Hostname: page-component-669899f699-cf6xr Total loading time: 0 Render date: 2025-04-24T20:37:41.168Z Has data issue: false hasContentIssue false

The influence of zinc levels on osteoarthritis: A comprehensive review

Published online by Cambridge University Press:  23 September 2024

İbrahim Tekeoğlu
Affiliation:
Sakarya University Faculty of Medicine, Department of Rheumatology, Sakarya University Training and Research Hospital, Sakarya, Türkiye
Muhammed Zahid Şahin*
Affiliation:
Sakarya University Faculty of Medicine, Department of Physical Medicine and Rehabilitation, Sakarya University Training and Research Hospital, Sakarya, Türkiye
Ayhan Kamanlı
Affiliation:
Sakarya University Faculty of Medicine, Department of Rheumatology, Sakarya University Training and Research Hospital, Sakarya, Türkiye
Kemal Nas
Affiliation:
Sakarya University Faculty of Medicine, Department of Rheumatology, Sakarya University Training and Research Hospital, Sakarya, Türkiye
*
*Corresponding author: Muhammed Zahid Şahin, email: [email protected]

Abstract

Osteoarthritis (OA), a disease with a multifactorial aetiology and an enigmatic root cause, affects the quality of life of many elderly patients. Even though there are certain medications utilised to reduce the symptomatic effects, a reliable treatment method to reverse the disease is yet to be discovered. Zinc is a cofactor of over 3000 proteins and is the only metal found in all six classes of enzymes. We explored zinc’s effect on the immune system and the bones as OA affects both. We also discussed zinc-dependent enzymes, highlighting their significant role in the disease’s pathogenesis. It is important to note that both excessive and deficient zinc levels can negatively affect bone health and immune function, thereby exacerbating OA. The purpose of this review is to offer a better understanding of zinc’s impact on OA pathogenesis and to provide clarity regarding its beneficial and detrimental outcomes. We searched thoroughly systematic reviews, meta-analysis, review articles, research articles and randomised controlled trials to ensure a comprehensive review. In brief, using zinc supplementation in the treatment of OA may act as a doubled-edged sword, offering potential benefits but also posing risks.

Type
Review Article
Copyright
© The Author(s), 2024. Published by Cambridge University Press on behalf of The Nutrition Society

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Article purchase

Temporarily unavailable

References

Robinson, WH, Lepus, CM, Wang, Q, et al. (2016) Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nat Rev Rheumatol 12, 580592. https://doi.org/10.1038/nrrheum.2016.136 CrossRefGoogle ScholarPubMed
Bliddal, H, Leeds, AR, Christensen, R (2014) Osteoarthritis, obesity and weight loss: evidence, hypotheses and horizons: a scoping review. Obes Rev Off J Int Assoc Study Obes 15, 578586. https://doi.org/10.1111/obr.12173 CrossRefGoogle ScholarPubMed
Allen, KD, Thoma, LM, Golightly, YM (2022) Epidemiology of osteoarthritis. Osteoarthr Cartil 30, 184195. https://doi.org/10.1016/j.joca.2021.04.020 CrossRefGoogle ScholarPubMed
Loeser, RF, Collins, JA, Diekman, BO (2016) Ageing and the pathogenesis of osteoarthritis. Nat Rev Rheumatol 12, 412420. https://doi.org/10.1038/nrrheum.2016.65 CrossRefGoogle ScholarPubMed
Zhou, J, Liu, C, Sun, Y, et al. (2021) Genetically predicted circulating levels of copper and zinc are associated with osteoarthritis but not with rheumatoid arthritis. Osteoarthr Cartil 29, 10291035. https://doi.org/10.1016/j.joca.2021.02.564 CrossRefGoogle Scholar
Mobasheri, A & Batt, M (2016) An update on the pathophysiology of osteoarthritis. Ann Phys Rehabil Med 59, 333339. https://doi.org/10.1016/j.rehab.2016.07.004 CrossRefGoogle ScholarPubMed
Pagano, G, Talamanca, AA, Castello, G, et al. (2014) Oxidative stress and mitochondrial dysfunction across broad-ranging pathologies: toward mitochondria-targeted clinical strategies. Oxid Med Cell Longevity, 2014, 541230. https://doi.org/10.1155/2014/541230 CrossRefGoogle ScholarPubMed
Loeser, RF (2009) Aging and osteoarthritis: the role of chondrocyte senescence and aging changes in the cartilage matrix. Osteoarthr Cartil 17, 971979. https://doi.org/10.1016/j.joca.2009.03.002 CrossRefGoogle ScholarPubMed
Kobayashi, M, Squires, GR, Mousa, A, et al. (2005) Role of interleukin-1 and tumor necrosis factor alpha in matrix degradation of human osteoarthritic cartilage. Arthritis Rheum 52, 128135. https://doi.org/10.1002/art.20776 CrossRefGoogle ScholarPubMed
Tetlow, LC, Adlam, DJ, Woolley, DE (2001) Matrix metalloproteinase and proinflammatory cytokine production by chondrocytes of human osteoarthritic cartilage: associations with degenerative changes. Arthritis Rheum 44, 585594. https://doi.org/10.1002/1529-0131(200103)44:3<585::AID-ANR107>3.0.CO;2-C 3.0.CO;2-C>CrossRefGoogle ScholarPubMed
Huang, LW, Huang, TC, Hu, YC, et al. (2020) Zinc protects chondrocytes from monosodium iodoacetate-induced damage by enhancing ATP and mitophagy. Biochem Biophys Res Commun 521, 5056. https://doi.org/10.1016/j.bbrc.2019.10.066 CrossRefGoogle ScholarPubMed
Reyes, C, Leyland, KM, Peat, G, et al. (2016) Association between overweight and obesity and risk of clinically diagnosed knee, hip, and hand osteoarthritis: a population-based cohort study. Arthritis Rheum 68, 18691875. https://doi.org/10.1002/art.39707 CrossRefGoogle ScholarPubMed
Conde, J, Scotece, M, Abella, V, et al. (2015) Identification of novel adipokines in the joint. Differential expression in healthy and osteoarthritis tissues. PLoS One 10, e0123601. https://doi.org/10.1371/journal.pone.0123601 CrossRefGoogle ScholarPubMed
Belluzzi, E, El Hadi, H, Granzotto, M, et al. (2017) Systemic and local adipose tissue in knee osteoarthritis. J Cell Physiol 232, 19711978. https://doi.org/10.1002/jcp.25716 CrossRefGoogle ScholarPubMed
Samvelyan, HJ, Hughes, D, Stevens, C, et al. (2021) Models of osteoarthritis: relevance and new insights. Calcif Tissue Int 109, 243256. https://doi.org/10.1007/s00223-020-00670-x CrossRefGoogle ScholarPubMed
Kuyinu, EL, Narayanan, G, Nair, LS, et al. (2016) Animal models of osteoarthritis: classification, update, and measurement of outcomes. J Orthop Surg Res 11, 19. https://doi.org/10.1186/s13018-016-0346-5 CrossRefGoogle ScholarPubMed
Chiu, PR, Hu, YC, Huang, TC, et al. (2016) Vitamin C protects chondrocytes against monosodium iodoacetate-induced osteoarthritis by multiple pathways. Int J Mol Sci 18, 38. https://doi.org/10.3390/ijms18010038 CrossRefGoogle ScholarPubMed
Huang, TC, Chang, WT, Hu, YC, et al. (2018) Zinc protects articular chondrocytes through changes in Nrf2-mediated antioxidants, cytokines and matrix metalloproteinases. Nutrients 10, 471. https://doi.org/10.3390/nu10040471 CrossRefGoogle ScholarPubMed
Kosik-Bogacka, DI, Lanocha-Arendarczyk, N, Kot, K, et al. (2018) Calcium, magnesium, zinc and lead concentrations in the structures forming knee joint in patients with osteoarthritis‬. J Trace Elem Med Biol 50, 409414. https://doi.org/10.1016/j.jtemb.2018.08.007 CrossRefGoogle Scholar
Zago, MP & Oteiza, PI (2001) The antioxidant properties of zinc: interactions with iron and antioxidants. Free Radic Biol Med 31, 266274. https://doi.org/10.1016/s0891-5849(01)00583-4 CrossRefGoogle ScholarPubMed
Yang, WM, Lv, JF, Wang, YY, et al. (2023) The daily intake levels of copper, selenium, and zinc are associated with osteoarthritis but not with rheumatoid arthritis in a cross-sectional study. Biol Trace Elem Res 201, 56625670. Advance online publication. https://doi.org/10.1007/s12011-023-03636-w CrossRefGoogle Scholar
McCall, KA, Huang, C, Fierke, CA (2000) Function and mechanism of zinc metalloenzymes. J Nutr 130(5S Suppl), 1437S1446S. https://doi.org/10.1093/jn/130.5.1437S CrossRefGoogle ScholarPubMed
Gálvez-Peralta, M, Wang, Z, Bao, S, et al. (2014) Tissue-specific induction of mouse ZIP8 and ZIP14 divalent cation/bicarbonate symporters by, and cytokine response to, inflammatory signals. Int J Toxicol 33, 246258. https://doi.org/10.1177/1091581814529310 CrossRefGoogle ScholarPubMed
Kim, JH, Jeon, J, Shin, M, et al. (2014) Regulation of the catabolic cascade in osteoarthritis by the zinc-ZIP8-MTF1 axis. Cell 156, 730743. https://doi.org/10.1016/j.cell.2014.01.007 CrossRefGoogle ScholarPubMed
Lee, M, Won, Y, Shin, Y, et al. (2016) Reciprocal activation of hypoxia-inducible factor (HIF)-2α and the zinc-ZIP8-MTF1 axis amplifies catabolic signaling in osteoarthritis. Osteoarthr Cartil 24, 134145. https://doi.org/10.1016/j.joca.2015.07.016 CrossRefGoogle ScholarPubMed
Mahmoud, RK, El-Ansary, AK, El-Eishi, HH, et al. (2005) Matrix metalloproteinases MMP-3 and MMP-1 levels in sera and synovial fluids in patients with rheumatoid arthritis and osteoarthritis. Ital J Biochem 54, 248257.Google ScholarPubMed
Cabrera, ÁJ (2015) Zinc, aging, and immunosenescence: an overview. Pathobiol Aging Age Relat Dis 5, 25592. https://doi.org/10.3402/pba.v5.25592 CrossRefGoogle ScholarPubMed
Chesters, JK & Will, M (1981) Zinc transport proteins in plasma. Br J Nutr 46, 111118. https://doi.org/10.1079/bjn19810014 CrossRefGoogle ScholarPubMed
Zaichick, V & Zaichick, S (2009) Instrumental neutron activation analysis of trace element contents in the rib bone of healthy men. J Radioanal Nucl Chem 281, 4752.CrossRefGoogle Scholar
MacDonald, RS (2000) The role of zinc in growth and cell proliferation. J Nutr 130(5S Suppl), 1500S1508S. https://doi.org/10.1093/jn/130.5.1500S CrossRefGoogle ScholarPubMed
Aceituno-Valenzuela, U, Micol-Ponce, R, Ponce, MR (2020) Genome-wide analysis of CCHC-type zinc finger (ZCCHC) proteins in yeast, Arabidopsis, and humans. Cell Mol Life Sci 77, 39914014. https://doi.org/10.1007/s00018-020-03518-7 CrossRefGoogle ScholarPubMed
Marreiro, DD, Cruz, KJ, Morais, JB, et al. (2017) Zinc and oxidative stress: current mechanisms. Antioxidants 6, 24. https://doi.org/10.3390/antiox6020024 CrossRefGoogle ScholarPubMed
King, LE, Frentzel, JW, Mann, JJ, et al. (2005) Chronic zinc deficiency in mice disrupted T cell lymphopoiesis and erythropoiesis while B cell lymphopoiesis and myelopoiesis were maintained. J Am Coll Nutr 24, 494502. https://doi.org/10.1080/07315724.2005.10719495 CrossRefGoogle Scholar
Kambe, T, Tsuji, T, Hashimoto, A, et al. (2015) The physiological, biochemical, and molecular roles of zinc transporters in zinc homeostasis and metabolism. Physiol Rev 95, 749784. https://doi.org/10.1152/physrev.00035.2014 CrossRefGoogle ScholarPubMed
Khader, A & Arinzeh, TL (2020) Biodegradable zinc oxide composite scaffolds promote osteochondral differentiation of mesenchymal stem cells. Biotechnol Bioeng 117, 194209. https://doi.org/10.1002/bit.27173 CrossRefGoogle ScholarPubMed
Rodríguez, JP & Rosselot, G (2001) Effects of zinc on cell proliferation and proteoglycan characteristics of epiphyseal chondrocytes. J Cell Biochem 82, 501511. https://doi.org/10.1002/jcb.1178 CrossRefGoogle ScholarPubMed
Klug, A (2010) The discovery of zinc fingers and their applications in gene regulation and genome manipulation. Annu Rev Biochem 79, 213231. https://doi.org/10.1146/annurev-biochem-010909-095056 CrossRefGoogle ScholarPubMed
Grzeszczak, K, Kwiatkowski, S, Kosik-Bogacka, D (2020) The role of Fe, Zn, and Cu in pregnancy. Biomolecules 10, 1176. https://doi.org/10.3390/biom10081176 CrossRefGoogle ScholarPubMed
Ciosek, Ż, Kot, K, Rotter, I (2023) Iron, zinc, copper, cadmium, mercury, and bone tissue. Int J Environ Res Public Health 20, 2197. https://doi.org/10.3390/ijerph20032197 CrossRefGoogle ScholarPubMed
Charles, CH, Cronin, MJ, Conforti, NJ, et al. (2001) Anticalculus efficacy of an antiseptic mouthrinse containing zinc chloride. J Am Dent Assoc 132, 9498. https://doi.org/10.14219/jada.archive.2001.0033 CrossRefGoogle ScholarPubMed
Eberle, J, Schmidmayer, S, Erben, RG, et al. (1999) Skeletal effects of zinc deficiency in growing rats. J Trace Elem Med Biol 13, 2126. https://doi.org/10.1016/S0946-672X(99)80019-4 CrossRefGoogle ScholarPubMed
Rocha, ÉD, de Brito, NJ, Dantas, MM, et al. (2015) Effect of zinc supplementation on GH, IGF1, IGFBP3, OCN, and ALP in non-zinc-deficient children. J Am Coll Nutr 34, 290299. https://doi.org/10.1080/07315724.2014.929511 CrossRefGoogle ScholarPubMed
Prasad, AS, Halsted, JA, Nadimi, M (1961) Syndrome of iron deficiency anemia, hepatosplenomegaly, hypogonadism, dwarfism and geophagia. Am J Med 31, 532546. https://doi.org/10.1016/0002-9343(61)90137-1 CrossRefGoogle ScholarPubMed
Kawai, S, Yamauchi, M, Wakisaka, S, et al. (2007) Zinc-finger transcription factor odd-skipped related 2 is one of the regulators in osteoblast proliferation and bone formation. J Bone Miner Res 22, 13621372. https://doi.org/10.1359/jbmr.070602 CrossRefGoogle ScholarPubMed
Peretz, A, Papadopoulos, T, Willems, D, et al. (2001) Zinc supplementation increases bone alkaline phosphatase in healthy men. J Trace Elem Med Biol 15, 175178. https://doi.org/10.1016/S0946-672X(01)80063-8 CrossRefGoogle ScholarPubMed
Rossi, L, Migliaccio, S, Corsi, A, et al. (2001) Reduced growth and skeletal changes in zinc-deficient growing rats are due to impaired growth plate activity and inanition. J Nutr 131, 11421146. https://doi.org/10.1093/jn/131.4.1142 CrossRefGoogle ScholarPubMed
Hie, M, Iitsuka, N, Otsuka, T, et al. (2011) Zinc deficiency decreases osteoblasts and osteoclasts associated with the reduced expression of Runx2 and RANK. Bone 49, 11521159. https://doi.org/10.1016/j.bone.2011.08.019 CrossRefGoogle ScholarPubMed
Cho, YE & Kwun, IS (2018) Zinc upregulates bone-specific transcription factor Runx2 expression via BMP-2 signaling and Smad-1 phosphorylation in osteoblasts. J Nutr Health 51, 2330.CrossRefGoogle Scholar
Guo, B, Yang, M, Liang, D, et al. (2012) Cell apoptosis induced by zinc deficiency in osteoblastic MC3T3-E1 cells via a mitochondrial-mediated pathway. Mol Cell Biochem 361, 209216. https://doi.org/10.1007/s11010-011-1105-x CrossRefGoogle Scholar
O’Connor, JP, Kanjilal, D, Teitelbaum, M, et al. (2020) Zinc as a therapeutic agent in bone regeneration. Materials 13, 2211. https://doi.org/10.3390/ma13102211 CrossRefGoogle ScholarPubMed
Hie, M & Tsukamoto, I (2011) Administration of zinc inhibits osteoclastogenesis through the suppression of RANK expression in bone. Eur J Pharmacol 668, 140146. https://doi.org/10.1016/j.ejphar.2011.07.003 CrossRefGoogle ScholarPubMed
Yamaguchi, M & Uchiyama, S (2004) Receptor activator of NF-kappaB ligand-stimulated osteoclastogenesis in mouse marrow culture is suppressed by zinc in vitro . Int J Mol Med 14, 8185.Google ScholarPubMed
Cerovic, A, Miletic, I, Sobajic, S, et al. (2007) Effects of zinc on the mineralization of bone nodules from human osteoblast-like cells. Biol Trace Elem Res 116, 6171. https://doi.org/10.1007/BF02685919 CrossRefGoogle ScholarPubMed
Gammoh, NZ & Rink, L (2017) Zinc in infection and inflammation. Nutrients 9, 624. https://doi.org/10.3390/nu9060624 CrossRefGoogle ScholarPubMed
Maywald, M & Rink, L (2015) Zinc homeostasis and immunosenescence. Trace Elem Med Biol 29, 2430. https://doi.org/10.1016/j.jtemb.2014.06.003 CrossRefGoogle ScholarPubMed
Prasad, AS, Beck, FW, Bao, B, et al. (2007) Zinc supplementation decreases incidence of infections in the elderly: effect of zinc on generation of cytokines and oxidative stress. Am J Clin Nutr 85, 837844. https://doi.org/10.1093/ajcn/85.3.837 CrossRefGoogle ScholarPubMed
Bhutta, ZA, Black, RE, Brown, KH, et al. (1999) Prevention of diarrhea and pneumonia by zinc supplementation in children in developing countries: pooled analysis of randomized controlled trials. J Pediatr 135, 689697. https://doi.org/10.1016/s0022-3476(99)70086-7 CrossRefGoogle ScholarPubMed
von Pein, JB, Stocks, CJ, Schembri, MA, et al. (2021) An alloy of zinc and innate immunity: galvanising host defence against infection. Cell Microbiol 23, e13268. https://doi.org/10.1111/cmi.13268 CrossRefGoogle ScholarPubMed
Kapetanovic, R, Bokil, NJ, Achard, ME, et al. (2016) Salmonella employs multiple mechanisms to subvert the TLR-inducible zinc-mediated antimicrobial response of human macrophages. FASEB J 30, 19011912. https://doi.org/10.1096/fj.201500061 CrossRefGoogle ScholarPubMed
Eide, DJ (2006) Zinc transporters and the cellular trafficking of zinc. BBA 1763, 711722. https://doi.org/10.1016/j.bbamcr.2006.03.005 Google ScholarPubMed
Stocks, CJ, Phan, MD, Achard, MES, et al. (2019) Uropathogenic Escherichia coli employs both evasion and resistance to subvert innate immune-mediated zinc toxicity for dissemination. PNAS 116, 63416350. https://doi.org/10.1073/pnas.1820870116 CrossRefGoogle ScholarPubMed
Ong, CY, Berking, O, Walker, MJ, et al. (2018) New insights into the role of zinc acquisition and zinc tolerance in group A streptococcal infection. Infect Immun 86, e0004818. https://doi.org/10.1128/IAI.00048-18 CrossRefGoogle ScholarPubMed
Botella, H, Peyron, P, Levillain, F, et al. (2011) Mycobacterial p(1)-type ATPases mediate resistance to zinc poisoning in human macrophages. Cell Host Microbe 10, 248259. https://doi.org/10.1016/j.chom.2011.08.006 CrossRefGoogle ScholarPubMed
Stafford, SL, Bokil, NJ, Achard, ME, et al. (2013) Metal ions in macrophage antimicrobial pathways: emerging roles for zinc and copper. Biosci Rep 33, e00049. https://doi.org/10.1042/BSR20130014 CrossRefGoogle ScholarPubMed
Eijkelkamp, BA, Morey, JR, Ween, MP, et al. (2014) Extracellular zinc competitively inhibits manganese uptake and compromises oxidative stress management in Streptococcus pneumoniae. PLoS One 9, e89427. https://doi.org/10.1371/journal.pone.0089427 CrossRefGoogle ScholarPubMed
Rosenkranz, E, Metz, CH, Maywald, M, et al. (2016) Zinc supplementation induces regulatory T cells by inhibition of Sirt-1 deacetylase in mixed lymphocyte cultures. Mol Nutr Food Res 60, 661671. https://doi.org/10.1002/mnfr.201500524 CrossRefGoogle ScholarPubMed
Hanidziar, D & Koulmanda, M (2010) Inflammation and the balance of Treg and Th17 cells in transplant rejection and tolerance. Curr Opin Organ Transplant 15, 411415. https://doi.org/10.1097/MOT.0b013e32833b7929 CrossRefGoogle ScholarPubMed
Rink, L & Haase, H (2007) Zinc homeostasis and immunity. Trends Immunol 28, 14. https://doi.org/10.1016/j.it.2006.11.005 CrossRefGoogle ScholarPubMed
Weston, WL, Huff, JC, Humbert, JR, et al. (1977) Zinc correction of defective chemotaxis in acrodermatitis enteropathica. Archiv Dermatolo 113, 422425.CrossRefGoogle ScholarPubMed
Maywald, M, Wessels, I, Rink, L (2017) Zinc signals and immunity. Int J Mol Sci 18, 2222. https://doi.org/10.3390/ijms18102222 CrossRefGoogle ScholarPubMed
Wessels, I & Cousins, RJ (2015) Zinc dyshomeostasis during polymicrobial sepsis in mice involves zinc transporter Zip14 and can be overcome by zinc supplementation. Am J Physiol Gastrointest Liver Physiol 309, G768G778. https://doi.org/10.1152/ajpgi.00179.2015 CrossRefGoogle ScholarPubMed
Mocchegiani, E & Malavolta, M (2007) Zinc dyshomeostasis, ageing and neurodegeneration: implications of A2M and inflammatory gene polymorphisms. J Alzheimer’s Dis 12, 101109. https://doi.org/10.3233/jad-2007-12110 CrossRefGoogle ScholarPubMed
Maret, W (2011) Metals on the move: zinc ions in cellular regulation and in the coordination dynamics of zinc proteins. Biometals 24, 411418. https://doi.org/10.1007/s10534-010-9406-1 CrossRefGoogle ScholarPubMed
Dardenne, M, Savino, W, Wade, S, et al. (1984) In vivo and in vitro studies of thymulin in marginally zinc-deficient mice. Eur J Immunol 14, 454458. https://doi.org/10.1002/eji.1830140513 CrossRefGoogle ScholarPubMed
Golden, MH, Jackson, AA, Golden, BE (1977) Effect of zinc on thymus of recently malnourished children. Lancet 2, 10571059. https://doi.org/10.1016/s0140-6736(77)91888-8 CrossRefGoogle ScholarPubMed
Fraker, PJ (2005) Roles for cell death in zinc deficiency. J Nutr 135, 359362. https://doi.org/10.1093/jn/135.3.359 CrossRefGoogle ScholarPubMed
Macfarlane, E, Seibel, MJ, Zhou, H (2020) Arthritis and the role of endogenous glucocorticoids. Bone Res 8, 33. https://doi.org/10.1038/s41413-020-00112-2 CrossRefGoogle ScholarPubMed
Chai, F, Truong-Tran, AQ, Evdokiou, A, et al. (2000) Intracellular zinc depletion induces caspase activation and p21 Waf1/Cip1 cleavage in human epithelial cell lines. J Infect Dis 182(Suppl 1), S85S92. https://doi.org/10.1086/315914 CrossRefGoogle ScholarPubMed
Safieh-Garabedian, B, Ahmed, K, Khamashta, MA, et al. (1993) Thymulin modulates cytokine release by peripheral blood mononuclear cells: a comparison between healthy volunteers and patients with systemic lupus erythematosus. Int Arch Allergy Immunol 101, 126131. https://doi.org/10.1159/000236509 CrossRefGoogle ScholarPubMed
Prasad, AS (2000) Effects of zinc deficiency on Th1 and Th2 cytokine shifts. J Infect Dis 182(Suppl 1), S62S68. https://doi.org/10.1086/315916 CrossRefGoogle ScholarPubMed
Bao, B, Prasad, AS, Beck, FW, et al. (2003) Zinc modulates mRNA levels of cytokines. Am J Physiol Endocrinol Metabol 285, E1095E1102. https://doi.org/10.1152/ajpendo.00545.2002 CrossRefGoogle ScholarPubMed
Gilbert, SJ, Blain, EJ, Mason, DJ (2022) Interferon-gamma modulates articular chondrocyte and osteoblast metabolism through protein kinase R-independent and dependent mechanisms. Biochem Biophys Rep 32, 101323. https://doi.org/10.1016/j.bbrep.2022.101323 Google ScholarPubMed
Prasad, AS, Bao, B, Beck, FW, et al. (2001) Zinc activates NF-kappaB in HUT-78 cells. J Lab Clin Med 138, 250256. https://doi.org/10.1067/mlc.2001.118108 CrossRefGoogle ScholarPubMed
Beck, FW, Prasad, AS, Kaplan, J, et al. (1997) Changes in cytokine production and T cell subpopulations in experimentally induced zinc-deficient humans. Am J Physiol 272(6 Pt 1), E1002E1007. https://doi.org/10.1152/ajpendo.1997.272.6.E1002 Google Scholar
Keen, CL & Gershwin, ME (1990) Zinc deficiency and immune function. Annu Rev Nutr 10, 415431. https://doi.org/10.1146/annurev.nu.10.070190.002215 CrossRefGoogle ScholarPubMed
Shi, HN, Scott, ME, Stevenson, MM, et al. (1998) Energy restriction and zinc deficiency impair the functions of murine T cells and antigen-presenting cells during gastrointestinal nematode infection. J Nutr 128, 2027. https://doi.org/10.1093/jn/128.1.20 CrossRefGoogle ScholarPubMed
Prasad, AS (2014) Zinc is an antioxidant and anti-Inflammatory agent: its role in human health. Front Nutr 1, 14. https://doi.org/10.3389/fnut.2014.00014 CrossRefGoogle Scholar
Langrish, CL, McKenzie, BS, Wilson, NJ, et al. (2004) IL-12 and IL-23: master regulators of innate and adaptive immunity. Immunol Rev 202, 96105. https://doi.org/10.1111/j.0105-2896.2004.00214.x CrossRefGoogle ScholarPubMed
Hosea, HJ, Rector, ES, Taylor, CG (2004) Dietary repletion can replenish reduced T cell subset numbers and lymphoid organ weight in zinc-deficient and energy-restricted rats. Br J Nutr 91, 741747. https://doi.org/10.1079/BJN20041104 CrossRefGoogle ScholarPubMed
Albert, MJ, Qadri, F, Wahed, MA, et al. (2003) Supplementation with zinc, but not vitamin A, improves seroconversion to vibriocidal antibody in children given an oral cholera vaccine. J Infect Dis 187, 909913. https://doi.org/10.1086/368132 CrossRefGoogle Scholar
Zhao, N, Wang, X, Zhang, Y, et al. (2013) Gestational zinc deficiency impairs humoral and cellular immune responses to hepatitis B vaccination in offspring mice. PLoS One 8, e73461. https://doi.org/10.1371/journal.pone.0073461 CrossRefGoogle ScholarPubMed
Hasan, R, Rink, L, Haase, H (2016) Chelation of free Zn2+ impairs chemotaxis, phagocytosis, oxidative burst, degranulation, and cytokine production by neutrophil granulocytes. Biol Trace Elem Res 171, 7988. https://doi.org/10.1007/s12011-015-0515-0 CrossRefGoogle ScholarPubMed
Dubben, S, Hönscheid, A, Winkler, K, et al. (2010) Cellular zinc homeostasis is a regulator in monocyte differentiation of HL-60 cells by 1 alpha,25-dihydroxyvitamin D3. J Leukocyte Biol 87, 833844. https://doi.org/10.1189/jlb.0409241 CrossRefGoogle ScholarPubMed
Hassan, KA, Pederick, VG, Elbourne, LD, et al. (2017) Zinc stress induces copper depletion in Acinetobacter baumannii . BMC Microbiol 17, 59. https://doi.org/10.1186/s12866-017-0965-y CrossRefGoogle ScholarPubMed
Xu, Z, Wang, P, Wang, H, Yu, ZH, et al. (2019) Zinc excess increases cellular demand for iron and decreases tolerance to copper in Escherichia coli . J Biol Chem 294, 1697816991. https://doi.org/10.1074/jbc.RA119.010023 CrossRefGoogle ScholarPubMed
Ong, CL, Walker, MJ, McEwan, AG (2015) Zinc disrupts central carbon metabolism and capsule biosynthesis in Streptococcus pyogenes . Sci Rep 5, 10799. https://doi.org/10.1038/srep10799 CrossRefGoogle ScholarPubMed
Duncan, A, Yacoubian, C, Watson, N, et al. (2015) The risk of copper deficiency in patients prescribed zinc supplements. J Clin Pathol 68, 723725. https://doi.org/10.1136/jclinpath-2014-202837 CrossRefGoogle ScholarPubMed
Li, G, Cheng, T, Yu, X (2021) The impact of trace elements on osteoarthritis. Front Med 8, 771297. https://doi.org/10.3389/fmed.2021.771297 CrossRefGoogle ScholarPubMed
Kitabayashi, C, Fukada, T, Kanamoto, M, et al. (2010) Zinc suppresses Th17 development via inhibition of STAT3 activation. Int Immunol 22, 375386. https://doi.org/10.1093/intimm/dxq017 CrossRefGoogle ScholarPubMed
Koropatnick, J & Zalups, RK (1997) Effect of non-toxic mercury, zinc or cadmium pretreatment on the capacity of human monocytes to undergo lipopolysaccharide-induced activation. Br J Pharmacol 120, 797806. https://doi.org/10.1038/sj.bjp.0700975 CrossRefGoogle ScholarPubMed
Hayashi, K, Ishizuka, S, Yokoyama, C, et al. (2008) Attenuation of interferon-gamma mRNA expression in activated Jurkat T cells by exogenous zinc via down-regulation of the calcium-independent PKC-AP-1 signaling pathway. Life Sci 83, 611. https://doi.org/10.1016/j.lfs.2008.04.022 CrossRefGoogle Scholar
Reinhold, D, Ansorge, S, Grüngreiff, K (1997) Zinc regulates DNA synthesis and IL-2, IL-6, and IL-10 production of PWM-stimulated PBMC and normalizes the periphere cytokine concentration in chronic liver disease. J Trace Elem Exp Med 10, 1927. https://doi.org/10.1002/(SICI)1520-670X(1997)10:1<19::AID-JTRA3>3.0.CO;2-# 3.0.CO;2-#>CrossRefGoogle Scholar
Hou, R, He, Y, Yan, G, et al. (2021) Zinc enzymes in medicinal chemistry. Eur J Med Chem 226, 113877. https://doi.org/10.1016/j.ejmech.2021.113877 CrossRefGoogle ScholarPubMed
Andreini, C, Banci, L, Bertini, I, et al. (2006) Zinc through the three domains of life. J Proteome Res 5, 31733178. https://doi.org/10.1021/pr0603699 CrossRefGoogle ScholarPubMed
Shankar, AH & Prasad, AS (1998) Zinc and immune function: the biological basis of altered resistance to infection. Am J Clin Nutr 68(2 Suppl), 447S463S. https://doi.org/10.1093/ajcn/68.2.447S CrossRefGoogle ScholarPubMed
Chakraborty, I, Kunti, S, Bandyopadhyay, M, et al. (2007) Evaluation of serum zinc level and plasma SOD activity in senile cataract patients under oxidative stress. Indian J Clin Biochem 22, 109113. https://doi.org/10.1007/BF02913326 CrossRefGoogle ScholarPubMed
Li, MS, Adesina, SE, Ellis, CL, et al. (2017) NADPH oxidase-2 mediates zinc deficiency-induced oxidative stress and kidney damage. Am J Physiol Cell Physiol 312, C47C55. https://doi.org/10.1152/ajpcell.00208.2016 CrossRefGoogle ScholarPubMed
Murphy, G (2017) Riding the metalloproteinase roller coaster. J Biol Chem 292, 77087718. https://doi.org/10.1074/jbc.X117.785295 CrossRefGoogle ScholarPubMed
Li, W, Xiong, Y, Chen, W, et al. (2020) Wnt/β-catenin signaling may induce senescence of chondrocytes in osteoarthritis. Exp Ther Med 20, 26312638. https://doi.org/10.3892/etm.2020.9022 Google ScholarPubMed
Little, CB, Barai, A, Burkhardt, D, et al. (2009) Matrix metalloproteinase 13-deficient mice are resistant to osteoarthritic cartilage erosion but not chondrocyte hypertrophy or osteophyte development. Arthritis Rheum 60, 37233733. https://doi.org/10.1002/art.25002 CrossRefGoogle ScholarPubMed
Wang, M, Sampson, ER, Jin, H, et al. (2013) MMP13 is a critical target gene during the progression of osteoarthritis. Arthritis Res Ther 15, R5. https://doi.org/10.1186/ar4133 CrossRefGoogle ScholarPubMed
Nuti, E, Casalini, F, Avramova, SI, et al. (2009) N-O-isopropyl sulfonamido-based hydroxamates: design, synthesis and biological evaluation of selective matrix metalloproteinase-13 inhibitors as potential therapeutic agents for osteoarthritis. J Med Chem 52, 47574773. https://doi.org/10.1021/jm900261f CrossRefGoogle ScholarPubMed
Wang, J, Ma, J, Gu, JH, et al. (2017) Regulation of type II collagen, matrix metalloproteinase-13 and cell proliferation by interleukin-1β is mediated by curcumin via inhibition of NF-κB signaling in rat chondrocytes. Mol Med Rep 16, 18371845. https://doi.org/10.3892/mmr.2017.6771 CrossRefGoogle ScholarPubMed
Gu, H, Jiao, Y, Yu, X, et al. (2017) Resveratrol inhibits the IL-1β-induced expression of MMP-13 and IL-6 in human articular chondrocytes via TLR4/MyD88-dependent and -independent signaling cascades. Int J Mol Med 39, 734740. https://doi.org/10.3892/ijmm.2017.2885 CrossRefGoogle Scholar
Klein, T & Bischoff, R (2011) Active metalloproteases of the A Disintegrin and Metalloprotease (ADAM) family: biological function and structure. J Proteome Res 10, 1733. https://doi.org/10.1021/pr100556z CrossRefGoogle ScholarPubMed
Yao, Q, Wu, X, Tao, C, et al. (2023) Osteoarthritis: pathogenic signaling pathways and therapeutic targets. Signal Transduct Targeted Therapy 8, 56. https://doi.org/10.1038/s41392-023-01330-w CrossRefGoogle ScholarPubMed
Glasson, SS, Askew, R, Sheppard, B, et al. (2005) Deletion of active ADAMTS5 prevents cartilage degradation in a murine model of osteoarthritis. Nature 434, 644648. https://doi.org/10.1038/nature03369 CrossRefGoogle Scholar
Guzman, RE, Evans, MG, Bove, S, et al. (2003) Mono-iodoacetate-induced histologic changes in subchondral bone and articular cartilage of rat femorotibial joints: an animal model of osteoarthritis. Toxicol Pathol 31, 619624. https://doi.org/10.1080/01926230390241800 CrossRefGoogle ScholarPubMed