516
Views
7
CrossRef citations to date
0
Altmetric
Review

Non-Alcoholic Fatty Liver Disease (NAFLD) in Patients with Psoriasis: A Review of the Hepatic Effects of Systemic Therapies

, &
Pages 151-168 | Published online: 07 Dec 2021

References

  • Armstrong AW, Mehta MD, Schupp CW, et al. Psoriasis prevalence in adults in the United States. JAMA Dermatol. 2021;157(8):940–946. doi:10.1001/jamadermatol.2021.2007
  • Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370:263–271. doi:10.1016/S0140-6736(07)61128-3
  • Balato N, Megna M, Palmisano F, et al. Psoriasis and sport: a new ally? J Eur Acad Dermatol Venereol. 2015;29(3):515–520. doi:10.1111/jdv.12607
  • Gisondi P, Bellinato F, Girolomoni G, et al. Pathogenesis of chronic plaque psoriasis and its intersection with cardio-metabolic comorbidities. Front Pharmacol. 2020;11:117. doi:10.3389/fphar.2020.00117
  • Prussick RB, Miele L. Non-alcoholic fatty liver disease in patients with psoriasis: a consequence of systemic inflammatory burden? Br J Dermatol. 2018;179(1):16–29. doi:10.1111/bjd.16239
  • Lønnberg AS, Skov L. Co-morbidity in psoriasis: mechanisms and implications for treatment. Expert Rev Clin Immunol. 2017;13:27–34. doi:10.1080/1744666X.2016.1213631
  • Elmets CA, Leonardi CL, Davis DMR, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with awareness and attention to comorbidities. J Am Acad Dermatol. 2019;80:1073–1113. doi:10.1016/j.jaad.2018.11.058
  • Sheka AC, Adeyi O, Thompson J, et al. Nonalcoholic steatohepatitis: a review. JAMA. 2020;323(12):1175–1183. doi:10.1001/jama.2020.2298
  • Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther. 2011;34(3):274–285. doi:10.1111/j.1365-2036.2011.04724.x
  • Younossi ZM. The epidemiology of nonalcoholic steatohepatitis. Clin Liver Dis (Hoboken). 2018;11(4):92–94. doi:10.1002/cld.710
  • Belinchón-Romero I, Bellot P, Romero-Pérez D, et al. Non-alcoholic fatty liver disease is associated with bacterial translocation and a higher inflammation response in psoriatic patients. Sci Rep. 2021;11(1):8593. doi:10.1038/s41598-021-88043-8
  • Rinella ME. Nonalcoholic fatty liver disease: a systematic review. JAMA. 2015;313(22):2263–2273. doi:10.1001/jama.2015.5370
  • Adams LA, Lymp JF, St Sauver J, et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology. 2005;129(1):113–121. doi:10.1053/j.gastro.2005.04.014
  • Harrison SA, Di Bisceglie AM. Advances in the understanding and treatment of nonalcoholic fatty liver disease. Drugs. 2003;63(22):2379–2394. doi:10.2165/00003495-200363220-00001
  • Magdaleno-Tapial J, Valenzuela-Oñate C, Ortiz-Salvador JM, et al. Prevalence of non-alcoholic fatty liver and liver fibrosis in patients with moderate-severe psoriasis: a cross-sectional cohort study. Australas J Dermatol. 2020;61(2):105–109. doi:10.1111/ajd.13175
  • Ortolan A, Lorenzin M, Tadiotto G, et al. Metabolic syndrome, non-alcoholic fatty liver disease and liver stiffness in psoriatic arthritis and psoriasis patients. Clin Rheumatol. 2019;38(10):2843–2850. doi:10.1007/s10067-019-04646-7
  • Olveira A, Herranz P, Montes ML. Psoriasis and fatty liver: a harmful synergy. Rev Esp Enferm Dig. 2019;111(4):314–319. doi:10.17235/reed.2019.6263/2019
  • Hamaguchi M, Kojima T, Takeda N, et al. The metabolic syndrome as a predictor of nonalcoholic fatty liver disease. Ann Intern Med. 2005;143(10):722–728. doi:10.7326/0003-4819-143-10-200511150-00009
  • Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American Gastroenterological Association, American Association for the Study of Liver Diseases, and American College of Gastroenterology. Gastroenterology. 2012;142(7):1592–1609. doi:10.1053/j.gastro.2012.04.001
  • Golabi P, Otgonsuren M, de Avila L, et al. Components of metabolic syndrome increase the risk of mortality in nonalcoholic fatty liver disease (NAFLD). Medicine (Baltimore). 2018;97(13):e0214. doi:10.1097/MD.0000000000010214
  • Takaki A, Kawai D, Yamamoto K. Multiple hits, including oxidative stress, as pathogenesis and treatment target in non-alcoholic steatohepatitis (NASH). Int J Mol Sci. 2013;14(10):20704–20728. doi:10.3390/ijms141020704
  • Gisondi P, Galvan A, Idolazzi L, et al. Management of moderate to severe psoriasis in patients with metabolic comorbidities. Front Med (Lausanne). 2015;2:1. doi:10.3389/fmed.2015.00001
  • Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, et al. Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis. Gastroenterology. 2015;149(2):367–378. doi:10.1053/j.gastro.2015.04.005
  • Lonardo A, Loria P, Carulli N. Concurrent non-alcoholic steatohepatitis and psoriasis. Report of three cases from the POLI.ST.E.N.A. study. Dig Liver Dis. 2001;33(1):86–87. doi:10.1016/S1590-8658(01)80144-4
  • Gisondi P, Targher G, Zoppini G, et al. Non-alcoholic fatty liver disease in patients with chronic plaque psoriasis. J Hepatol. 2009;51(4):758–764. doi:10.1016/j.jhep.2009.04.020
  • Gisondi P, Barba E, Girolomoni G. Non-alcoholic fatty liver disease fibrosis score in patients with psoriasis. J Eur Acad Dermatol Venereol. 2016;30(2):282–287. doi:10.1111/jdv.13456
  • van der Voort EA, Koehler EM, Dowlatshahi EA, et al. Psoriasis is independently associated with nonalcoholic fatty liver disease in patients 55 years old or older: results from a population-based study. J Am Acad Dermatol. 2014;70(3):517–524. doi:10.1016/j.jaad.2013.10.044
  • Madanagobalane S, Anandan S. The increased prevalence of non-alcoholic fatty liver disease in psoriatic patients: a study from South India. Australas J Dermatol. 2012;53(3):190–197. doi:10.1111/j.1440-0960.2012.00905.x
  • Abedini R, Salehi M, Lajevardi V, et al. Patients with psoriasis are at a higher risk of developing nonalcoholic fatty liver disease. Clin Exp Dermatol. 2015;40(7):722–727. doi:10.1111/ced.12672
  • Ogdie A, Grewal SK, Noe MH, et al. Risk of incident liver disease in patients with psoriasis, psoriatic arthritis, and rheumatoid arthritis: a population-based study. J Invest Dermatol. 2018;138(4):760–767. doi:10.1016/j.jid.2017.10.024
  • Candia R, Ruiz A, Torres-Robles R, et al. Risk of non-alcoholic fatty liver disease in patients with psoriasis: a systematic review and meta-analysis. J Eur Acad Dermatol Venereol. 2015;29(4):656–662. doi:10.1111/jdv.12847
  • Phan K, Onggo J, Charlton O, et al. Relationship between psoriasis and non-alcoholic fatty liver disease - Updated systematic review and adjusted meta-analysis. Australas J Dermatol. 2019;60(4):e352–e355. doi:10.1111/ajd.13015
  • Yang YW, Keller JJ, Lin HC. Medical comorbidity associated with psoriasis in adults: a population-based study. Br J Dermatol. 2011;165(5):1037–1043. doi:10.1111/j.1365-2133.2011.10494.x
  • Gandha N, Wibawa LP, Jacoeb TNA, et al. Correlation between psoriasis severity and nonalcoholic fatty liver disease degree measured using controlled attenuation parameter. Psoriasis (Auckl). 2020;10:39–44. doi:10.2147/PTT.S272286
  • Roberts KK, Cochet AE, Lamb PB, et al. The prevalence of NAFLD and NASH among patients with psoriasis in a tertiary care dermatology and rheumatology clinic. Aliment Pharmacol Ther. 2015;41(3):293–300. doi:10.1111/apt.13042
  • Romero-Pérez D, Belinchón-Romero I, Bellot P, et al. Nonalcoholic fatty liver disease puts patients with psoriasis at greater cardiovascular risk. Australas J Dermatol. 2019;60:e304–e310. doi:10.1111/ajd.13098
  • National Guideline Centre (UK). Non-alcoholic fatty liver disease: assessment and management. London: National Institute for Health and Care Excellence (UK); 2016. (NICE Guideline, No. 49.) 5, Risk factors for NAFLD. Available from: https://www.ncbi.nlm.nih.gov/books/NBK384735. Accessed November 10, 2021.
  • Friedman SL, Neuschwander-Tetri BA, Rinella M, et al. Mechanisms of NAFLD development and therapeutic strategies. Nat Med. 2018;24(7):908–922. doi:10.1038/s41591-018-0104-9
  • Kamiya K, Kishimoto M, Sugai J, et al. Risk factors for the development of psoriasis. Int J Mol Sci. 2019;20:4347. doi:10.3390/ijms20184347
  • Ganzetti G, Campanati A, Molinelli E, et al. Psoriasis, non-alcoholic fatty liver disease, and cardiovascular disease: three different diseases on a unique background. World J Cardiol. 2016;8(2):120–131. doi:10.4330/wjc.v8.i2.120
  • Grželj J, Mlinarič-Raščan I, Marko PB, et al. Polymorphisms in GNMT and DNMT3b are associated with methotrexate treatment outcome in plaque psoriasis. Biomed Pharmacother. 2021;138:111456. doi:10.1016/j.biopha.2021.111456
  • Heitmann J, Frings VG, Geier A, et al. Non-alcoholic fatty liver disease and psoriasis - is there a shared proinflammatory network? J Dtsch Dermatol Ges. 2021;19(4):517–528.
  • Harden JL, Krueger JG, Bowcock AM. The immunogenetics of psoriasis: a comprehensive review. J Autoimmun. 2015;64:66–73. doi:10.1016/j.jaut.2015.07.008
  • Liang Y, Sarkar MK, Tsoi LC, et al. Psoriasis: a mixed autoimmune and autoinflammatory disease. Curr Opin Immunol. 2017;49:1–8. doi:10.1016/j.coi.2017.07.007
  • Rendon A, Schäkel K. Psoriasis pathogenesis and treatment. Int J Mol Sci. 2019;20(6):E1475. doi:10.3390/ijms20061475
  • 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
  • Lowes MA, Suárez-Fariñas M, Krueger JG. Immunology of psoriasis. Annu Rev Immunol. 2014;32:227–255. doi:10.1146/annurev-immunol-032713-120225
  • Mantovani A, Gisondi P, Lonardo A, et al. Relationship between non-alcoholic fatty liver disease and psoriasis: a novel hepato-dermal axis? Int J Mol Sci. 2016;17(2):217. doi:10.3390/ijms17020217
  • Hawkes JE, Yan BY, Chan TC, et al. Discovery of the IL-23/IL-17 signaling pathway and the treatment of psoriasis. J Immunol. 2018;201(6):1605–1613. doi:10.4049/jimmunol.1800013
  • Ruiz de Morales JMG, Puig L, Daudén E, et al. Critical role of interleukin (IL)-17 in inflammatory and immune disorders: an updated review of the evidence focusing on controversies. Autoimmun Rev. 2020;19(1):102429. doi:10.1016/j.autrev.2019.102429
  • Streba LA, Vere CC, Rogoveanu I, et al. Nonalcoholic fatty liver disease, metabolic risk factors, and hepatocellular carcinoma: an open question. World J Gastroenterol. 2015;21(14):4103–4110. doi:10.3748/wjg.v21.i14.4103
  • Carr RM, Oranu A, Khungar V. Nonalcoholic fatty liver disease: pathophysiology and management. Gastroenterol Clin North Am. 2016;45(4):639–652. doi:10.1016/j.gtc.2016.07.003
  • Anstee QM, Daly AK, Day CP. Genetic modifiers of non-alcoholic fatty liver disease progression. Biochim Biophys Acta. 2011;1812(11):1557–1566. doi:10.1016/j.bbadis.2011.07.017
  • Eslam M, Valenti L, Romeo S. Genetics and epigenetics of NAFLD and NASH: clinical impact. J Hepatol. 2018;68(2):268–279. doi:10.1016/j.jhep.2017.09.003
  • Yousaf A, Raiker R, Davis SM, et al. Association between psoriasis, psoriatic arthritis and gastrointestinal disease. Wien Klin Wochenschr. 2021;133:586–593. doi:10.1007/s00508-020-01740-8
  • Angulo P, Lindor KD. Non-alcoholic fatty liver disease. J Gastroenterol Hepatol. 2002;17(Suppl):S186–190. doi:10.1046/j.1440-1746.17.s1.10.x
  • Ganzetti G, Campanati A, Offidani A. Non-alcoholic fatty liver disease and psoriasis: so far, so near. World J Hepatol. 2015;7(3):315–326. doi:10.4254/wjh.v7.i3.315
  • Young S, Tariq R, Provenza J, et al. Prevalence and profile of nonalcoholic fatty liver disease in lean adults: systematic review and meta-analysis. Hepatol Commun. 2020;4(7):953–972. doi:10.1002/hep4.1519
  • Meli R, Mattace Raso G, Calignano A. Role of innate immune response in non-alcoholic fatty liver disease: metabolic complications and therapeutic tools. Front Immunol. 2014;5:177. doi:10.3389/fimmu.2014.00177
  • Jarrar M, Baranova A, Collantes R, et al. Adipokines and cytokines in non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2008;27:412–421. doi:10.1111/j.1365-2036.2007.03586.x
  • Carrascosa JM, Bonanad C, Dauden E, et al. Psoriasis and nonalcoholic fatty liver disease. Actas Dermosifiliogr. 2017;108(6):506–514. doi:10.1016/j.ad.2016.12.017
  • Martin DA, Towne JE, Kricorian G, et al. The emerging role of IL-17 in the pathogenesis of psoriasis: preclinical and clinical findings. J Invest Dermatol. 2013;133:17–26. doi:10.1038/jid.2012.194
  • Theocharidou E, Papademetriou M, Reklou A, et al. The role of PCSK9 in the pathogenesis of non-alcoholic fatty liver disease and the effect of PCSK9 inhibitors. Curr Pharm Des. 2018;24(31):3654–3657. doi:10.2174/1381612824666181010123127
  • Luan C, Chen X, Zhu Y, et al. Potentiation of psoriasis-like inflammation by PCSK9. J Invest Dermatol. 2019;139(4):859–867. doi:10.1016/j.jid.2018.07.046
  • Piaserico S, Messina F, Russo FP. Managing psoriasis in patients with HBV or HCV infection: practical considerations. Am J Clin Dermatol. 2019;20:829–845. doi:10.1007/s40257-019-00457-3
  • Taylor WJ, Korendowych E, Nash P, et al. Drug use and toxicity in psoriatic disease: focus on methotrexate. J Rheumatol. 2008;35:1454–1457.
  • Shetty A, Cho W, Alazawi W, et al. Methotrexate hepatotoxicity and the impact of nonalcoholic fatty liver disease. Am J Med Sci. 2017;354(2):172–181. doi:10.1016/j.amjms.2017.03.014
  • Nast A, Gisondi P, Ormerod AD, et al. European S3-guidelines on the systemic treatment of psoriasis vulgaris—update 2015—short version—EDF in cooperation with EADV and IPC. J Eur Acad Dermatol Venereol. 2015;29(12):2277–2294. doi:10.1111/jdv.13354
  • Balak DMW, Gerdes S, Parodi A, et al. Long-term safety of oral systemic therapies for psoriasis: a comprehensive review of the literature. Dermatol Ther (Heidelb). 2020;10(4):589–613. doi:10.1007/s13555-020-00409-4
  • Gelfand JM, Wan J, Zhang H, et al. Risk of liver disease in patients with psoriasis, psoriatic arthritis, and rheumatoid arthritis receiving methotrexate: a population-based study. J Am Acad Dermatol. 2021;84(6):1636–1643. doi:10.1016/j.jaad.2021.02.019
  • Bath RK, Brar NK, Forouhar FA, et al. A review of methotrexate-associated hepatotoxicity. J Dig Dis. 2014;15(10):517–524. doi:10.1111/1751-2980.12184
  • Ezhilarasan D. Hepatotoxic potentials of methotrexate: understanding the possible toxicological molecular mechanisms. Toxicology. 2021;458:152840. doi:10.1016/j.tox.2021.152840
  • European Medicines Agency. Summary of product characteristics. Nordimet (methotrexate) 7.5–25mg solution for injection in pre-filled pen. Available from: https://www.ema.europa.eu/en/documents/product-information/nordimet-epar-product-information_en.pdf. Accessed November 10, 2021.
  • Electronic Medicines Compendium (EMC). Sandimmun concentrate for solution for infusion 50 mg/mL; 2020. Available from: https://www.medicines.org.uk/emc/product/1036/smpc. Accessed November 10, 2021.
  • Prussick R, Prussick L, Nussbaum D. Nonalcoholic fatty liver disease and psoriasis: what a dermatologist needs to know. J Clin Aesthet Dermatol. 2015;8(3):43–45.
  • European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO). EASL-EASD-EASO clinical practice guidelines for the management of non-alcoholic fatty liver disease. J Hepatol. 2016;64(6):1388–1402. doi:10.1016/j.jhep.2015.11.004
  • Electronic Medicines Compendium (EMC). Skilarence 30 mg gastro-resistant tablets; 2020. Available from: www.medicines.org.uk/emc/product/752/smpc#gref. Accessed November 10, 2021.
  • LiverTox: clinical and research information on drug-induced liver injury [Internet]. Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases. Available from: https://europepmc.org/article/NBK/NBK548013. Accessed November 10, 2021.
  • Roenigk HH, Callen JP, Guzzo CA, et al. Effects of Acitretin on the liver. J Am Acad Dermatol. 1999;41(4):584–588.
  • Dunn LK, Gaar LR, Yentzer BA, et al. Acitretin in dermatology: a review. J Drugs Dermatol. 2011;10:772–782.
  • Ghabril M, Bonkovsky HL, Kum C, et al. Liver injury from tumor necrosis factor-alpha antagonists: analysis of thirty-four cases. Clin Gastroenterol Hepatol. 2013;11:558–564.
  • Rossi RE, Parisi I, Despott EJ, et al. Anti-tumour necrosis factor agent and liver injury: literature review, recommendations for management. World J Gastroenterol. 2014;20(46):17352–17359. doi:10.3748/wjg.v20.i46.17352
  • Mikhaylov D, Hashim PW, Nektalova T, et al. Systemic psoriasis therapies and comorbid disease in patients with psoriasis: a review of potential risks and benefits. J Clin Aesthet Dermatol. 2019;12(6):46–54.
  • Wu MY, Yu CL, Yang SJ, et al. Change in body weight and body mass index in psoriasis patients receiving biologics: a systematic review and network meta-analysis. J Am Acad Dermatol. 2020;82(1):101–109. doi:10.1016/j.jaad.2019.07.103
  • Llamas-Velasco M, Concha-Garzon MJ, Garcia-Diez A, et al. Liver injury in psoriasis patients receiving ustekinumab: a retrospective study of 44 patients treated in the clinical practice setting. Actas Dermosifiliogr. 2015;106(6):470–476. doi:10.1016/j.ad.2015.02.002
  • Gebreselassie A, Aduli F, Howell CD. Rheumatologic diseases and the liver. Clin Liver Dis. 2019;23(2):247–261. doi:10.1016/j.cld.2018.12.007
  • Polat Ekinci A, Bölük KN, Babuna Kobaner G. Secukinumab and Acitretin as a combination therapy for three clinical forms of severe psoriasis in multi-drug refractory patients: a case series of high efficacy and safety profile. Dermatol Ther. 2021;34(1):e14704. doi:10.1111/dth.14704
  • Gisondi P, Conti A, Galdo G, et al. Ustekinumab does not increase body mass index in patients with chronic plaque psoriasis: a prospective cohort study. Br J Dermatol. 2013;168(5):1124–1127. doi:10.1111/bjd.12235
  • Krahel JA, Baran A, Kamiński TW, et al. Methotrexate decreases the level of PCSK9-a novel indicator of the risk of proatherogenic lipid profile in psoriasis. the preliminary data. J Clin Med. 2020;9(4):910. doi:10.3390/jcm9040910
  • Kiluk P, Baran A, Kaminski TW, et al. The level of FGF 21 as a new risk factor for the occurrence of cardiometabolic disorders amongst the psoriatic patients. J Clin Med. 2019;8(12):2206. doi:10.3390/jcm8122206
  • Mangoni AA, Zinellu A, Sotgia S, et al. Protective effects of methotrexate against proatherosclerotic cytokines: a review of the evidence. Mediators Inflamm. 2017;2017:9632846. doi:10.1155/2017/9632846
  • Mangoni AA, Tommasi S, Zinellu A, et al. Methotrexate and vasculoprotection: mechanistic insights and potential therapeutic applications in old age. Curr Pharm Des. 2019;25(39):4175–4184. doi:10.2174/1381612825666191112091700
  • Michalska-Bańkowska A, Grabarek B, Wcisło-Dziadecka D, et al. The impact of diabetes and metabolic syndromes to the effectiveness of cyclosporine a pharmacotherapy in psoriatic patients. Dermatol Ther. 2019;32(3):e12881. doi:10.1111/dth.12881
  • Mazzilli S, Lanna C, Chiaramonte C, et al. Real life experience of apremilast in psoriasis and arthritis psoriatic patients: preliminary results on metabolic biomarkers. J Dermatol. 2020;47(6):578–582. doi:10.1111/1346-8138.15293
  • Mehta NN, Shin DB, Joshi AA, et al. Effect of 2 psoriasis treatments on vascular inflammation and novel inflammatory cardiovascular biomarkers: a randomized placebo-controlled trial. Circ Cardiovasc Imaging. 2018;11(6):e007394. doi:10.1161/CIRCIMAGING.117.007394
  • Holzer G, Hoke M, Sabeti-Sandor S, et al. Disparate effects of Adalimumab and fumaric acid esters on cardiovascular risk factors in psoriasis patients: results from a prospective, randomized, observer-blinded head-to-head trial. J Eur Acad Dermatol Venereol. 2021;35(2):441–449. doi:10.1111/jdv.16635
  • 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):1784–1793. doi:10.1016/j.jid.2020.01.025
  • Gerdes S, Pinter A, Papavassilis C, et al. Effects of secukinumab on metabolic and liver parameters in plaque psoriasis patients. J Eur Acad Dermatol Venereol. 2020;34(3):533–541. doi:10.1111/jdv.16004
  • Ikumi K, Odanaka M, Shime H, et al. Hyperglycemia is associated with psoriatic inflammation in both humans and mice. J Invest Dermatol. 2019;139(6):1329–1338. doi:10.1016/j.jid.2019.01.029
  • Makavos G, Ikonomidis I, Andreadou I, et al. Effects of interleukin 17A inhibition on myocardial deformation and vascular function in psoriasis. Can J Cardiol. 2020;36(1):100–111. doi:10.1016/j.cjca.2019.06.021
  • Gelfand JM, Shin DB, Alavi A, et al. A phase IV, randomized, double-blind, placebo-controlled crossover study of the effects of ustekinumab on vascular inflammation in psoriasis (the VIP-U trial). J Invest Dermatol. 2020;140(1):85–93. doi:10.1016/j.jid.2019.07.679
  • Zhu K, Mrowietz U. Inhibition of dendritic cell differentiation by fumaric acid esters. J Invest Dermatol. 2001;116(2):203–208. doi:10.1046/j.1523-1747.2001.01159.x
  • Peng H, Guerau-de-arellano M, Mehta VB, et al. Dimethyl fumarate inhibits dendritic cell maturation via nuclear factor κB (NF-κB) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) and mitogen stress-activated kinase 1 (MSK1) signaling. J Biol Chem. 2012;287(33):28017–28026. doi:10.1074/jbc.M112.383380
  • Takasu C, Vaziri ND, Li S, et al. Treatment with dimethyl fumarate ameliorates liver ischemia/reperfusion injury. World J Gastroenterol. 2017;23(25):4508–4516. doi:10.3748/wjg.v23.i25.4508
  • Kornberg MD, Bhargava P, Kim PM, et al. Dimethyl fumarate targets GAPDH and aerobic glycolysis to modulate immunity. Science. 2018;360(6387):449–453. doi:10.1126/science.aan4665
  • Brück J, Glocova I, Geisel J, et al. Dimethyl fumarate-induced IL-17 low IFN-γ low IL-4 + Th cells protect mice from severe encephalomyelitis. Eur J Immunol. 2018;48(9):1588–1591. doi:10.1002/eji.201747435
  • Abdelrahman RS, Abdel-Rahman N. Dimethyl fumarate ameliorates Acetaminophen-induced hepatic injury in mice dependent of Nrf-2/HO-1 pathway. Life Sci. 2019;217:251–260. doi:10.1016/j.lfs.2018.12.013
  • Dwivedi DK, Jena G, Kumar V. Dimethyl fumarate protects thioacetamide-induced liver damage in rats: studies on Nrf2, NLRP3, and NF-κB. J Biochem Mol Toxicol. 2020;34(6):e22476. doi:10.1002/jbt.22476
  • Korman NJ. Management of psoriasis as a systemic disease: what is the evidence? Br J Dermatol. 2020;182(4):840–848. doi:10.1111/bjd.18245
  • Ridker PM, Everett BM, Pradhan A, et al. Low-dose methotrexate for the prevention of atherosclerotic events. N Engl J Med. 2019;380(8):752–762. doi:10.1056/NEJMoa1809798
  • Karadag AS, Ertugrul DT, Kalkan G, et al. The effect of Acitretin treatment on insulin resistance, retinol-binding protein-4, leptin, and adiponectin in psoriasis vulgaris: a noncontrolled study. Dermatology. 2013;227(2):103–108. doi:10.1159/000351769
  • Michalska-Bańkowska A, Wcisło-Dziadecka D, Grabarek B, et al. Clinical and molecular evaluation of therapy with the use of cyclosporine A in patients with psoriasis vulgaris. Int J Dermatol. 2019;58(4):477–482. doi:10.1111/ijd.14275
  • Ure DR, Trepanier DJ, Mayo PR, et al. Cyclophilin inhibition as a potential treatment for nonalcoholic steatohepatitis (NASH). Expert Opin Investig Drugs. 2020;29(2):163–178. doi:10.1080/13543784.2020.1703948
  • Lanna C, Cesaroni GM, Mazzilli S, et al. Small molecules, big promises: improvement of psoriasis severity and glucidic markers with apremilast: a case report. Diabetes Metab Syndr Obes. 2019;12:2685–2688. doi:10.2147/DMSO.S229549
  • Gualtierotti R, De Lucia O. Efficacy and metabolic effect on serum lipids of apremilast in psoriatic arthritis: a case report. J Clin Med. 2019;8(3):398. doi:10.3390/jcm8030398
  • Brück J, Dringen R, Amasuno A, et al. A review of the mechanisms of action of dimethylfumarate in the treatment of psoriasis. Exp Dermatol. 2018;27(6):611–624. doi:10.1111/exd.13548
  • McGuire VA, Ruiz-Zorrilla Diez T, Emmerich CH, et al. Dimethyl fumarate blocks pro-inflammatory cytokine production via inhibition of TLR induced M1 and K63 ubiquitin chain formation. Sci Rep. 2016;6:31159. doi:10.1038/srep31159
  • Kiortsis DN, Mavridis AK, Vasakos S, et al. Effects of infliximab treatment on insulin resistance in patients with rheumatoid arthritis and ankylosing spondylitis. Ann Rheum Dis. 2005;64(5):765–766. doi:10.1136/ard.2004.026534
  • Gonzalez-Gay MA, De Matias JM, Gonzalez-Juanatey C, et al. Anti-tumor necrosis factor-alpha blockade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp Rheumatol. 2006;24(1):83–86.
  • Miranda-Filloy JA, Llorca J, Carnero-López B, et al. TNF-alpha antagonist therapy improves insulin sensitivity in non-diabetic ankylosing spondylitis patients. Clin Exp Rheumatol. 2012;30(6):850–855.
  • Dominguez H, Storgaard H, Rask-Madsen C, et al. Metabolic and vascular effects of tumor necrosis factor-alpha blockade with etanercept in obese patients with type 2 diabetes. J Vasc Res. 2005;42(6):517–525. doi:10.1159/000088261
  • Bernstein LE, Berry J, Kim S, et al. Effects of etanercept in patients with the metabolic syndrome. Arch Intern Med. 2006;166:902–908. doi:10.1001/archinte.166.8.902
  • Wascher TC, Lindeman JH, Sourij H, et al. Chronic TNF-α neutralization does not improve insulin resistance or endothelial function in “healthy” men with metabolic syndrome. Mol Med. 2011;17(3–4):189–193. doi:10.2119/molmed.2010.00221
  • Seitz M, Reichenbach S, Möller B, et al. Hepatoprotective effect of tumour necrosis factor alpha blockade in psoriatic arthritis: a cross-sectional study. Ann Rheum Dis. 2010;69(6):1148–1150. doi:10.1136/ard.2009.116194