Amino Acid Metabolism in Rheumatoid Arthritis: Friend or Foe?

Biomolecules - Tập 10 Số 9 - Trang 1280
Eleonora Panfili1, Roberto Gerli2, Ursula Grohmann1, Maria Teresa Pallotta1
1Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy
2Department of Medicine, University of Perugia, 06132 Perugia, Italy

Tóm tắt

In mammals, amino acid metabolism has evolved to act as a critical regulator of innate and adaptive immune responses. Rheumatoid arthritis (RA) is the most common form of inflammatory arthropathy sustained by autoimmune responses. We examine here the current knowledge of tryptophan and arginine metabolisms and the main immunoregulatory pathways in amino acid catabolism, in both RA patients and experimental models of arthritis. We found that l-tryptophan (Trp) metabolism and, in particular, the kynurenine pathway would exert protective effects in all experimental models and in some, but not all, RA patients, possibly due to single nucleotide polymorphisms in the gene coding for indoleamine 2,3-dioxygenase 1 (IDO1; the enzyme catalyzing the rate-limiting step of the kynurenine pathway). The function, i.e., either protective or pathogenetic, of the l-arginine (Arg) metabolism in RA was less clear. In fact, although immunoregulatory arginase 1 (ARG1) was highly induced at the synovial level in RA patients, its true functional role is still unknown, possibly because of few available preclinical data. Therefore, our analysis would indicate that amino acid metabolism represents a fruitful area of research for new drug targets for a more effective and safe therapy of RA and that further studies are demanding to pursue such an important objective.

Từ khóa


Tài liệu tham khảo

Karami, 2019, Genetic implications in the pathogenesis of rheumatoid arthritis; an updated review, Gene, 702, 8, 10.1016/j.gene.2019.03.033

Calabresi, 2018, One year in review 2018: Pathogenesis of rheumatoid arthritis, Clin. Exp. Rheumatol., 36, 175

Weyand, 2017, Immunometabolism in early and late stages of rheumatoid arthritis, Nat. Rev. Rheumatol., 13, 291, 10.1038/nrrheum.2017.49

Freitag, 2016, Immunometabolism and autoimmunity, Immunol. Cell Biol., 94, 925, 10.1038/icb.2016.77

Grohmann, 2010, Control of immune response by amino acid metabolism, Immunol. Rev., 236, 243, 10.1111/j.1600-065X.2010.00915.x

Mondanelli, 2019, Amino acid metabolism as drug target in autoimmune diseases, Autoimmun. Rev., 18, 334, 10.1016/j.autrev.2019.02.004

Murray, 2016, Amino acid auxotrophy as a system of immunological control nodes, Nat. Immunol., 17, 132, 10.1038/ni.3323

Mondanelli, 2017, The immune regulation in cancer by the amino acid metabolizing enzymes ARG and IDO, Curr. Opin. Pharmacol., 35, 30, 10.1016/j.coph.2017.05.002

Marchesi, 2015, The vocabulary of microbiome research: A proposal, Microbiome, 3, 31, 10.1186/s40168-015-0094-5

Bessede, 2014, Aryl hydrocarbon receptor control of a disease tolerance defence pathway, Nature, 511, 184, 10.1038/nature13323

Zelante, 2013, Tryptophan Catabolites from Microbiota Engage Aryl Hydrocarbon Receptor and Balance Mucosal Reactivity via Interleukin-22, Immunity, 39, 372, 10.1016/j.immuni.2013.08.003

Maeda, 2019, Host–microbiota interactions in rheumatoid arthritis, Exp. Mol. Med., 51, 1, 10.1038/s12276-019-0283-6

Lovelace, 2017, Recent evidence for an expanded role of the kynurenine pathway of tryptophan metabolism in neurological diseases, Neuropharmacology, 112, 373, 10.1016/j.neuropharm.2016.03.024

Sugimoto, 2006, Crystal structure of human indoleamine 2,3-dioxygenase: Catalytic mechanism of O2 incorporation by a heme-containing dioxygenase, Proc. Natl. Acad. Sci. USA, 103, 2611, 10.1073/pnas.0508996103

Schwarcz, 2004, The kynurenine pathway of tryptophan degradation as a drug target, Curr. Opin. Pharmacol., 4, 12, 10.1016/j.coph.2003.10.006

Cheong, 2018, Targeting the IDO1/TDO2-KYN-AhR Pathway for Cancer Immunotherapy—Challenges and Opportunities, Trends Pharmacol. Sci., 39, 307, 10.1016/j.tips.2017.11.007

Takikawa, 1988, Mechanism of interferon-gamma action. Characterization of indoleamine 2,3-dioxygenase in cultured human cells induced by interferon-gamma and evaluation of the enzyme-mediated tryptophan degradation in its anticellular activity, J. Biol. Chem., 263, 2041, 10.1016/S0021-9258(19)77982-4

Tykocinski, 2017, Synovial Fibroblasts Selectively Suppress Th1 Cell Responses through IDO1-Mediated Tryptophan Catabolism, J. Immunol., 198, 3109, 10.4049/jimmunol.1600600

Mellor, 2004, Ido expression by dendritic cells: Tolerance and tryptophan catabolism, Nat. Rev. Immunol., 4, 762, 10.1038/nri1457

Grohmann, 2003, Tolerance, DCs and tryptophan: Much ado about IDO, Trends Immunol., 24, 242, 10.1016/S1471-4906(03)00072-3

Pallotta, 2011, Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells, Nat. Immunol., 12, 870, 10.1038/ni.2077

Orabona, 2012, Different Partners, Opposite Outcomes: A New Perspective of the Immunobiology of Indoleamine 2,3-Dioxygenase, Mol. Med., 18, 834, 10.2119/molmed.2012.00029

Igari, 1987, Alteration of tryptophan metabolism in the synovial fluid of patients with rheumatoid arthritis and osteoarthritis, Tohoku J. Exp. Med., 153, 79, 10.1620/tjem.153.79

Schroecksnadel, 2005, Tryptophan degradation increases with stage in patients with rheumatoid arthritis, Clin. Rheumatol., 25, 334, 10.1007/s10067-005-0056-6

Forrest, 2003, Kynurenine and Neopterin Levels in Patients with Rheumatoid Arthritis and Osteoporosis During Drug Treatment, Adv. Exp. Med. Biol., 527, 287, 10.1007/978-1-4615-0135-0_32

Suzuki, 2010, Increased serum kynurenine/tryptophan ratio correlates with disease progression in lung cancer, Lung Cancer, 67, 361, 10.1016/j.lungcan.2009.05.001

Ozkan, 2011, Tryptophan degradation and neopterin levels in treated rheumatoid arthritis patients, Clin. Rheumatol., 31, 29, 10.1007/s10067-011-1767-5

Park, 2011, Myeloid differentiation primary response protein 88 blockade upregulates indoleamine 2,3-dioxygenase expression in rheumatoid synovial fibroblasts, Exp. Mol. Med., 43, 446, 10.3858/emm.2011.43.8.050

Rzeski, 2006, Kynurenic acid, an endogenous constituent of rheumatoid arthritis synovial fluid, inhibits proliferation of synoviocytes in vitro, Rheumatol. Int., 26, 422, 10.1007/s00296-005-0057-4

Kang, 2015, Downregulation of Tryptophan-related Metabolomic Profile in Rheumatoid Arthritis Synovial Fluid, J. Rheumatol., 42, 2003, 10.3899/jrheum.141505

Cribbs, 2014, Treg Cell Function in Rheumatoid Arthritis Is Compromised by CTLA-4 Promoter Methylation Resulting in a Failure to Activate the Indoleamine 2,3-Dioxygenase Pathway, Arthritis Rheumatol., 66, 2344, 10.1002/art.38715

Muz, 2009, Hypoxia. The role of hypoxia and HIF-dependent signalling events in rheumatoid arthritis, Arthritis Res. Ther., 11, 201, 10.1186/ar2568

Kaul, 2019, Hypoxia decreases the T helper cell-suppressive capacity of synovial fibroblasts by downregulating IDO1-mediated tryptophan metabolism, Rheumatology, 59, 1148, 10.1093/rheumatology/kez587

Koreny, 2007, Inhibition of indoleamine 2,3-dioxygenase-mediated tryptophan catabolism accelerates collagen-induced arthritis in mice, Arthritis Res. Ther., 9, R50, 10.1186/ar2205

Criado, 2009, Indoleamine 2,3 dioxygenase-mediated tryptophan catabolism regulates accumulation of Th1/Th17 cells in the joint in collagen-induced arthritis, Arthritis Rheum., 60, 1342, 10.1002/art.24446

Paleolog, 2010, Kynurenine metabolism in health and disease, Amino Acids, 41, 1173

2013, Systemic metabolism of tryptophan and its catabolites, kynurenine and 3-HAA, in mice with inflammatory arthritis, Gene, 512, 23, 10.1016/j.gene.2012.09.122

Chen, 2011, Amelioration of Rat Collagen-Induced Arthritis Through CD4+ T Cells Apoptosis and Synovial Interleukin-17 Reduction by Indoleamine 2,3-Dioxygenase Gene Therapy, Hum. Gene Ther., 22, 145, 10.1089/hum.2009.217

Chalise, 2016, IDO1 and TGF-beta Mediate Protective Effects of IFN-alpha in Antigen-Induced Arthritis, J. Immunol., 197, 3142, 10.4049/jimmunol.1502125

Royzman, 2019, Soluble CD83 Triggers Resolution of Arthritis and Sustained Inflammation Control in IDO Dependent Manner, Front. Immunol., 10, 633, 10.3389/fimmu.2019.00633

Orabona, C., Mondanelli, G., Pallotta, M.T., Carvalho, A., Albini, E., Fallarino, F., Vacca, C., Volpi, C., Belladonna, M.L., and Berioli, M.G. (2018). Deficiency of immunoregulatory indoleamine 2,3-dioxygenase 1in juvenile diabetes. JCI Insight, 3.

Mondanelli, 2020, Positive allosteric modulation of indoleamine 2,3-dioxygenase 1 restrains neuroinflammation, Proc. Natl. Acad. Sci. USA, 117, 3848, 10.1073/pnas.1918215117

Mondanelli, G., Di Battista, V., Pellanera, F., Mammoli, A., Macchiarulo, A., Gargaro, M., Mavridou, E., Matteucci, C., Ruggeri, L., and Orabona, C. (2020). A novel mutation of indoleamine 2,3-dioxygenase 1 causes a rapid proteasomal degradation and compromises protein function. J. Autoimmun., 102509.

Kouskoff, 1996, Organ-Specific Disease Provoked by Systemic Autoimmunity, Cell, 87, 811, 10.1016/S0092-8674(00)81989-3

Merlo, 2014, IDO2 is a critical mediator of autoantibody production and inflammatory pathogenesis in a mouse model of autoimmune arthritis, J. Immunol., 192, 2082, 10.4049/jimmunol.1303012

Merlo, 2016, IDO2 Modulates T Cell-Dependent Autoimmune Responses through a B Cell-Intrinsic Mechanism, J. Immunol., 196, 4487, 10.4049/jimmunol.1600141

Merlo, 2017, Therapeutic antibody targeting of indoleamine-2,3-dioxygenase (IDO2) inhibits autoimmune arthritis, Clin. Immunol., 179, 8, 10.1016/j.clim.2017.01.016

Merlo, 2016, IDO2: A Pathogenic Mediator of Inflammatory Autoimmunity, Clin. Med. Insights: Pathol., 9, 21

Platten, 2019, Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond, Nat. Rev. Drug Discov., 18, 379, 10.1038/s41573-019-0016-5

Haber, 1993, Identification of Tryptophan 2,3-Dioxygenase RNA in Rodent Brain, J. Neurochem., 60, 1159, 10.1111/j.1471-4159.1993.tb03269.x

Mezrich, 2010, An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells, J. Immunol., 185, 3190, 10.4049/jimmunol.0903670

Rothhammer, 2019, The aryl hydrocarbon receptor: An environmental sensor integrating immune responses in health and disease, Nat. Rev. Immunol., 19, 184, 10.1038/s41577-019-0125-8

Stockinger, 2014, The Aryl Hydrocarbon Receptor: Multitasking in the Immune System, Annu. Rev. Immunol., 32, 403, 10.1146/annurev-immunol-032713-120245

Nguyen, 2010, Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism, Proc. Natl. Acad. Sci. USA, 107, 19961, 10.1073/pnas.1014465107

Grohmann, 2015, The Coevolution of IDO1 and AhR in the Emergence of Regulatory T-Cells in Mammals, Front. Immunol., 6, 58, 10.3389/fimmu.2015.00058

Nguyen, 2013, Aryl hydrocarbon receptor and experimental autoimmune arthritis, Semin. Immunopathol., 35, 637, 10.1007/s00281-013-0392-6

Kazantseva, 2012, Dendritic cells provide a potential link between smoking and inflammation in rheumatoid arthritis, Arthritis Res. Ther., 14, R208, 10.1186/ar4046

O’Donnell, E.F., Saili, K.S., Koch, D.C., Kopparapu, P.R., Farrer, D., Bisson, W.H., Mathew, L.K., Sengupta, S., Kerkvliet, N.I., and Tanguay, R.L. (2010). The Anti-Inflammatory Drug Leflunomide Is an Agonist of the Aryl Hydrocarbon Receptor. PLoS ONE, 5.

Ogando, 2016, Notch-regulated miR-223 targets the aryl hydrocarbon receptor pathway and increases cytokine production in macrophages from rheumatoid arthritis patients, Sci. Rep., 6, 20223, 10.1038/srep20223

2008, Measurement of antioxidant ability of melatonin and serotonin by the DMPD and CUPRAC methods as trolox equivalent, J. Enzym. Inhib. Med. Chem., 23, 871, 10.1080/14756360701626223

Yoo, 2017, Neuroprotective action of N-acetyl serotonin in oxidative stress-induced apoptosis through the activation of both TrkB/CREB/BDNF pathway and Akt/Nrf2/Antioxidant enzyme in neuronal cells, Redox Biol., 11, 592, 10.1016/j.redox.2016.12.034

Zhao, 2019, Potential role of melatonin in autoimmune diseases, Cytokine Growth Factor Rev., 48, 1, 10.1016/j.cytogfr.2019.07.002

Skarlis, 2019, The role of melatonin in Multiple Sclerosis, Neurol. Sci., 41, 769, 10.1007/s10072-019-04137-2

Voog, 2004, Progression of radiographic changes in the temporomandibular joints of patients with rheumatoid arthritis in relation to inflammatory markers and mediators in the blood, Acta Odontol. Scand., 62, 7, 10.1080/00016350310007860

Bernardes, 2017, Serum serotonin levels and bone in rheumatoid arthritis patients, Rheumatol. Int., 37, 1891, 10.1007/s00296-017-3836-9

Wang, 2004, Leukotrienes mediate 5-hydroxytryptamine-induced plasm extravasation in the rat knee joint via CysLT-type receptors, Inflamm. Res., 53, 66, 10.1007/s00011-003-1224-2

Seidel, 2008, Serotonin mediates PGE2 overexpression through 5-HT2A and 5-HT3 receptor subtypes in serum-free tissue culture of macrophage-like synovial cells, Rheumatol. Int., 28, 1017, 10.1007/s00296-008-0564-1

Fiebich, 2004, Antiinflammatory effects of 5-HT3receptor antagonists in lipopolysaccharide-stimulated primary human monocytes, Scand. J. Rheumatol., 33, 28, 10.1080/03009740410006998

Fakhfouri, 2012, Impact of 5-HT3 receptor antagonists on peripheral and central diseases, Drug Discov. Today, 17, 741, 10.1016/j.drudis.2012.02.009

Coman, 2016, Serotonin Is Involved in Autoimmune Arthritis through Th17 Immunity and Bone Resorption, Am. J. Pathol., 186, 927, 10.1016/j.ajpath.2015.11.018

Snir, 2012, Genetic variation in the serotonin receptor gene affects immune responses in rheumatoid arthritis, Genes Immun., 14, 83, 10.1038/gene.2012.56

Kling, 2008, Genetic variations in the serotonin 5-HT2A receptor gene (HTR2A) are associated with rheumatoid arthritis, Ann. Rheum. Dis., 67, 1111, 10.1136/ard.2007.074948

Kling, 2006, Decreased density of serotonin 5-HT2A receptors in rheumatoid arthritis, Ann. Rheum. Dis., 65, 816, 10.1136/ard.2005.042473

Yang, 2015, Supplement of 5-hydroxytryptophan before induction suppresses inflammation and collagen-induced arthritis, Arthritis Res. Ther., 17, 1, 10.1186/s13075-015-0884-y

Brown, 2018, TNFalpha-dependent anhedonia and upregulation of hippocampal serotonin transporter activity in a mouse model of collagen-induced arthritis, Neuropharmacology, 137, 211, 10.1016/j.neuropharm.2018.04.023

Kim, 2012, Brain indoleamine 2,3-dioxygenase contributes to the comorbidity of pain and depression, J. Clin. Investig., 122, 2940, 10.1172/JCI61884

Schwarz, 2007, The immune-mediated alteration of serotonin and glutamate: Towards an integrated view of depression, Mol. Psychiatry, 12, 988, 10.1038/sj.mp.4002006

Nerurkar, 2019, Rheumatoid arthritis and depression: An inflammatory perspective, Lancet Psychiatry, 6, 164, 10.1016/S2215-0366(18)30255-4

Krishnadas, 2011, Sustained remission of rheumatoid arthritis with a specific serotonin reuptake inhibitor antidepressant: A case report and review of the literature, J. Med. Case Rep., 5, 112, 10.1186/1752-1947-5-112

Sacre, 2010, Fluoxetine and citalopram exhibit potent antiinflammatory activity in human and murine models of rheumatoid arthritis and inhibit toll-like receptors, Arthritis Rheum., 62, 683, 10.1002/art.27304

Liou, Y.-S., Lin, T.-K., Chen, H.-Y., and Jong, G.-P. (2019). Medications associated with fracture risk in patients with rheumatoid arthritis. Ann. Rheum. Dis.

Tosini, 2012, N-acetylserotonin: Neuroprotection, neurogenesis, and the sleepy brain, Neuroscientist, 18, 645, 10.1177/1073858412446634

Macdonald, I.J., Huang, C.-C., Liu, S.-C., and Tang, C. (2020). Reconsidering the Role of Melatonin in Rheumatoid Arthritis. Int. J. Mol. Sci., 21.

Hansson, 1990, Constant darkness enhances autoimmunity to type II collagen and exaggerates development of collagen-induced arthritis in DBA/1 mice, J. Neuroimmunol., 27, 79, 10.1016/0165-5728(90)90139-E

Hansson, 1992, The pineal hormone melatonin exaggerates development of collagen-induced arthritis in mice, J. Neuroimmunol., 39, 23, 10.1016/0165-5728(92)90171-G

Hansson, 1993, Pinealectomy ameliorates collagen II-induced arthritis in mice, Clin. Exp. Immunol., 92, 432, 10.1111/j.1365-2249.1993.tb03416.x

Bang, 2010, Melatonin attenuates clock gene Cryptochrome1, which may aggravate mouse anti-type II collagen antibody-induced arthritis, Rheumatol. Int., 32, 379, 10.1007/s00296-010-1641-9

Molinero, 2005, Dual effect of melatonin as proinflammatory and antioxidant in collagen-induced arthritis in rats, J. Pineal Res., 38, 93, 10.1111/j.1600-079X.2004.00175.x

Huang, 2019, Melatonin attenuates TNF-alpha and IL-1beta expression in synovial fibroblasts and diminishes cartilage degradation: Implications for the treatment of rheumatoid arthritis, J. Pineal Res., 66, e12560, 10.1111/jpi.12560

Chen, 2002, Effects and mechanisms of melatonin on inflammatory and immune responses of adjuvant arthritis rat, Int. Immunopharmacol., 2, 1443, 10.1016/S1567-5769(02)00088-7

Saleh, 2007, Serum melatonin in juvenile rheumatoid arthritis: Correlation with disease activity, Pak. J. Biol. Sci., 10, 1471, 10.3923/pjbs.2007.1471.1476

Lin, 2013, Modulation by Melatonin of the Pathogenesis of Inflammatory Autoimmune Diseases, Int. J. Mol. Sci., 14, 11742, 10.3390/ijms140611742

Sulli, 2002, Melatonin serum levels in rheumatoid arthritis, Ann. N. Y. Acad. Sci., 966, 276, 10.1111/j.1749-6632.2002.tb04227.x

Afkhamizadeh, 2014, Morning melatonin serum values do not correlate with disease activity in rheumatoid arthritis: A cross-sectional study, Rheumatol. Int., 34, 1145, 10.1007/s00296-013-2930-x

Ha, 2005, Positive relationship between melatonin receptor type 1B polymorphism and rheumatoid factor in rheumatoid arthritis patients in the Korean population, J. Pineal Res., 39, 201, 10.1111/j.1600-079X.2005.00237.x

Korkmaz, 2008, Melatonin as an adjuvant therapy in patients with rheumatoid arthritis, Br. J. Clin. Pharmacol., 66, 316, 10.1111/j.1365-2125.2008.03181.x

Bronte, 2005, Regulation of immune responses by L-arginine metabolism, Nat. Rev. Immunol., 5, 641, 10.1038/nri1668

Rodriguez, 2006, l-arginine availability regulates T-lymphocyte cell-cycle progression, Blood, 109, 1568, 10.1182/blood-2006-06-031856

Wu, 1998, Arginine metabolism: Nitric oxide and beyond, Biochem. J., 336, 1, 10.1042/bj3360001

Mills, 2012, M1 and M2 Macrophages: Oracles of Health and Disease, Crit. Rev. Immunol., 32, 463, 10.1615/CritRevImmunol.v32.i6.10

Cardounel, 2006, Evidence for the Pathophysiological Role of Endogenous Methylarginines in Regulation of Endothelial NO Production and Vascular Function, J. Biol. Chem., 282, 879, 10.1074/jbc.M603606200

Konya, 2015, Asymmetric dimethylarginine, a biomarker of cardiovascular complications in diabetes mellitus, World J. Exp. Med., 5, 110, 10.5493/wjem.v5.i2.110

Wu, 2008, Arginine metabolism and nutrition in growth, health and disease, Amino Acids, 37, 153, 10.1007/s00726-008-0210-y

Durante, 2013, Role of Arginase in Vessel Wall Remodeling, Front. Immunol., 4, 111, 10.3389/fimmu.2013.00111

Ming, 2012, Arginase II Promotes Macrophage Inflammatory Responses Through Mitochondrial Reactive Oxygen Species, Contributing to Insulin Resistance and Atherogenesis, J. Am. Heart Assoc., 1, e000992, 10.1161/JAHA.112.000992

Munder, 2009, Arginase: An emerging key player in the mammalian immune system, Br. J. Pharmacol., 158, 638, 10.1111/j.1476-5381.2009.00291.x

Dzik, 2014, Evolutionary Roots of Arginase Expression and Regulation, Front. Immunol., 5, 544, 10.3389/fimmu.2014.00544

Corraliza, 2002, Increased expression of arginase II in patients with different forms of arthritis. Implications of the regulation of nitric oxide, J. Rheumatol., 29, 2261

Chandrasekharan, 2018, Elevated levels of plasma symmetric dimethylarginine and increased arginase activity as potential indicators of cardiovascular comorbidity in rheumatoid arthritis, Arthritis Res., 20, 123, 10.1186/s13075-018-1616-x

Prati, 2012, Treatment with the arginase inhibitor Nw-hydroxy-nor-L-arginine restores endothelial function in rat adjuvant-induced arthritis, Arthritis Res. Ther., 14, R130, 10.1186/ar3860

Bordy, 2018, Methotrexate did not improve endothelial function in rheumatoid arthritis: A study in rats with adjuvant-induced arthritis, Clin. Exp. Rheumatol., 37, 81

Miyoshi, 2016, Changes in the pattern of cytokine production from peripheral blood mononuclear cells in patients with rheumatoid arthritis treated with infliximab and their relation to plasma arginase activity, Int. J. Rheum. Dis., 21, 1907, 10.1111/1756-185X.12864

Hannemann, 2019, Transcription factor Fra-1 targets arginase-1 to enhance macrophage-mediated inflammation in arthritis, J. Clin. Investig., 129, 2669, 10.1172/JCI96832

Mishra, 2016, c-Jun Is Required for Nuclear Factor-κB–Dependent, LPS-Stimulated Fos-Related Antigen-1 Transcription in Alveolar Macrophages, Am. J. Respir. Cell Mol. Biol., 55, 667, 10.1165/rcmb.2016-0028OC

Pegg, 2016, Functions of Polyamines in Mammals, J. Biol. Chem., 291, 14904, 10.1074/jbc.R116.731661

Medina, 2019, Polyamines in mammalian pathophysiology, Cell. Mol. Life Sci., 76, 3987, 10.1007/s00018-019-03196-0

Watanabe, 1991, Estimation of polyamine binding to macromolecules and ATP in bovine lymphocytes and rat liver, J. Biol. Chem., 266, 20803, 10.1016/S0021-9258(18)54780-3

Karouzakis, 2012, Increased recycling of polyamines is associated with global DNA hypomethylation in rheumatoid arthritis synovial fibroblasts, Arthritis Rheum., 64, 1809, 10.1002/art.34340

Friedman, 2019, Methotrexate mechanism in treatment of rheumatoid arthritis, Jt. Bone Spine, 86, 301, 10.1016/j.jbspin.2018.07.004

Iezaki, 2012, Amelioration by the natural polyamine spermine of cartilage and bone destruction in rats with collagen-induced arthritis, J. Pharmacol. Sci., 119, 107, 10.1254/jphs.11241SC

Yamamoto, 2012, The natural polyamines spermidine and spermine prevent bone loss through preferential disruption of osteoclastic activation in ovariectomized mice, Br. J. Pharmacol., 166, 1084, 10.1111/j.1476-5381.2012.01856.x

Yeon, 2014, Natural polyamines inhibit the migration of preosteoclasts by attenuating Ca2+-PYK2-Src-NFATc1 signaling pathways, Amino Acids, 46, 2605, 10.1007/s00726-014-1797-9

Yeon, 2015, Arginase 1 is a negative regulator of osteoclast differentiation, Amino Acids, 48, 559, 10.1007/s00726-015-2112-0

Sandri, 2014, The Emerging Immunological Role of Post-Translational Modifications by Reactive Nitrogen Species in Cancer Microenvironment, Front. Immunol., 5, 69

Nagy, 2010, Central role of nitric oxide in the pathogenesis of rheumatoid arthritis and systemic lupus erythematosus, Arthritis Res., 12, 210, 10.1186/ar3045

Wilkinson, 1996, Increased expression of blood mononuclear cell nitric oxide synthase type 2 in rheumatoid arthritis patients, J. Exp. Med., 184, 1173, 10.1084/jem.184.3.1173

Nilforoushan, 2009, Nitric oxide enhances osteoclastogenesis possibly by mediating cell fusion, Nitric Oxide, 21, 27, 10.1016/j.niox.2009.04.002

Chae, 1997, Nitric oxide is a regulator of bone remodelling, J. Pharm. Pharmacol., 49, 897, 10.1111/j.2042-7158.1997.tb06132.x

Fulton, M.D., Brown, T., and Zheng, Y.G. (2019). The Biological Axis of Protein Arginine Methylation and Asymmetric Dimethylarginine. Int. J. Mol. Sci., 20.

Brown, 2019, Gut Microbiota Regulation of T Cells During Inflammation and Autoimmunity, Annu. Rev. Immunol., 37, 599, 10.1146/annurev-immunol-042718-041841

Wu, 2010, Gut-Residing Segmented Filamentous Bacteria Drive Autoimmune Arthritis via T Helper 17 Cells, Immunology, 32, 815

Tong, Y., Marion, T., Schett, G., Luo, Y., and Liu, Y. (2020). Microbiota and metabolites in rheumatic diseases. Autoimmun. Rev., 102530.

Rogier, 2017, Aberrant intestinal microbiota due to IL-1 receptor antagonist deficiency promotes IL-17- and TLR4-dependent arthritis, Microbiome, 5, 63, 10.1186/s40168-017-0278-2

Roager, 2018, Microbial tryptophan catabolites in health and disease, Nat. Commun., 9, 3294, 10.1038/s41467-018-05470-4

Gao, 2018, Impact of the Gut Microbiota on Intestinal Immunity Mediated by Tryptophan Metabolism, Front. Microbiol., 8, 13, 10.3389/fcimb.2018.00013

Wang, 2019, Bridging intestinal immunity and gut microbiota by metabolites, Cell. Mol. Life Sci., 76, 3917, 10.1007/s00018-019-03190-6

Rooks, 2016, Gut microbiota, metabolites and host immunity, Nat. Rev. Immunol., 16, 341, 10.1038/nri.2016.42

Holmes, 2008, Metabolic Phenotyping in Health and Disease, Cell, 134, 714, 10.1016/j.cell.2008.08.026

Qiu, 2012, The Aryl Hydrocarbon Receptor Regulates Gut Immunity through Modulation of Innate Lymphoid Cells, Immunology, 36, 92

Miani, 2018, Gut Microbiota-Stimulated Innate Lymphoid Cells Support beta-Defensin 14 Expression in Pancreatic Endocrine Cells, Preventing Autoimmune Diabetes, Cell Metab., 28, 557, 10.1016/j.cmet.2018.06.012

Ji, Y., Gao, Y., Chen, H., Yin, Y., and Zhang, W. (2019). Indole-3-Acetic Acid Alleviates Nonalcoholic Fatty Liver Disease in Mice via Attenuation of Hepatic Lipogenesis, and Oxidative and Inflammatory Stress. Nutrients, 11.

Lamas, 2016, CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands, Nat. Med., 22, 598, 10.1038/nm.4102

Scher, 2015, Decreased bacterial diversity characterizes the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease, Arthritis Rheumatol., 67, 128, 10.1002/art.38892

Wilck, 2017, Salt-responsive gut commensal modulates TH17 axis and disease, Nature, 551, 585, 10.1038/nature24628

Ma, 2019, Immunomodulatory effect of human umbilical cord mesenchymal stem cells on T lymphocytes in rheumatoid arthritis, Int. Immunopharmacol., 74, 105687, 10.1016/j.intimp.2019.105687

Li, 2020, Human Umbilical Mesenchymal Stem Cells Display Therapeutic Potential in Rheumatoid Arthritis by Regulating Interactions Between Immunity and Gut Microbiota via the Aryl Hydrocarbon Receptor, Front. Cell Dev. Biol., 8, 131, 10.3389/fcell.2020.00131

Sakurai, T., Odamaki, T., and Xiao, J.-Z. (2019). Production of Indole-3-Lactic Acid by Bifidobacterium Strains Isolated fromHuman Infants. Microorganisms, 7.

Sassi, 2017, Gut Microbiota, Immune System, and Bone, Calcif. Tissue Int., 102, 415

Asquith, 2017, Intestinal Metabolites Are Profoundly Altered in the Context of HLA-B27 Expression and Functionally Modulate Disease in a Rat Model of Spondyloarthritis, Arthritis Rheumatol., 69, 1984, 10.1002/art.40183

Zhang, 2015, The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment, Nat. Med., 21, 895, 10.1038/nm.3914

Shoenfeld, 2019, The microbiome in autoimmune diseases, Clin. Exp. Immunol., 195, 74

2017, Intestinal Dysbiosis and Rheumatoid Arthritis: A Link between Gut Microbiota and the Pathogenesis of Rheumatoid Arthritis, J. Immunol. Res., 2017, 1

Rosser, 2020, Microbiota-Derived Metabolites Suppress Arthritis by Amplifying Aryl-Hydrocarbon Receptor Activation in Regulatory B Cells, Cell Metab., 31, 837, 10.1016/j.cmet.2020.03.003