheumatoid arthritis rats. J Chromatogr B Analyt Technol Biomed Life Sci. 2019; 1105:93–103.
Article
124. Cheng B, Zheng H, Wu F, Wu J, Liu X, Tang C, et al. Metabolomics analysis of Danggui Sini decoction on treatment of collagen-induced arthritis in rats. J Chromatogr B Analyt Technol Biomed Life Sci. 2017; 1061-1062:282–91.
Article
125. Yun X, Dong S, Hu Q, Dai Y, Xia Y. 1H NMR-based metabolomics approach to investigate the urine samples of collagen-induced arthritis rats and the intervention of tetrandrine. J Pharm Biomed Anal. 2018; 154:302–11.
Article
126. Sweeney SR, Kavanaugh A, Lodi A, Wang B, Boyle D, Tiziani S, et al. Metabolomic profiling predicts outcome of rituximab therapy in rheumatoid arthritis. RMD Open. 2016; 2:e000289.
Article
127. Wang Z, Chen Z, Yang S, Wang Y, Yu L, Zhang B, et al. (1) H NMR-based metabolomic analysis for identifying serum biomarkers to evaluate methotrexate treatment in patients with early rheumatoid arthritis. Exp Ther Med. 2012; 4:165–71.
128. Malek Mahdavi A, Javadivala Z. A systematic review of preclinical studies on the efficacy of taurine for the treatment of rheumatoid arthritis. Amino Acids. 2021; 53:783–800.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr