34
Views
0
CrossRef citations to date
0
Altmetric
Review

The role of adipokines in the pathogenesis of psoriasis – a focus on resistin, omentin-1 and vaspin

, , &
Received 15 Feb 2024, Accepted 28 Jun 2024, Published online: 05 Jul 2024

References

  • Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin Endocrinol Metab. 2004;89(6):2548–2556. doi: 10.1210/jc.2004-0395
  • Wang X, Zhang S, Li Z. Adipokines in glucose and lipid metabolism. Adipocyte. 2023;12(1):2202976. doi: 10.1080/21623945.2023.2202976
  • Boehncke WH, Schön MP. Psoriasis. Lancet. 2015;386(9997):983–994. doi: 10.1016/S0140-6736(14)61909-7
  • Słuczanowska-Głabowska S, Staniszewska M, Marchlewicz M, et al. Adiponectin, leptin and resistin in patients with psoriasis. J Clin Med. 2023;12(2):663. doi: 10.3390/jcm12020663
  • Takahashi H, Tsuji H, Honma M, et al. Increased plasma resistin and decreased omentin levels in Japanese patients with psoriasis. Arch Dermatol Res. 2013;305(2):113–116. doi: 10.1007/s00403-012-1310-9
  • Noe MH, Shin DB, Wan MT, et al. Objective measures of psoriasis severity predict mortality: a prospective population-based cohort study. J Invest Dermatol. 2018;138(1):228–230. doi: 10.1016/j.jid.2017.07.841
  • Egeberg A, Andersen YMF, Thyssen JP. Prevalence and characteristics of psoriasis in Denmark: findings from the Danish skin cohort. BMJ Open. 2019;9(3):e028116. doi: 10.1136/bmjopen-2018-028116
  • Damiani G, Bragazzi NL, Karimkhani Aksut C, et al. The Global, Regional, and National Burden of Psoriasis: results and insights from the Global Burden of Disease 2019 study. Front Med. 2021;8:743180. doi: 10.3389/fmed.2021.743180
  • Kim WB, Jerome D, Yeung J. Diagnosis and management of psoriasis. Can Fam Physician. 2017;63(4):278–285.
  • Micali G, Verzì AE, Giuffrida G, et al. Inverse psoriasis: from diagnosis to current treatment options. Clin Cosmet Investig Dermatol. 2019;12:953–959. doi: 10.2147/CCID.S189000
  • Menter A, Van Voorhees AS, Hsu S. Pustular psoriasis: a narrative review of recent developments in pathophysiology and therapeutic options. Dermatol Ther (Heidelb). 2021;11(6):1917–1929. doi: 10.1007/s13555-021-00612-x
  • Singh RK, Lee KM, Ucmak D, et al. Erythrodermic psoriasis: pathophysiology and current treatment perspectives. Psoriasis. 2016;6:93–104. doi: 10.2147/PTT.S101232
  • FitzGerald O, Ogdie A, Chandran V, et al. Psoriatic arthritis. Nat Rev Dis Primers. 2021;7(1):59. doi: 10.1038/s41572-021-00293-y
  • Loo WY, Tee YC, Han WH, et al. Predictive factors of psoriatic arthritis in a diverse population with psoriasis. J Int Med Res. 2024;52(1):3000605231221014. doi: 10.1177/03000605231221014
  • Prashant P, Garg R, Kataria U, et al. Autoimmune thyroid disease in psoriasis patients: a case-control study. Cureus. 2023;15(12):e50197. doi: 10.7759/cureus.50197
  • Daugaard C, Iversen L, Hjuler KF. Comorbidity in adult psoriasis: considerations for the clinician. Psoriasis (Auckl). 2022;12:139–150. doi: 10.2147/PTT.S328572
  • Trafford AM, Parisi R, Kontopantelis E, et al. Association of psoriasis with the risk of developing or dying of cancer: a systematic review and meta-analysis. JAMA Dermatol. 2019;155(12):1390–1403. doi: 10.1001/jamadermatol.2019.3056
  • Sagi L, Trau H. The koebner phenomenon. Clin Dermatol. 2011;29(2):231–236. doi: 10.1016/j.clindermatol.2010.09.014
  • Vičić M, Kaštelan M, Brajac I, et al. Current concepts of psoriasis immunopathogenesis. Int J Mol Sci. 2021;22(21):11574. doi: 10.3390/ijms222111574
  • Ikutama R, Peng G, Tsukamoto S, et al. Cathelicidin LL-37 activates human keratinocyte autophagy through the P2X₇, mechanistic target of rapamycin, and MAPK pathways. J Invest Dermatol. 2023;143(5):751–61.e7. doi: 10.1016/j.jid.2022.10.020
  • Lande R, Botti E, Jandus C, et al. The antimicrobial peptide LL37 is a T-cell autoantigen in psoriasis. Nat Commun. 2014;5(1):5621. doi: 10.1038/ncomms6621
  • Kim TG, Kim SH, Lee MG. The origin of skin dendritic cell network and its role in psoriasis. Int J Mol Sci. 2017;19(1):42. doi: 10.3390/ijms19010042
  • Schlaak JF, Buslau M, Jochum W, et al. T cells involved in psoriasis vulgaris belong to the Th1 subset. J Invest Dermatol. 1994;102(2):145–149. doi: 10.1111/1523-1747.ep12371752
  • Kurtovic NO, Halilovic EK. Serum concentrations of Interferon Gamma (IFN-γ) in patients with psoriasis: correlation with clinical type and severity of the disease. Med Arch. 2018;72(6):410–413. doi: 10.5455/medarh.2018.72.410-413
  • Aguilar-Flores C, Castro-Escamilla O, Ortega-Rocha EM, et al. Association of pathogenic Th17 cells with the disease severity and its potential implication for biological treatment selection in psoriasis patients. Mediators Inflamm. 2020;2020:8065147. doi: 10.1155/2020/8065147
  • Chen L, Deshpande M, Grisotto M, et al. Skin expression of IL-23 drives the development of psoriasis and psoriatic arthritis in mice. Sci Rep. 2020;10(1):8259. doi: 10.1038/s41598-020-65269-6
  • Hou Y, Zhu L, Tian H, et al. IL-23-induced macrophage polarization and its pathological roles in mice with imiquimod-induced psoriasis. Protein Cell. 2018;9(12):1027–1038. doi: 10.1007/s13238-018-0505-z
  • Puig L. The role of IL 23 in the treatment of psoriasis. Expert Rev Clin Immunol. 2017;13(6):525–534. doi: 10.1080/1744666X.2017.1292137
  • Van Belle AB, de Heusch M, Lemaire MM, et al. IL-22 is required for imiquimod-induced psoriasiform skin inflammation in mice. J Immunol. 2012;188(1):462–469. doi: 10.4049/jimmunol.1102224
  • Mashiko S, Bouguermouh S, Rubio M, et al. Human mast cells are major IL-22 producers in patients with psoriasis and atopic dermatitis. J Allergy Clin Immunol. 2015;136(2):351–9.e1. doi: 10.1016/j.jaci.2015.01.033
  • Niu J, Song Z, Yang X, et al. Increased circulating follicular helper T cells and activated B cells correlate with disease severity in patients with psoriasis. J Eur Acad Dermatol Venereol. 2015;29(9):1791–1796. doi: 10.1111/jdv.13027
  • Sato Y, Ogawa E, Okuyama R. Role of innate immune cells in psoriasis. Int J Mol Sci. 2020;21(18):6604. doi: 10.3390/ijms21186604
  • Shao S, Fang H, Dang E, et al. Neutrophil extracellular traps promote inflammatory responses in psoriasis via activating epidermal TLR4/IL-36R crosstalk. Front Immunol. 2019;10:746. doi: 10.3389/fimmu.2019.00746
  • Benhadou F, Glitzner E, Brisebarre A, et al. Epidermal autonomous VEGFA/Flt1/Nrp1 functions mediate psoriasis-like disease. Sci Adv. 2020;6(2):eaax5849. doi: 10.1126/sciadv.aax5849
  • Chen Y, Tai Z, Zhu C, et al. Vascular endothelial growth factor a VEGFA inhibition: an effective treatment strategy for psoriasis. Int J Mol Sci. 2023;25(1):59. doi: 10.3390/ijms25010059
  • Choi CW, Kim BR, Yang S, et al. Regulatory T cells suppress skin inflammation in the imiquimod-induced psoriasis-like mouse model. J Dermatol Sci. 2020;98(3):199–202. doi: 10.1016/j.jdermsci.2020.04.008
  • Karagianni F, Pavlidis A, Malakou LS, et al. Predominant role of mTOR signaling in skin diseases with therapeutic potential. Int J Mol Sci. 2022;23(3):1693. doi: 10.3390/ijms23031693
  • Shin MJ, Kim HS, Lee P, et al. Mechanistic investigation of WWOX function in NF-kB-induced skin inflammation in psoriasis. Int J Mol Sci. 2023;25(1):167. doi: 10.3390/ijms25010167
  • Stuart PE, Tsoi LC, Nair RP, et al. Transethnic analysis of psoriasis susceptibility in South Asians and Europeans enhances fine-mapping in the MHC and genomewide. HGG Adv. 2022;3(1):100069. doi: 10.1016/j.xhgg.2021.100069
  • Mateu-Arrom L, Puig L. Genetic and epigenetic mechanisms of psoriasis. Genes (Basel). 2023;14(8):1619. doi: 10.3390/genes14081619
  • Snekvik I, Smith CH, Nilsen TIL, et al. Obesity, waist circumference, weight change, and risk of incident psoriasis: prospective data from the HUNT study. J Invest Dermatol. 2017;137(12):2484–2490. doi: 10.1016/j.jid.2017.07.822
  • Han JH, Lee JH, Han KD, et al. Increased risk of psoriasis in subjects with abdominal obesity: a nationwide population-based study. J Dermatol. 2019;46(8):695–701. doi: 10.1111/1346-8138.14939
  • Norden A, Rekhtman S, Strunk A, et al. Risk of psoriasis according to body mass index: a retrospective cohort analysis. J Am Acad Dermatol. 2022;86(5):1020–1026. doi: 10.1016/j.jaad.2021.06.012
  • Osman A, Nigro A, Taylor AC, et al. The effects of cardiometabolic comorbidities on biologic treatment for psoriasis with respect to PASI scores: a qualitative systematic review. Psoriasis (Auckl). 2024;14:1–10. doi: 10.2147/PTT.S441642
  • Armstrong AW, Pathophysiology RC. Clinical presentation, and treatment of psoriasis: a review. JAMA. 2020;323(19):1945–1960. doi: 10.1001/jama.2020.4006
  • Steppan CM, Bailey ST, Bhat S, et al. The hormone resistin links obesity to diabetes. Nature. 2001;409(6818):307–312. doi: 10.1038/35053000
  • Holcomb IN, Kabakoff RC, Chan B, et al. FIZZ1, a novel cysteine-rich secreted protein associated with pulmonary inflammation, defines a new gene family. Embo J. 2000;19(15):4046–4055. doi: 10.1093/emboj/19.15.4046
  • Steppan CM, Brown EJ, Wright CM, et al. A family of tissue-specific resistin-like molecules. Proc Natl Acad Sci USA. 2001;98(2):502–506. doi: 10.1073/pnas.98.2.502
  • Patel L, Buckels AC, Kinghorn IJ, et al. Resistin is expressed in human macrophages and directly regulated by PPAR gamma activators. Biochem Biophys Res Commun. 2003;300(2):472–476. doi: 10.1016/S0006-291X(02)02841-3
  • McManus DD, Lyass A, Ingelsson E, et al. Relations of circulating resistin and adiponectin and cardiac structure and function: the Framingham Offspring Study. Obesity (Silver Spring). 2012;20(9):1882–1886. doi: 10.1038/oby.2011.32
  • Lin Q, Johns RA. Resistin family proteins in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol. 2020;319(3):L422–L34. doi: 10.1152/ajplung.00040.2020
  • Harrison WJ, Bull JJ, Seltmann H, et al. Expression of lipogenic factors galectin-12, resistin, SREBP-1, and SCD in human sebaceous glands and cultured sebocytes. J Invest Dermatol. 2007;127(6):1309–1317. doi: 10.1038/sj.jid.5700743
  • Reilly MP, Lehrke M, Wolfe ML, et al. Resistin is an inflammatory marker of atherosclerosis in humans. Circulation. 2005;111(7):932–939. doi: 10.1161/01.CIR.0000155620.10387.43
  • Haugen F, Jørgensen A, Drevon CA, et al. Inhibition by insulin of resistin gene expression in 3T3-L1 adipocytes. FEBS Lett. 2001;507(1):105–108. doi: 10.1016/S0014-5793(01)02968-4
  • Shojima N, Sakoda H, Ogihara T, et al. Humoral regulation of resistin expression in 3T3-L1 and mouse adipose cells. Diabetes. 2002;51(6):1737–1744. doi: 10.2337/diabetes.51.6.1737
  • Rajala MW, Lin Y, Ranalletta M, et al. Cell type-specific expression and coregulation of murine resistin and resistin-like molecule-alpha in adipose tissue. Mol Endocrinol. 2002;16(8):1920–1930. doi: 10.1210/me.2002-0048
  • Kaser S, Kaser A, Sandhofer A, et al. Resistin messenger-RNA expression is increased by proinflammatory cytokines in vitro. Biochem Biophys Res Commun. 2003;309(2):286–290. doi: 10.1016/j.bbrc.2003.07.003
  • Bokarewa M, Nagaev I, Dahlberg L, et al. Resistin, an adipokine with potent proinflammatory properties. J Immunol. 2005;174(9):5789–5795. doi: 10.4049/jimmunol.174.9.5789
  • Jiang S, Park DW, Tadie JM, et al. Human resistin promotes neutrophil proinflammatory activation and neutrophil extracellular trap formation and increases severity of acute lung injury. J Immunol. 2014;192(10):4795–4803. doi: 10.4049/jimmunol.1302764
  • Kusminski CM, da Silva NF, Creely SJ, et al. The in vitro effects of resistin on the innate immune signaling pathway in isolated human subcutaneous adipocytes. J Clin Endocrinol Metab. 2007;92(1):270–276. doi: 10.1210/jc.2006-1151
  • Azzam ZE, Elneily D, Elgayar N, et al. Serum levels of resistin and its relationship with some pro-inflammatory cytokines in a cohort of Egyptian patients with Alzheimer’s disease. Endocr Metabolic Sci. 2020;1:100054. doi: 10.1016/j.endmts.2020.100054
  • Gao J, Deng M, Li Y, et al. Resistin as a systemic inflammation-related biomarker for sarcopenia in patients with chronic obstructive pulmonary disease. Front Nutr. 2022;9:921399. doi: 10.3389/fnut.2022.921399
  • Chen WC, Lu YC, Kuo SJ, et al. Resistin enhances IL-1β and TNF-α expression in human osteoarthritis synovial fibroblasts by inhibiting miR-149 expression via the MEK and ERK pathways. Faseb J. 2020;34(10):13671–13684. doi: 10.1096/fj.202001071R
  • Son YM, Ahn SM, Jang MS, et al. Immunomodulatory effect of resistin in human dendritic cells stimulated with lipoteichoic acid from Staphylococcus aureus. Biochem Biophys Res Commun. 2008;376(3):599–604. doi: 10.1016/j.bbrc.2008.09.037
  • Son YM, Ahn SM, Kim GR, et al. Resistin enhances the expansion of regulatory T cells through modulation of dendritic cells. BMC Immunol. 2010;11(1):33. doi: 10.1186/1471-2172-11-33
  • Li Y, Yang Q, Cai D, et al. Resistin, a novel host defense peptide of innate immunity. Front Immunol. 2021;12:699807. doi: 10.3389/fimmu.2021.699807
  • Kim HJ, Lee YS, Won EH, et al. Expression of resistin in the prostate and its stimulatory effect on prostate cancer cell proliferation. BJU Int. 2011;108(2 Pt 2):E77–83. doi: 10.1111/j.1464-410X.2010.09813.x
  • Senolt L, Housa D, Vernerová Z, et al. Resistin in rheumatoid arthritis synovial tissue, synovial fluid and serum. Ann Rheum Dis. 2007;66(4):458–463. doi: 10.1136/ard.2006.054734
  • Nieva-Vazquez A, Pérez-Fuentes R, Torres-Rasgado E, et al. Serum resistin levels are associated with adiposity and insulin sensitivity in obese Hispanic subjects. Metab Syndr Relat Disord. 2014;12(2):143–148. doi: 10.1089/met.2013.0118
  • Schlenker SM, Munhoz SI, Busanello AR, et al. Resistin serum levels and its association with clinical profile and carotid intima-media thickness in psoriasis: a cross-sectional study. An Bras Dermatol. 2023;98(6):799–805. doi: 10.1016/j.abd.2022.10.011
  • Coimbra S, Oliveira H, Reis F, et al. Circulating adipokine levels in Portuguese patients with psoriasis vulgaris according to body mass index, severity and therapy. J Eur Acad Dermatol Venereol. 2010;24(12):1386–1394. doi: 10.1111/j.1468-3083.2010.03647.x
  • Huang H, Shen E, Tang S, et al. Increased serum resistin levels correlate with psoriasis: a meta-analysis. Lipids Health Dis. 2015;14(1):44. doi: 10.1186/s12944-015-0039-9
  • Johnston A, Arnadottir S, Gudjonsson JE, et al. Obesity in psoriasis: leptin and resistin as mediators of cutaneous inflammation. Br J Dermatol. 2008;159(2):342–350. doi: 10.1111/j.1365-2133.2008.08655.x
  • Dikbas O, Tosun M, Bes C, et al. Serum levels of visfatin, resistin and adiponectin in patients with psoriatic arthritis and associations with disease severity. Int J Rheum Dis. 2016;19(7):672–677. doi: 10.1111/1756-185X.12444
  • Kyriakou A, Patsatsi A, Sotiriadis D, et al. Effects of treatment for psoriasis on circulating levels of leptin, adiponectin and resistin: a systematic review and meta-analysis. Br J Dermatol. 2018;179(2):273–281. doi: 10.1111/bjd.16437
  • Takahashi H, Tsuji H, Ishida-Yamamoto A, et al. Serum level of adiponectin increases and those of leptin and resistin decrease following the treatment of psoriasis. J Dermatol. 2013;40(6):475–476. doi: 10.1111/1346-8138.12122
  • Cacciapuoti S, Megna M, Salza E, et al. The effect of tildrakizumab on adipokines production in patients affected by psoriasis and obesity: preliminary results from a single center real-life study. J Dermatol Treat. 2024;35(1):2291323. doi: 10.1080/09546634.2023.2291323
  • Goodman WA, Levine AD, Massari JV, et al. IL-6 signaling in psoriasis prevents immune suppression by regulatory T cells. J Immunol. 2009;183(5):3170–3176. doi: 10.4049/jimmunol.0803721
  • Grossman RM, Krueger J, Yourish D, et al. Interleukin 6 is expressed in high levels in psoriatic skin and stimulates proliferation of cultured human keratinocytes. Proc Natl Acad Sci USA. 1989;86(16):6367–6371. doi: 10.1073/pnas.86.16.6367
  • Furue K, Ito T, Furue M. Differential efficacy of biologic treatments targeting the TNF-α/IL-23/IL-17 axis in psoriasis and psoriatic arthritis. Cytokine. 2018;111:182–188. doi: 10.1016/j.cyto.2018.08.025
  • Pine GM, Batugedara HM, Nair MG. Here, there and everywhere: Resistin-like molecules in infection, inflammation, and metabolic disorders. Cytokine. 2018;110:442–451. doi: 10.1016/j.cyto.2018.05.014
  • Pivarcsi A, Bodai L, Réthi B, et al. Expression and function of Toll-like receptors 2 and 4 in human keratinocytes. Int Immunol. 2003;15(6):721–730. doi: 10.1093/intimm/dxg068
  • Song PI, Park YM, Abraham T, et al. Human keratinocytes express functional CD14 and toll-like receptor 4. J Invest Dermatol. 2002;119(2):424–432. doi: 10.1046/j.1523-1747.2002.01847.x
  • Xu F, Xu J, Xiong X, et al. Salidroside inhibits MAPK, NF-κB, and STAT3 pathways in psoriasis-associated oxidative stress via SIRT1 activation. Redox Rep. 2019;24(1):70–74. doi: 10.1080/13510002.2019.1658377
  • Garcia-Rodriguez S, Arias-Santiago S, Perandrés-López R, et al. Increased gene expression of Toll-like receptor 4 on peripheral blood mononuclear cells in patients with psoriasis. J Eur Acad Dermatol Venereol. 2013;27(2):242–250. doi: 10.1111/j.1468-3083.2011.04372.x
  • Lai R, Xian D, Xiong X, et al. Proanthocyanidins: novel treatment for psoriasis that reduces oxidative stress and modulates Th17 and treg cells. Redox Rep. 2018;23(1):130–135. doi: 10.1080/13510002.2018.1462027
  • Yu XJ, Li CY, Dai HY, et al. Expression and localization of the activated mitogen-activated protein kinase in lesional psoriatic skin. Exp Mol Pathol. 2007;83(3):413–418. doi: 10.1016/j.yexmp.2007.05.002
  • Li Z, Wang X, Pan H, et al. Resistin promotes CCL4 expression through toll-like receptor-4 and activation of the p38-MAPK and NF-κB signaling pathways: implications for intervertebral disc degeneration. Osteoarthritis Cartilage. 2017;25(2):341–350. doi: 10.1016/j.joca.2016.10.002
  • Tsai HC, Cheng SP, Han CK, et al. Resistin enhances angiogenesis in osteosarcoma via the MAPK signaling pathway. Aging (Albany NY). 2019;11(21):9767–9777. doi: 10.18632/aging.102423
  • Hsieh YY, Shen CH, Huang WS, et al. Resistin-induced stromal cell-derived factor-1 expression through toll-like receptor 4 and activation of p38 MAPK/NFκB signaling pathway in gastric cancer cells. J Biomed Sci. 2014;21(1):59. doi: 10.1186/1423-0127-21-59
  • Silswal N, Singh AK, Aruna B, et al. Human resistin stimulates the pro-inflammatory cytokines TNF-alpha and IL-12 in macrophages by NF-kappaB-dependent pathway. Biochem Biophys Res Commun. 2005;334(4):1092–1101. doi: 10.1016/j.bbrc.2005.06.202
  • Lowes MA, Suárez- Fariñfariñas M, Krueger JG. Immunology of psoriasis. Annu Rev Immunol. 2014;32(1):227–255. doi: 10.1146/annurev-immunol-032713-120225
  • Kagami S, Rizzo HL, Lee JJ, et al. Circulating Th17, Th22, and Th1 cells are increased in psoriasis. J Invest Dermatol. 2010;130(5):1373–1383. doi: 10.1038/jid.2009.399
  • Eyerich S, Eyerich K, Pennino D, et al. Th22 cells represent a distinct human T cell subset involved in epidermal immunity and remodeling. J Clin Invest. 2009;119(12):3573–3585. doi: 10.1172/JCI40202
  • Zhuang L, Ma W, Yan J, et al. Evaluation of the effects of IL‑22 on the proliferation and differentiation of keratinocytes in vitro. Mol Med Rep. 2020;22(4):2715–2722. doi: 10.3892/mmr.2020.11348
  • Di Cesare A, Di Meglio P, Nestle FO. The IL-23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol. 2009;129(6):1339–1350. doi: 10.1038/jid.2009.59
  • Zhou L, Ivanov II, Spolski R, et al. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol. 2007;8(9):967–974. doi: 10.1038/ni1488
  • Veldhoen M, Hocking RJ, Atkins CJ, et al. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 2006;24(2):179–189. doi: 10.1016/j.immuni.2006.01.001
  • Bovenschen HJ, van de Kerkhof PC, van Erp PE, et al. Foxp3+ regulatory T cells of psoriasis patients easily differentiate into IL-17A-producing cells and are found in lesional skin. J Invest Dermatol. 2011;131(9):1853–1860. doi: 10.1038/jid.2011.139
  • Di T, Zhai C, Zhao J, et al. Taxifolin inhibits keratinocyte proliferation and ameliorates imiquimod-induced psoriasis-like mouse model via regulating cytoplasmic phospholipase A2 and PPAR-γ pathway. Int Immunopharmacol. 2021;99:107900. doi: 10.1016/j.intimp.2021.107900
  • Sivasami P, Elkins C, Diaz-Saldana PP, et al. Obesity-induced dysregulation of skin-resident PPARγ + Treg cells promotes IL-17A-mediated psoriatic inflammation. Immunity. 2023;56(8):1844–61.e6. doi: 10.1016/j.immuni.2023.06.021
  • Yang RZ, Lee MJ, Hu H, et al. Identification of omentin as a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action. Am J Physiol Endocrinol Metab. 2006;290(6):E1253–61. doi: 10.1152/ajpendo.00572.2004
  • Pan X, Kaminga AC, Wen SW, et al. Omentin-1 in diabetes mellitus: A systematic review and meta-analysis. PLOS ONE. 2019;14(12):e0226292. doi: 10.1371/journal.pone.0226292
  • Sperling M, Grzelak T, Pelczyńska M, et al. Association of Serum Omentin-1 concentration with the content of adipose tissue and glucose tolerance in subjects with central obesity. Biomedicines. 2023;11(2):331. doi: 10.3390/biomedicines11020331
  • Bakry OA, El Farargy S, Ghanayem N, et al. Serum Omentin-1 in Psoriasis. Indian J Dermatol. 2018;63(1):85–87. doi: 10.4103/ijd.IJD_222_17
  • Zhang C, Zhu KJ, Liu JL, et al. Omentin-1 plasma levels and omentin-1 expression are decreased in psoriatic lesions of psoriasis patients. Arch Dermatol Res. 2015;307(5):455–459. doi: 10.1007/s00403-015-1549-z
  • Ismail SA, Mohamed SA. Serum levels of visfatin and omentin-1 in patients with psoriasis and their relation to disease severity. Br J Dermatol. 2012;167(2):436–439. doi: 10.1111/j.1365-2133.2012.10980.x
  • Ko CY, Lin YY, Achudhan D, et al. Omentin-1 ameliorates the progress of osteoarthritis by promoting IL-4-dependent anti-inflammatory responses and M2 macrophage polarization. Int J Biol Sci. 2023;19(16):5275–5289. doi: 10.7150/ijbs.86701
  • Watanabe K, Watanabe R, Konii H, et al. Counteractive effects of omentin-1 against atherogenesis†. Cardiovasc Res. 2016;110(1):118–128. doi: 10.1093/cvr/cvw016
  • Yamawaki H, Kuramoto J, Kameshima S, et al. Omentin, a novel adipocytokine inhibits TNF-induced vascular inflammation in human endothelial cells. Biochem Biophys Res Commun. 2011;408(2):339–343. doi: 10.1016/j.bbrc.2011.04.039
  • Wang J, Gao Y, Lin F, et al. Omentin-1 attenuates lipopolysaccharide (LPS)-induced U937 macrophages activation by inhibiting the TLR4/MyD88/NF-κB signaling. Arch Biochem Biophys. 2020;679:108187. doi: 10.1016/j.abb.2019.108187
  • Tao M, Yan W, Chen C, et al. Omentin-1 ameliorates experimental inflammatory bowel disease via Nrf2 activation and redox regulation. Life Sci. 2023;328:121847. doi: 10.1016/j.lfs.2023.121847
  • Zhou H, Zhang Z, Qian G, et al. Omentin-1 attenuates adipose tissue inflammation via restoration of TXNIP/NLRP3 signaling in high-fat diet-induced obese mice. Fundam Clin Pharmacol. 2020;34(6):721–735. doi: 10.1111/fcp.12575
  • Swanson KV, Deng M, Ting JP. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat Rev Immunol. 2019;19(8):477–489. doi: 10.1038/s41577-019-0165-0
  • Salvoza N, Giraudi P, Gazzin S, et al. The potential role of omentin-1 in obesity-related metabolic dysfunction-associated steatotic liver disease: evidence from translational studies. J Transl Med. 2023;21(1):906. doi: 10.1186/s12967-023-04770-8
  • Ma L, Zhang X, Zhang C, et al. Omentin-1 attenuates inflammation and barrier damage in DSS-induced ulcerative colitis in mice by inhibiting endoplasmic reticulum stress. Gen Physiol Biophys. 2022;41(3):221–230. doi: 10.4149/gpb_2022012
  • D’Amico F, Costantino G, Salvatorelli L, et al. Inverse correlation between the expression of AMPK/SIRT1 and NAMPT in psoriatic skin: a pilot study. Adv Med Sci. 2022;67(2):262–268. doi: 10.1016/j.advms.2022.07.001
  • Shen H, Sha Y, Huang J, et al. The roles of AMPK-mediated autophagy and mitochondrial autophagy in a mouse model of imiquimod-induced psoriasis. Am J Transl Res. 2021;13(11):12626–12637.
  • Lin SH, Chuang HY, Ho JC, et al. Treatment with TNF-α inhibitor rectifies M1 macrophage polarization from blood CD14+ monocytes in patients with psoriasis independent of STAT1 and IRF-1 activation. J Dermatol Sci. 2018;91(3):276–284. doi: 10.1016/j.jdermsci.2018.05.009
  • Rebholz B, Haase I, Eckelt B, et al. Crosstalk between keratinocytes and adaptive immune cells in an IkappaBalpha protein-mediated inflammatory disease of the skin. Immunity. 2007;27(2):296–307. doi: 10.1016/j.immuni.2007.05.024
  • Ren X, Zhong W, Li W, et al. Human umbilical cord-derived mesenchymal stem cells alleviate psoriasis through TNF-α/NF-κB/MMP13 pathway. Inflammation. 2023;46(3):987–1001. doi: 10.1007/s10753-023-01785-7
  • Hseu JH, Chan CI, Vadivalagan C, et al. Tranexamic acid improves psoriasis-like skin inflammation: Evidence from in vivo and in vitro studies. Biomed Pharmacother. 2023;166:115307. doi: 10.1016/j.biopha.2023.115307
  • Zhang C, Tang B, Zheng X, et al. Analysis of the potential pyroptosis mechanism in psoriasis and experimental validation of NLRP3 in vitro and in vivo. Int Immunopharmacol. 2023;124(Pt A):110811. doi: 10.1016/j.intimp.2023.110811
  • Mao J, Ma X, Zhu J, et al. Ginsenoside Rg1 ameliorates psoriasis-like skin lesions by suppressing proliferation and NLRP3 inflammasomes in keratinocytes. J Food Biochem. 2022;46(5):e14053. doi: 10.1111/jfbc.14053
  • Qiao P, Zhang C, Yu J, et al. Quinolinic acid, a tryptophan metabolite of the skin microbiota, Negatively Regulates NLRP3 Inflammasome through AhR in psoriasis. J Invest Dermatol. 2022;142(8):2184–93.e6. doi: 10.1016/j.jid.2022.01.010
  • Lee YJ, Bae JH, Kang SG, et al. Pro-oxidant status and Nrf2 levels in psoriasis vulgaris skin tissues and dimethyl fumarate-treated HaCaT cells. Arch Pharm Res. 2017;40(9):1105–1116. doi: 10.1007/s12272-017-0955-5
  • Ma C, Gu C, Lian P, et al. Sulforaphane alleviates psoriasis by enhancing antioxidant defense through KEAP1-NRF2 pathway activation and attenuating inflammatory signaling. Cell Death Dis. 2023;14(11):768. doi: 10.1038/s41419-023-06234-9
  • He Y, Jia H, Yang Q, et al. Specific activation of CB2R ameliorates psoriasis-like skin lesions by inhibiting inflammation and oxidative stress. Inflammation. 2023;46(4):1255–1271. doi: 10.1007/s10753-023-01805-6
  • Yang L, Fan X, Cui T, et al. Nrf2 promotes keratinocyte proliferation in psoriasis through up-regulation of keratin 6, keratin 16, and keratin 17. J Invest Dermatol. 2017;137(10):2168–2176. doi: 10.1016/j.jid.2017.05.015
  • Zhao M, Luo J, Xiao B, et al. Endoplasmic reticulum stress links psoriasis vulgaris with keratinocyte inflammation. Postepy Dermatol Alergol. 2020;37(1):34–40. doi: 10.5114/ada.2020.93382
  • Xue Y, Jiang L, Cheng Q, et al. Adipokines in psoriatic arthritis patients: the correlations with osteoclast precursors and bone erosions. PLOS ONE. 2012;7(10):e46740. doi: 10.1371/journal.pone.0046740
  • Senolt L, Polanská M, Filková M, et al. Vaspin and omentin: new adipokines differentially regulated at the site of inflammation in rheumatoid arthritis. Ann Rheum Dis. 2010;69(7):1410–1411. doi: 10.1136/ard.2009.119735
  • Wu Y, Li X, Ke J, et al. Increased expression of omentin-1 is associated with synovitis and bone destruction in rheumatoid arthritis. Clin Exp Rheumatol. 2024. doi: 10.55563/clinexprheumatol/7pcgv7
  • Xu L, Zhu GB, Wang L, et al. Synovial fluid omentin-1 levels are inversely correlated with radiographic severity of knee osteoarthritis. J Investig Med. 2012;60(3):583–586. doi: 10.2310/JIM.0b013e31824443cb
  • Hida K, Wada J, Eguchi J, et al. Visceral adipose tissue-derived serine protease inhibitor: a unique insulin-sensitizing adipocytokine in obesity. Proc Natl Acad Sci USA. 2005;102(30):10610–10615. doi: 10.1073/pnas.0504703102
  • Kurowska P, Mlyczyńska E, Dawid M, et al. Review: Vaspin (SERPINA12) expression and function in endocrine cells. Cells. 2021;10(7):1710. doi: 10.3390/cells10071710
  • Nakatsuka A, Wada J, Iseda I, et al. Vaspin is an adipokine ameliorating ER stress in obesity as a ligand for cell-surface GRP78/MTJ-1 complex. Diabetes. 2012;61(11):2823–2832. doi: 10.2337/db12-0232
  • Kurowska P, Mlyczyńska E, Dawid M, et al. Role of vaspin in porcine ovary: effect on signaling pathways and steroid synthesis via GRP78 receptor and protein kinase A†. Biol Reprod. 2020;102(6):1290–1305. doi: 10.1093/biolre/ioaa027
  • Kurowska P, Mlyczyńska E, Dawid M, et al. In vitro effects of Vaspin on porcine granulosa cell proliferation, cell cycle progression, and apoptosis by activation of GRP78 receptor and several kinase signaling pathways including MAP3/1, AKT, and STAT3. Int J Mol Sci. 2019;20(22):5816. doi: 10.3390/ijms20225816
  • Weiner J, Zieger K, Pippel J, et al. Molecular mechanisms of vaspin action - from adipose tissue to skin and bone, from blood vessels to the brain. Adv Exp Med Biol. 2019;1111:159–188.
  • Tindall CA, Möhlis K, Rapöhn I, et al. LRP1 is the cell-surface endocytosis receptor for vaspin in adipocytes. FEBS J. 2024;291(10):2134–2154. doi: 10.1111/febs.16991
  • Dizen-Namdar N, Akcilar R, Bayat Z. Association between vaspin rs2236242 gene polymorphism and psoriasis vulgaris. Skin Pharmacol Physiol. 2020;33(6):317–322. doi: 10.1159/000512124
  • Sayed KS, Said ER, Mohammed FN, et al. Down-regulation of tissue levels of serine protease inhibitor (vaspin) in psoriasis vulgaris patients: a possible mechanism of narrowband ultraviolet B radiation. Arch Dermatol Res. 2019;311(8):637–641. doi: 10.1007/s00403-019-01947-4
  • Ataseven A, Kesli R. Novel inflammatory markers in psoriasis vulgaris: vaspin, vascular adhesion protein-1 (VAP-1), and YKL-40. G Ital Dermatol Venereol. 2016;151(3):244–250.
  • Kiluk P, Baran A, Kaminski TW, et al. Decreased levels of vaspin and its potential association with cardiometabolic risk in patients with psoriasis: preliminary results. Postepy Dermatol Alergol. 2022;39(2):307–315. doi: 10.5114/ada.2021.103383
  • Saalbach A, Tremel J, Herbert D, et al. Anti-inflammatory action of keratinocyte-derived Vaspin: relevance for the pathogenesis of psoriasis. Am J Pathol. 2016;186(3):639–651. doi: 10.1016/j.ajpath.2015.10.030
  • Coban M, Tasli L, Turgut S, et al. Association of adipokines, insulin resistance, hypertension and dyslipidemia in patients with psoriasis vulgaris. Ann Dermatol. 2016;28(1):74–79. doi: 10.5021/ad.2016.28.1.74
  • Saalbach A, Vester K, Rall K, et al. Vaspin–a link of obesity and psoriasis? Exp Dermatol. 2012;21(4):309–312. doi: 10.1111/j.1600-0625.2012.01460.x
  • Zieger K, Weiner J, Krause K, et al. Vaspin suppresses cytokine-induced inflammation in 3T3-L1 adipocytes via inhibition of NFκB pathway. Mol Cell Endocrinol. 2018;460:181–188. doi: 10.1016/j.mce.2017.07.022
  • Zhang GZ, Zhang K, Yang SQ, et al. VASPIN reduces inflammation and endoplasmic reticulum stress of renal tubular epithelial cells by inhibiting HMGB1 and relieves renal ischemia-reperfusion injury. Eur Rev Med Pharmacol Sci. 2020;24(17):8968–8977. doi: 10.26355/eurrev_202009_22839
  • Chen J, Fu Y, Xiong S. Keratinocyte derived HMGB1 aggravates psoriasis dermatitis via facilitating inflammatory polarization of macrophages and hyperproliferation of keratinocyte. Mol Immunol. 2023;163:1–12. doi: 10.1016/j.molimm.2023.09.004
  • Wang Z, Zhou H, Zheng H, et al. Autophagy-based unconventional secretion of HMGB1 by keratinocytes plays a pivotal role in psoriatic skin inflammation. Autophagy. 2021;17(2):529–552. doi: 10.1080/15548627.2020.1725381
  • Bao JP, Xu LH, Ran JS, et al. Vaspin prevents leptin‑induced inflammation and catabolism by inhibiting the activation of nuclear factor‑κB in rat chondrocytes. Mol Med Rep. 2017;16(3):2925–2930. doi: 10.3892/mmr.2017.6911
  • Cerman AA, Bozkurt S, Sav A, et al. Serum leptin levels, skin leptin and leptin receptor expression in psoriasis. Br J Dermatol. 2008;159(4):820–826. doi: 10.1111/j.1365-2133.2008.08742.x
  • Ke X, Chen X, Yan L, et al. Vaspin contributes to autophagy and endothelial-to-mesenchymal transition via PI3K-/AKT-mTOR pathway. Acta Histochem. 2022;124(4):151881. doi: 10.1016/j.acthis.2022.151881
  • Li X, Ke X, Li Z, et al. Vaspin prevents myocardial injury in rats model of diabetic cardiomyopathy by enhancing autophagy and inhibiting inflammation. Biochem Biophys Res Commun. 2019;514(1):1–8. doi: 10.1016/j.bbrc.2019.04.110
  • Xia J, Zhang Y, Wang Q, et al. Cycloastragenol restrains keratinocyte hyperproliferation by promoting autophagy via the miR-145/STC1/Notch1 axis in psoriasis. Immunopharmacol Immunotoxicol. 2024;46(2):229–239. doi: 10.1080/08923973.2023.2300310
  • Wang J, Kaplan N, Wang S, et al. Autophagy plays a positive role in induction of epidermal proliferation. Faseb J. 2020;34(8):10657–10667. doi: 10.1096/fj.202000770RR
  • Colak S, Omma A, Sandikci SC, et al. Vaspin, neutrophil gelatinase-associated lipocalin and apolipoprotein levels in patients with psoriatic arthritis. Bratisl Lek Listy. 2019;120(1):65–69. doi: 10.4149/BLL_2019_010
  • Pedersen OK. Fetuin, a New Globulin Isolated from Serum. Nature. 1944;154(3914):575. doi: 10.1038/154575a0
  • Olmaz R, Selen T, Gungor O. Vascular calcification inhibitors and cardiovascular events in peritoneal dialysis patients. Ther Apher Dial. 2024;28(2):169–181. doi: 10.1111/1744-9987.14091
  • Pérez-Sotelo D, Roca-Rivada A, Larrosa-García M, et al. Visceral and subcutaneous adipose tissue express and secrete functional alpha2hsglycoprotein (fetuin a) especially in obesity. Endocrine. 2017;55(2):435–446. doi: 10.1007/s12020-016-1132-1
  • Pal D, Dasgupta S, Kundu R, et al. Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance. Nat Med. 2012;18(8):1279–1285. doi: 10.1038/nm.2851
  • Ren Q, Liu Z, Wu L, et al. C/EBPβ: The structure, regulation, and its roles in inflammation-related diseases. Biomed Pharmacother. 2023;169:115938. doi: 10.1016/j.biopha.2023.115938
  • Wang H, Sama AE. Anti-inflammatory role of fetuin-A in injury and infection. Curr Mol Med. 2012;12(5):625–633. doi: 10.2174/156652412800620039
  • Shehata WA, Basha MA, Gayed IM, et al. Relationship between disease severity and fetuin-A levels in patients with psoriasis. Indian J Dermatol Venereol Leprol. 2020;86(5):586–588. doi: 10.4103/ijdvl.IJDVL_355_19
  • Borsky P, Fiala Z, Andrys C, et al. C-reactive protein, chemerin, fetuin-A and osteopontin as predictors of cardiovascular risks in persons with psoriasis vulgaris. Physiol Res. 2021;70(3):383–391. doi: 10.33549/physiolres.934654
  • Genc M, Can M, Guven B, et al. Evaluation of serum Fetuin-A and Osteoprotegerin Levels in patients with psoriasis. Indian J Clin Biochem. 2017;32(1):90–94. doi: 10.1007/s12291-016-0570-0
  • Przepiera-Będzak H, Fischer K, Brzosko M. Serum Interleukin-18, Fetuin-A, soluble intercellular adhesion molecule-1, and Endothelin-1 in Ankylosing Spondylitis, Psoriatic Arthritis, and SAPHO Syndrome. Int J Mol Sci. 2016;17(8):1255. doi: 10.3390/ijms17081255
  • Demirbaş A, Kurtipek GS, Tunçez A, et al. The role of cystatin-C and fetuin-A in the determination of early atherosclerotic risk in psoriasis patients. Dermatol Ther. 2020;33(6):e13898. doi: 10.1111/dth.13898
  • Mohany KM, Elkady S, Youssef EMK, et al. Pigment epithelium-derived factor (PEDF) represses the glucose transporter 1 (GLUT1) mRNA expression and may be a potential therapeutic agent in psoriasis: a case-control and experimental study. Sci Rep. 2023;13(1):21424. doi: 10.1038/s41598-023-48565-9
  • Okan G, Baki AM, Yorulmaz E, et al. Serum Visfatin, Fetuin-A, and Pentraxin 3 levels in patients with psoriasis and their relation to disease severity. Clin Lab Anal. 2016;30(4):284–289. doi: 10.1002/jcla.21850
  • Uysal S, Yılmaz FM, Karatoprak K, et al. The levels of serum pentraxin3, CRP, fetuin-A, and insulin in patients with psoriasis. Eur Rev Med Pharmacol Sci. 2014;18(22):3453–3458.
  • Ix JH, Wassel CL, Kanaya AM, et al. Fetuin-A and incident diabetes mellitus in older persons. JAMA. 2008;300(2):182–188. doi: 10.1001/jama.300.2.182
  • Gelfand JM, Shin DB, Armstrong AW, et al. Association of Apremilast with vascular inflammation and cardiometabolic function in patients with psoriasis: the VIP-A Phase 4, open-label, nonrandomized clinical trial. JAMA Dermatol. 2022;158(12):1394–1403. doi: 10.1001/jamadermatol.2022.3862
  • Birukov A, Polemiti E, Jäger S, et al. Fetuin-A and risk of diabetes-related vascular complications: a prospective study. Cardiovasc Diabetol. 2022;21(1):6. doi: 10.1186/s12933-021-01439-8
  • Gelfand JM, Shin DB, Duffin KC, et al. A randomized placebo-controlled trial of Secukinumab on aortic vascular inflammation in moderate-to-severe plaque psoriasis (VIP-S). J Invest Dermatol. 2020;140(9):1784–93.e2. doi: 10.1016/j.jid.2020.01.025
  • Kanai M, Raz A, Goodman DS. Retinol-binding protein: the transport protein for vitamin a in human plasma. J Clin Invest. 1968;47(9):2025–2044. doi: 10.1172/JCI105889
  • Nono Nankam PA, Blüher M. Retinol-binding protein 4 in obesity and metabolic dysfunctions. Mol Cell Endocrinol. 2021;531:111312. doi: 10.1016/j.mce.2021.111312
  • Fan J, Hu J. Retinol binding protein 4 and type 2 diabetes: from insulin resistance to pancreatic β-cell function. Endocrine. 2024. doi: 10.1007/s12020-024-03777-5
  • Li X, Zhu S, Song G, et al. Retinol-binding protein 4 is closely correlated to blood pressure level and E/A in untreated essential hypertension patients. Ann Palliat Med. 2019;8(5):645–650. doi: 10.21037/apm.2019.11.07
  • Gao G, Cui Y, Cheng H. Association between retinol binding protein-4 and psoriasis vulgaris: a systematic review and meta-analysis. Front Med. 2023;10:1208969. doi: 10.3389/fmed.2023.1208969
  • Capo A, Di Nicola M, Costantini E, et al. Circulating levels of Apelin-36 in patients with mild to moderate psoriasis. G Ital Dermatol Venereol. 2020;155(5):646–651. doi: 10.23736/S0392-0488.18.05981-3
  • Baran A, Swiderska M, Flisiak I. The effect of topical treatment and psoriasis severity on serum retinol-binding protein-4 levels. J Dermatol Treat. 2016;27(2):114–119. doi: 10.3109/09546634.2015.1079300
  • Wang HM, Wu C, Jiang YY, et al. Retinol and vitamin a metabolites accumulate through RBP4 and STRA6 changes in a psoriasis murine model. Nutr Metab. 2020;17(1):5. doi: 10.1186/s12986-019-0423-y
  • Kjeldsen L, Johnsen AH, Sengeløv H, et al. Isolation and primary structure of NGAL, a novel protein associated with human neutrophil gelatinase. J Biol Chem. 1993;268(14):10425–10432. doi: 10.1016/S0021-9258(18)82217-7
  • Wang D, Fang L, Pan G. Association of serum lipocalin-2 concentrations with psoriasis and psoriatic arthritis: an updated meta-analysis. Dis Markers. 2019;2019:7361826. doi: 10.1155/2019/7361826
  • Nguyen CTH, Nguyen OPT. Increased plasma lipocalin-2 levels correlate with disease severity and may be a marker of acute inflammatory response in patients with psoriasis. Dermatol Rep. 2022;14(4):9469. doi: 10.4081/dr.2022.9469
  • Baran A, Świderska M, Myśliwiec H, et al. Effect of psoriasis activity and topical treatment on serum lipocalin-2 levels. J Dermatol Treat. 2017;28(2):136–140. doi: 10.1080/09546634.2016.1180340
  • Abdel Hay R, Samir N, Safwat M, et al. Tissue lipocalin-2 in psoriasis: is it a marker of metabolic disturbance or a possible marker of therapeutic efficacy after narrow band ultraviolet B? J Dermatol Treat. 2020;31(5):519–523. doi: 10.1080/09546634.2019.1605141
  • El-Hadidi H, Samir N, Shaker OG, et al. Estimation of tissue and serum lipocalin-2 in psoriasis vulgaris and its relation to metabolic syndrome. Arch Dermatol Res. 2014;306(3):239–245. doi: 10.1007/s00403-013-1414-x
  • Hau CS, Kanda N, Tada Y, et al. Lipocalin-2 exacerbates psoriasiform skin inflammation by augmenting T-helper 17 response. J Dermatol. 2016;43(7):785–794. doi: 10.1111/1346-8138.13227
  • Shao S, Cao T, Jin L, et al. Increased Lipocalin-2 contributes to the pathogenesis of psoriasis by modulating neutrophil chemotaxis and cytokine secretion. J Invest Dermatol. 2016;136(7):1418–1428. doi: 10.1016/j.jid.2016.03.002

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.