Exploring the links between Obesity and Psoriasis: A comprehensive review

datacite.rightshttp://purl.org/coar/access_right/c_abf2eng
dc.contributor.authorBarros, Gabriela
dc.contributor.authorDuran, Pablo
dc.contributor.authorVera, Ivana
dc.contributor.authorBermúdez, Valmore
dc.date.accessioned2022-11-16T16:38:46Z
dc.date.available2022-11-16T16:38:46Z
dc.date.issued2022
dc.description.abstractObesity is a major public health issue worldwide since it is associated with the development of chronic comorbidities such as type 2 diabetes, dyslipidemias, atherosclerosis, some cancer forms and skin diseases, including psoriasis. Scientific evidence has indicated that the possible link between obesity and psoriasis may be multifactorial, highlighting dietary habits, lifestyle, certain genetic factors and the microbiome as leading factors in the progress of both pathologies because they are associated with a chronic pro-inflammatory state. Thus, inflammation management in obesity is a plausible target for psoriasis, not only because of the sick adipose tissue secretome profile but also due to the relationship of obesity with the rest of the immune derangements associated with psoriasis initiation and maintenance. Hence, this review will provide a general and molecular overview of the relationship between both pathologies and present recent therapeutic advances in treating this problem.eng
dc.format.mimetypepdfspa
dc.identifier.citationBarros, G., Duran, P., Vera, I., & Bermúdez, V. (2022). Exploring the Links between Obesity and Psoriasis: A Comprehensive Review. International Journal of Molecular Sciences, 23(14), 7499. https://doi.org/10.3390/ijms23147499eng
dc.identifier.doihttps://doi.org/10.3390/ijms23147499
dc.identifier.issn14220067
dc.identifier.urihttps://hdl.handle.net/20.500.12442/11331
dc.language.isoengeng
dc.publisherMDPIspa
dc.rightsAttribution-NonCommercial-NoDerivatives 4.0 Internacional*
dc.rights.accessrightsinfo:eu-repo/semantics/openAccesseng
dc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/4.0/*
dc.sourceInternational Journal of Molecular Scienceseng
dc.sourceVol. 23, Issue 14 (2022)eng
dc.subjectadipokineseng
dc.subjectcytokineseng
dc.subjectinflammationeng
dc.subjectObesityeng
dc.subjectPsoriasiseng
dc.subjectmicrobiotaeng
dc.subjectBody Mass Indexeng
dc.titleExploring the links between Obesity and Psoriasis: A comprehensive revieweng
dc.type.driverinfo:eu-repo/semantics/articleeng
dc.type.spaArtículo científicospa
dcterms.referencesJensen, P.; Skov, L. Psoriasis and Obesity. Dermatology 2016, 232, 633–639.eng
dcterms.referencesSu, X.; Cheng, Y.; Chang, D. The Important Role of Leptin in Modulating the Risk of Dermatological Diseases. Front. Immunol. 2021, 11, 593564.eng
dcterms.referencesKanda, N.; Hoashi, T.; Saeki, H. Nutrition and Psoriasis. Int. J. Mol. Sci. 2020, 21, 5405eng
dcterms.referencesBonanad, C.; González-Parra, E.; Rivera, R.; Carrascosa, J.M.; Daudén, E.; Olveira, A.; Botella-Estrada, R. Clinical, Diagnostic, and Therapeutic Implications in Psoriasis Associated With Cardiovascular Disease. Actas Dermosifiliogr. 2017, 108, 800–808.eng
dcterms.referencesTimmins, K.A.; Leech, R.D.; Batt, M.E.; Edwards, K.L. Running and Knee Osteoarthritis: A Systematic Review and Meta-analysis. Am. J. Sports Med. 2017, 45, 1447–1457.eng
dcterms.referencesYe, Q.; Zou, B.; Yeo, Y.H.; Li, J.; Huang, D.Q.; Wu, Y.; Yang, H.; Liu, C.; Kam, L.Y.; Tan, X.X.E.; et al. Global prevalence, incidence, and outcomes of non-obese or lean non-alcoholic fatty liver disease: A systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2020, 5, 739–752.eng
dcterms.referencesLuppino, F.S.; de Wit, L.M.; Bouvy, P.F.; Stijnen, T.; Cuijpers, P.; Penninx, B.W.J.H.; Zitman, F.G. Overweight, obesity, and depression: A systematic review and meta-analysis of longitudinal studies. Arch. Gen. Psychiatry 2010, 67, 220–229eng
dcterms.referencesCortese, S.; Tessari, L. Attention-Deficit/Hyperactivity Disorder (ADHD) and Obesity: Update 2016. Curr. Psychiatry Rep. 2017, 19, 4.eng
dcterms.referencesKelishadi, R.; Roufarshbaf, M.; Soheili, S.; Payghambarzadeh, F.; Masjedi, M. Association of Childhood Obesity and the Immune System: A Systematic Review of Reviews. Child. Obes. 2017, 13, 332–346.eng
dcterms.referencesBlüher, M. Obesity: Global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 2019, 15, 288–298.eng
dcterms.referencesCarretero, J.; Ena, J.; Arévalo, J.C.; Seguí, J.M.; Carrasco, F.J.; Gómez, R.; Pérez, M.I.; Delgado, J.; Pérez, P. Obesity is a chronic disease. Positioning statement of the Diabetes, Obesity and Nutrition Workgroup of the Spanish Society of Internal Medicine (SEMI) for an approach centred on individuals with obesity. Rev. Clínica Esp. 2021, 221, 509–516.eng
dcterms.referencesPan, X.-F.; Wang, L.; Pan, A. Epidemiology and determinants of obesity in China. Lancet Diabetes Endocrinol. 2021, 9, 373–392.eng
dcterms.referencesCDC CDC Overweight & Obesity. Centers for Disease Control and Prevention. 2021. Available online: https://www.cdc.gov/obesity/index.htmleng
dcterms.referencesOrloff, J.; Kaminetsky, J.; Aziz, M. Psoriasis and Obesity: A Review of the Current Literature. SKIN J. Cutan. Med. 2018, 2, 364–379.eng
dcterms.referencesChooi, Y.C.; Ding, C.; Magkos, F. The epidemiology of obesity. Metabolism 2019, 92, 6–10.eng
dcterms.referencesAn, R.; Ji, M.; Zhang, S. Global warming and obesity: A systematic review. Obes. Rev. 2018, 19, 150–163.eng
dcterms.referencesJaacks, L.M.; Vandevijvere, S.; Pan, A.; McGowan, C.J.; Wallace, C.; Imamura, F.; Mozaffarian, D.; Swinburn, B.; Ezzati, M. The obesity transition: Stages of the global epidemic. Lancet Diabetes Endocrinol. 2019, 7, 231–240.eng
dcterms.referencesNga, V.T.; Dung, V.N.T.; Chu, D.-T.; Tien, N.L.B.; Van Thanh, V.; Ngoc, V.T.N.; Hoan, L.N.; Phuong, N.T.; Pham, V.-H.; Tao, Y.; et al. School education and childhood obesity: A systemic review. Diabetes Metab. Syndr. 2019, 13, 2495–2501.eng
dcterms.referencesKoch, C.A.; Sharda, P.; Patel, J.; Gubbi, S.; Bansal, R.; Bartel, M.J. Climate Change and Obesity. Horm. Metab. Res. 2021, 53, 575–587.eng
dcterms.referencesDhurandhar, E.J. The food-insecurity obesity paradox: A resource scarcity hypothesis. Physiol. Behav. 2016, 162, 88–92.eng
dcterms.referencesFigueira, T.C.F.; Corrente, J.E.; Miot, L.D.B.; Papini, S.J.; Miot, H.A. Dietary patterns of patients with psoriasis at a public healthcare institution in Brazil. An. Bras. Dermatol. 2020, 95, 452–458.eng
dcterms.referencesChen, G.; Chen, Z.-M.; Fan, X.-Y.; Jin, Y.-L.; Li, X.; Wu, S.-R.; Ge, W.-W.; Lv, C.-H.; Wang, Y.-K.; Chen, J.-G. Gut-Brain-Skin Axis in Psoriasis: A Review. Derm. Ther. 2021, 11, 25–38.eng
dcterms.referencesLiakou, A.; Zouboulis, C. Links and risks associated with psoriasis and metabolic syndrome. Psoriasis Targets Ther. 2015, 5, 125.eng
dcterms.referencesLangan, S.M.; Seminara, N.M.; Shin, D.B.; Troxel, A.B.; Kimmel, S.E.; Mehta, N.N.; Margolis, D.J.; Gelfand, J.M. Prevalence of Metabolic Syndrome in Patients with Psoriasis: A Population-Based Study in the United Kingdom. J. Investig. Dermatol. 2012, 132, 556–562.eng
dcterms.referencesGisondi, P.; Fostini, A.C.; Fossà, I.; Girolomoni, G.; Targher, G. Psoriasis and the metabolic syndrome. Clin. Dermatol. 2018, 36, 21–28.eng
dcterms.referencesLarabi, A.; Barnich, N.; Nguyen, H.T.T. New insights into the interplay between autophagy, gut microbiota and inflammatory responses in IBD. Autophagy 2020, 16, 38–51.eng
dcterms.referencesSafari, Z.; Gérard, P. The links between the gut microbiome and non-alcoholic fatty liver disease (NAFLD). Cell Mol. Life Sci. 2019, 76, 1541–1558.eng
dcterms.referencesMegur, A.; Baltriukienė, D.; Bukelskienė, V.; Burokas, A. The Microbiota-Gut-Brain Axis and Alzheimer’s Disease: Neuroinflammation Is to Blame? Nutrients 2020, 13, 37.eng
dcterms.referencesMyers, B.; Brownstone, N.; Reddy, V.; Chan, S.; Thibodeaux, Q.; Truong, A.; Bhutani, T.; Chang, H.-W.; Liao, W. The gut microbiome in psoriasis and psoriatic arthritis. Best Pract. Res. Clin. Rheumatol. 2019, 33, 101494.eng
dcterms.referencesVerhaar, B.J.H.; Prodan, A.; Nieuwdorp, M.; Muller, M. Gut Microbiota in Hypertension and Atherosclerosis: A Review. Nutrients 2020, 12, 2982.eng
dcterms.referencesArmstrong, A.W.; Read, C. Pathophysiology, Clinical Presentation, and Treatment of Psoriasis: A Review. JAMA 2020, 323, 1945–1960.eng
dcterms.referencesLiang, Y.; Sarkar, M.K.; Tsoi, L.C.; Gudjonsson, J.E. Psoriasis: A mixed autoimmune and autoinflammatory disease. Curr. Opin. Immunol. 2017, 49, 1–8.eng
dcterms.referencesKunz, M.; Simon, J.C.; Saalbach, A. Psoriasis: Obesity and Fatty Acids. Front. Immunol. 2019, 10, 1807.eng
dcterms.referencesKim, H.-N.; Han, K.; Park, Y.-G.; Lee, J.H. Metabolic syndrome is associated with an increased risk of psoriasis: A nationwide population-based study. Metabolism 2019, 99, 19–24eng
dcterms.referencesKyriakou, A.; Patsatsi, A.; Sotiriadis, D.; Goulis, D.G. Effects of treatment for psoriasis on circulating levels of leptin, adiponectin and resistin: A systematic review and meta-analysis. Br. J. Derm. 2018, 179, 273–281.eng
dcterms.referencesOgawa, K.; Stuart, P.E.; Tsoi, L.C.; Suzuki, K.; Nair, R.P.; Mochizuki, H.; Elder, J.T.; Okada, Y. A Transethnic Mendelian Randomization Study Identifies Causality of Obesity on Risk of Psoriasis. J. Investig. Dermatol. 2019, 139, 1397–1400.eng
dcterms.referencesVerdugo, A. Prevalencia de sobrepeso y obesidad en adultos de la ciudad de Cuenca-Ecuador, 2014. Rev. Fac. Cienc. Méd. Univ. Cuenca 2018, 36, 54–60.eng
dcterms.referencesHirt, P.A.; Castillo, D.E.; Yosipovitch, G.; Keri, J.E. Skin changes in the obese patient. J. Am. Acad. Dermatol. 2019, 81, 1037–1057.eng
dcterms.referencesParoutoglou, K.; Papadavid, E.; Christodoulatos, G.S.; Dalamaga, M. Deciphering the Association Between Psoriasis and Obesity: Current Evidence and Treatment Considerations. Curr. Obes. Rep. 2020, 9, 165–178.eng
dcterms.referencesKong, Y.; Zhang, S.; Wu, R.; Su, X.; Peng, D.; Zhao, M.; Su, Y. New insights into different adipokines in linking the pathophysiology of obesity and psoriasis. Lipids Health Dis. 2019, 18, 171.eng
dcterms.referencesStjernholm, T.; Ommen, P.; Langkilde, A.; Johansen, C.; Iversen, L.; Rosada, C.; Stenderup, K. Leptin deficiency in mice counteracts imiquimod (IMQ)-induced psoriasis-like skin inflammation while leptin stimulation induces inflammation in human keratinocytes. Exp. Derm. 2017, 26, 338–345.eng
dcterms.referencesSnekvik, I.; Smith, C.H.; Nilsen, T.I.L.; Langan, S.M.; Modalsli, E.H.; Romundstad, P.R.; Saunes, M. Obesity, Waist Circumference, Weight Change, and Risk of Incident Psoriasis: Prospective Data from the HUNT Study. J. Investig. Dermatol. 2017, 137, 2484–2490.eng
dcterms.referencesKumar, S.; Han, J.; Li, T.; Qureshi, A.A. Obesity, waist circumference, weight change and the risk of psoriasis in US women: Risk of psoriasis in US women. J. Eur. Acad. Dermatol. Venereol. 2013, 27, 1293–1298.eng
dcterms.referencesKo, S.-H.; Chi, C.-C.; Yeh, M.-L.; Wang, S.-H.; Tsai, Y.-S.; Hsu, M.-Y. Lifestyle changes for treating psoriasis. Cochrane Database Syst. Rev. 2019, 7, 1–68.eng
dcterms.referencesLebwohl, M.G. Psoriasis: Which therapy for which patient. J. Am. Acad. Dermatol. 2019, 80, 27–40.eng
dcterms.referencesDopytalska, K.; Baranowska-Bik, A.; Roszkiewicz, M.; Bik, W.; Walecka, I. The role of leptin in selected skin diseases. Lipids Health Dis. 2020, 19, 215.eng
dcterms.referencesTollefson, M.M.; Crowson, C.S.; McEvoy, M.T.; Maradit Kremers, H. Incidence of psoriasis in children: A population-based study. J. Am. Acad. Dermatol. 2010, 62, 979–987.eng
dcterms.referencesCastaldo, G.; Rastrelli, L.; Galdo, G.; Molettieri, P.; Rotondi Aufiero, F.; Cereda, E. Aggressive weight-loss program with a ketogenic induction phase for the treatment of chronic plaque psoriasis: A proof-of-concept, single-arm, open-label clinical trial. Nutrition 2020, 74, 110757.eng
dcterms.referencesLlamas-Velasco, M.; Ovejero-Merino, E.; Salgado-Boquete, L. Obesidad: Factor de riesgo para psoriasis y COVID-19. Actas Dermo-Sifiliográficas 2020, 112, 489–494.eng
dcterms.referencesTakeshita, J.; Grewal, S.; Langan, S.M.; Mehta, N.N.; Ogdie, A.; Van Voorhees, A.S.; Gelfand, J.M. Psoriasis and comorbid diseases. J. Am. Acad. Dermatol. 2017, 76, 377–390.eng
dcterms.referencesVanderpuye-Orgle, J.; Zhao, Y.; Lu, J.; Shrestha, A.; Sexton, A.; Seabury, S.; Lebwohl, M. Evaluating the economic burden of psoriasis in the United States. J. Am. Acad. Derm. 2015, 72, 961–967.e5.eng
dcterms.referencesParisi, R.; Iskandar, I.Y.K.; Kontopantelis, E.; Augustin, M.; Griffiths, C.E.M.; Ashcroft, D.M. National, regional, and worldwide epidemiology of psoriasis: Systematic analysis and modelling study. BMJ 2020, 369, m1590.eng
dcterms.referencesIcen, M.; Crowson, C.S.; McEvoy, M.T.; Dann, F.J.; Gabriel, S.E.; Maradit Kremers, H. Trends in incidence of adult-onset psoriasis over three decades: A population-based study. J. Am. Acad. Dermatol. 2009, 60, 394–401.eng
dcterms.referencesPrevalence of Psoriasis in Italy 2010–2017. Statista. Available online: https://www.statista.com/statistics/944680/prevalence-of-psoriasis-in-italy/ (accessed on 22 October 2021).eng
dcterms.referencesOrtega, F.B.; Lavie, C.J.; Blair, S.N. Obesity and Cardiovascular Disease. Circ. Res. 2016, 118, 1752–1770.eng
dcterms.referencesXie, W.; Huang, H.; Deng, X.; Gao, D.; Zhang, Z. Modifiable lifestyle and environmental factors associated with onset of psoriatic arthritis in patients with psoriasis: A systematic review and meta-analysis of observational studies. J. Am. Acad. Derm. 2021, 84, 701–711.eng
dcterms.referencesHsu, S.; Green, L.J.; Lebwohl, M.G.; Wu, J.J.; Blauvelt, A.; Jacobson, A.A. Comparable efficacy and safety of brodalumab in obese and nonobese patients with psoriasis: Analysis of two randomized controlled trials. Br. J. Derm. 2020, 182, 880–888.eng
dcterms.referencesNorden, A.; Rekhtman, S.; Strunk, A.; Garg, A. Risk of psoriasis according to body mass index: A retrospective cohort analysis. J. Am. Acad. Dermatol. 2021, 86, 1020–1026.eng
dcterms.referencesKarmacharya, P.; Ogdie, A.; Eder, L. Psoriatic arthritis and the association with cardiometabolic disease: A narrative review. Adv. Musculoskelet. Dis. 2021, 13, 1759720X21998279.eng
dcterms.referencesBavoso, N.C.; Pinto, J.M.; Soares, M.M.S.; dos Santos Diniz, M.; Teixeira Júnior, A.L. Psoriasis in obesity: Comparison of serum levels of leptin and adiponectin in obese subjects—Cases and controls. An. Bras. Dermatol. 2019, 94, 192–197.eng
dcterms.referencesChen, L.; Li, J.; Zhu, W.; Kuang, Y.; Liu, T.; Zhang, W.; Chen, X.; Peng, C. Skin and Gut Microbiome in Psoriasis: Gaining Insight Into the Pathophysiology of It and Finding Novel Therapeutic Strategies. Front. Microbiol. 2020, 11, 589726.eng
dcterms.referencesSetty, A.R. Obesity, Waist Circumference, Weight Change, and the Risk of Psoriasis in Women: Nurses’ Health Study II. Arch. Intern. Med. 2007, 167, 1670–1675.eng
dcterms.referencesHidalgo-Cantabrana, C.; Gómez, J.; Delgado, S.; Requena-López, S.; Queiro-Silva, R.; Margolles, A.; Coto, E.; Sánchez, B.; Coto-Segura, P. Gut microbiota dysbiosis in a cohort of patients with psoriasis. Br. J. Derm. 2019, 181, 1287–1295.eng
dcterms.referencesHeitmann, J.; Frings, V.G.; Geier, A.; Goebeler, M.; Kerstan, A. Non-alcoholic fatty liver disease and psoriasis—Is there a shared proinflammatory network? J. Dtsch. Derm. Ges. 2021, 19, 517–528.eng
dcterms.referencesHedin, C.R.H.; Sonkoly, E.; Eberhardson, M.; Ståhle, M. Inflammatory bowel disease and psoriasis: Modernizing the multidisciplinary approach. J. Intern. Med. 2021, 290, 257–278.eng
dcterms.referencesTodberg, T.; Egeberg, A.; Zachariae, C.; Sørensen, N.; Pedersen, O.; Skov, L. Patients with psoriasis have a dysbiotic taxonomic and functional gut microbiota. Br. J. Derm. 2022, bjd.21245.eng
dcterms.referencesPolak, K.; Bergler-Czop, B.; Szczepanek, M.; Wojciechowska, K.; Frątczak, A.; Kiss, N. Psoriasis and Gut Microbiome-Current State of Art. Int. J. Mol. Sci. 2021, 22, 4529.eng
dcterms.referencesAoun, A.; Darwish, F.; Hamod, N. The Influence of the Gut Microbiome on Obesity in Adults and the Role of Probiotics, Prebiotics, and Synbiotics for Weight Loss. Prev. Nutr. Food Sci. 2020, 25, 113–123.eng
dcterms.referencesMuscogiuri, G.; Cantone, E.; Cassarano, S.; Tuccinardi, D.; Barrea, L.; Savastano, S.; Colao, A. Gut microbiota: A new path to treat obesity. Int. J. Obes. Supp. 2019, 9, 10–19.eng
dcterms.referencesTurnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031.eng
dcterms.referencesJohn, G.K.; Mullin, G.E. The Gut Microbiome and Obesity. Curr. Oncol. Rep. 2016, 18, 45.eng
dcterms.referencesLv, Y.; Qin, X.; Jia, H.; Chen, S.; Sun, W.; Wang, X. The association between gut microbiota composition and BMI in Chinese male college students, as analysed by next-generation sequencing. Br. J. Nutr. 2019, 122, 986–995.eng
dcterms.referencesBarengolts, E.; Green, S.J.; Chlipala, G.E.; Layden, B.T.; Eisenberg, Y.; Priyadarshini, M.; Dugas, L.R. Predictors of Obesity among Gut Microbiota Biomarkers in African American Men with and without Diabetes. Microorganisms 2019, 7, 320.eng
dcterms.referencesKim, K.N.; Yao, Y.; Ju, S.Y. Short Chain Fatty Acids and Fecal Microbiota Abundance in Humans with Obesity: A Systematic Review and Meta-Analysis. Nutrients 2019, 11, 2512.eng
dcterms.referencesGomes, A.C.; Hoffmann, C.; Mota, J.F. The human gut microbiota: Metabolism and perspective in obesity. Gut. Microbes 2018, 9, 308–325.eng
dcterms.referencesVamanu, E.; Rai, S.N. The Link between Obesity, Microbiota Dysbiosis, and Neurodegenerative Pathogenesis. Diseases 2021, 9, 45.eng
dcterms.referencesOlejniczak-Staruch, I.; Ciążyńska, M.; Sobolewska-Sztychny, D.; Narbutt, J.; Skibińska, M.; Lesiak, A. Alterations of the Skin and Gut Microbiome in Psoriasis and Psoriatic Arthritis. Int. J. Mol. Sci. 2021, 22, 3998.eng
dcterms.referencesChen, Y.; Ho, H.J.; Tseng, C.; Lai, Z.; Shieh, J.; Wu, C. Intestinal microbiota profiling and predicted metabolic dysregulation in psoriasis patients. Exp. Derm. 2018, 27, 1336–1343.eng
dcterms.referencesGhezzal, S.; Postal, B.G.; Quevrain, E.; Brot, L.; Seksik, P.; Leturque, A.; Thenet, S.; Carrière, V. Palmitic acid damages gut epithelium integrity and initiates inflammatory cytokine production. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2020, 1865, 158530.eng
dcterms.referencesGenser, L.; Aguanno, D.; Soula, H.A.; Dong, L.; Trystram, L.; Assmann, K.; Salem, J.-E.; Vaillant, J.-C.; Oppert, J.-M.; Laugerette, F.; et al. Increased jejunal permeability in human obesity is revealed by a lipid challenge and is linked to inflammation and type 2 diabetes. J. Pathol. 2018, 246, 217–230.eng
dcterms.referencesTsai, Y.-W.; Lu, C.-H.; Chang, R.C.-A.; Hsu, Y.-P.; Ho, L.-T.; Shih, K.-C. Palmitoleic acid ameliorates palmitic acid-induced proinflammation in J774A.1 macrophages via TLR4-dependent and TNF-α-independent signallings. Prostaglandins Leukot. Essent. Fat. Acids 2021, 169, 102270.eng
dcterms.referencesZhou, B.; Zhang, J.; Zhang, Q.; Permatasari, F.; Xu, Y.; Wu, D.; Yin, Z.; Luo, D. Palmitic Acid Induces Production of Proinflammatory Cytokines Interleukin-6, Interleukin-1β, and Tumor Necrosis Factor-α via a NF-κB-Dependent Mechanism in HaCaT Keratinocytes. Mediat. Inflamm. 2013, 2013, 530429.eng
dcterms.referencesGong, J.; Campos, H.; McGarvey, S.; Wu, Z.; Goldberg, R.; Baylin, A. Adipose tissue palmitoleic acid and obesity in humans: Does it behave as a lipokine? 123. Am. J. Clin. Nutr. 2011, 93, 186–191.eng
dcterms.referencesBermúdez, V.; Durán, P.; Rojas, E.; Díaz, M.P.; Rivas, J.; Nava, M.; Chacín, M.; Cabrera de Bravo, M.; Carrasquero, R.; Ponce, C.C.; et al. The Sick Adipose Tissue: New Insights Into Defective Signaling and Crosstalk With the Myocardium. Front Endocrinol. 2021, 12, 735070.eng
dcterms.referencesSharmin, M.M.; Mizusawa, M.; Hayashi, S.; Arai, W.; Sakata, S.; Yonekura, S. Effects of fatty acids on inducing endoplasmic reticulum stress in bovine mammary epithelial cells. J. Dairy Sci. 2020, 103, 8643–8654.eng
dcterms.referencesBen-Dror, K.; Birk, R. Oleic acid ameliorates palmitic acid-induced ER stress and inflammation markers in naive and cerulein-treated exocrine pancreas cells. Biosci. Rep. 2019, 39, BSR20190054.eng
dcterms.referencesHetz, C. The unfolded protein response: Controlling cell fate decisions under ER stress and beyond. Nat. Rev. Mol. Cell Biol. 2012, 13, 89–102.eng
dcterms.referencesHetz, C.; Zhang, K.; Kaufman, R.J. Mechanisms, regulation and functions of the unfolded protein response. Nat. Rev. Mol. Cell Biol. 2020, 21, 421–438.eng
dcterms.referencesRozpędek, W.; Pytel, D.; Mucha, B.; Leszczyńska, H.; Diehl, J.A.; Majsterek, I. The Role of the PERK/eIF2α/ATF4/CHOP Signaling Pathway in Tumor Progression During Endoplasmic Reticulum Stress. Curr. Mol. Med. 2016, 16, 533–544.eng
dcterms.referencesSuzuki, T.; Gao, J.; Ishigaki, Y.; Kondo, K.; Sawada, S.; Izumi, T.; Uno, K.; Kaneko, K.; Tsukita, S.; Takahashi, K.; et al. ER Stress Protein CHOP Mediates Insulin Resistance by Modulating Adipose Tissue Macrophage Polarity. Cell Rep. 2017, 18, 2045–2057.eng
dcterms.referencesMaya-Monteiro, C.M.; Bozza, P.T. Leptin and mTOR: Partners in metabolism and inflammation. Cell Cycle 2008, 7, 1713–1717.eng
dcterms.referencesCooper, P.O.; Haas, M.R.; Noonepalle, S.K.R.; Shook, B.A. Dermal Drivers of Injury-Induced Inflammation: Contribution of Adipocytes and Fibroblasts. Int. J. Mol. Sci. 2021, 22, 1933.eng
dcterms.referencesRivera-Gonzalez, G.; Shook, B.; Horsley, V. Adipocytes in Skin Health and Disease. Cold Spring Harb. Perspect. Med. 2014, 4, a015271.eng
dcterms.referencesGianfrancesco, M.A.; Dehairs, J.; L’homme, L.; Herinckx, G.; Esser, N.; Jansen, O.; Habraken, Y.; Lassence, C.; Swinnen, J.V.; Rider, M.H.; et al. Saturated fatty acids induce NLRP3 activation in human macrophages through K+ efflux resulting from phospholipid saturation and Na, K-ATPase disruption. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2019, 1864, 1017–1030.eng
dcterms.referencesAlmeida, L.; Everts, B. Fa(c)t checking: How fatty acids shape metabolism and function of macrophages and dendritic cells. Eur. J. Immunol. 2021, 51, 1628–1640.eng
dcterms.referencesStelzner, K.; Herbert, D.; Popkova, Y.; Lorz, A.; Schiller, J.; Gericke, M.; Klöting, N.; Blüher, M.; Franz, S.; Simon, J.C.; et al. Free fatty acids sensitize dendritic cells to amplify TH1/TH17-immune responses. Eur. J. Immunol. 2016, 46, 2043–2053.eng
dcterms.referencesSchumann, J. It is all about fluidity: Fatty acids and macrophage phagocytosis. Eur. J. Pharm. 2016, 785, 18–23.eng
dcterms.referencesZhao, M.; Luo, J.; Xiao, B.; Tang, H.; Song, F.; Ding, X.; Yang, G. Endoplasmic reticulum stress links psoriasis vulgaris with keratinocyte inflammation. Postepy Derm. Alergol. 2020, 37, 34–40eng
dcterms.referencesKanemaru, K.; Matsuyuki, A.; Nakamura, Y.; Fukami, K. Obesity exacerbates imiquimod-induced psoriasis-like epidermal hyperplasia and interleukin-17 and interleukin-22 production in mice. Exp. Derm. 2015, 24, 436–442.eng
dcterms.referencesVasseur, P.; Serres, L.; Jégou, J.-F.; Pohin, M.; Delwail, A.; Petit-Paris, I.; Levillain, P.; Favot, L.; Samson, M.; Yssel, H.; et al. High-Fat Diet-Induced IL-17A Exacerbates Psoriasiform Dermatitis in a Mouse Model of Steatohepatitis. Am. J. Pathol. 2016, 186, 2292–2301.eng
dcterms.referencesKlapan, K.; Simon, D.; Karaulov, A.; Gomzikova, M.; Rizvanov, A.; Yousefi, S.; Simon, H.-U. Autophagy and Skin Diseases. Front. Pharmacol. 2022, 13, 380–389.eng
dcterms.referencesChávez-Castillo, M.; Ortega, Á.; Cudris-Torres, L.; Duran, P.; Rojas, M.; Manzano, A.; Garrido, B.; Salazar, J.; Silva, A.; Rojas-Gomez, D.M.; et al. Specialized Pro-Resolving Lipid Mediators: The Future of Chronic Pain Therapy? Int. J. Mol. Sci. 2021, 22, 10370.eng
dcterms.referencesBasil, M.C.; Levy, B.D. Specialized pro-resolving mediators: Endogenous regulators of infection and inflammation. Nat. Rev. Immunol. 2016, 16, 51–67.eng
dcterms.referencesSugimoto, M.A.; Sousa, L.P.; Pinho, V.; Perretti, M.; Teixeira, M.M. Resolution of Inflammation: What Controls Its Onset? Front. Immunol. 2016, 7, 160.eng
dcterms.referencesFullerton, J.N.; Gilroy, D.W. Resolution of inflammation: A new therapeutic frontier. Nat. Rev. Drug Discov. 2016, 15, 551–567.eng
dcterms.referencesHeadland, S.E.; Norling, L.V. The resolution of inflammation: Principles and challenges. Semin. Immunol. 2015, 27, 149–160.eng
dcterms.referencesSorokin, A.V.; Domenichiello, A.F.; Dey, A.K.; Yuan, Z.-X.; Goyal, A.; Rose, S.M.; Playford, M.P.; Ramsden, C.E.; Mehta, N.N. Bioactive Lipid Mediator Profiles in Human Psoriasis Skin and Blood. J. Investig. Derm. 2018, 138, 1518–1528.eng
dcterms.referencesShim, J.H. Prostaglandin E2 Induces Skin Aging via E-Prostanoid 1 in Normal Human Dermal Fibroblasts. Int. J. Mol. Sci. 2019, 20, 5555eng
dcterms.referencesSaito-Sasaki, N.; Sawada, Y.; Nakamura, M. Maresin-1 and Inflammatory Disease. Int. J. Mol. Sci. 2022, 23, 1367eng
dcterms.referencesKrzyszczyk, P.; Schloss, R.; Palmer, A.; Berthiaume, F. The Role of Macrophages in Acute and Chronic Wound Healing and Interventions to Promote Pro-wound Healing Phenotypes. Front Physiol. 2018, 9, 419.eng
dcterms.referencesWang, W.; Xu, R.-L.; He, P.; Chen, R. MAR1 suppresses inflammatory response in LPS-induced RAW 264.7 macrophages and human primary peripheral blood mononuclear cells via the SIRT1/PGC-1α/PPAR-γ pathway. J. Inflamm. 2021, 18, 8.eng
dcterms.referencesChiurchiù, V.; Leuti, A.; Dalli, J.; Jacobsson, A.; Battistini, L.; Maccarrone, M.; Serhan, C.N. Proresolving lipid mediators resolvin D1, resolvin D2, and maresin 1 are critical in modulating T cell responses. Sci. Transl. Med. 2016, 8, 353ra111.eng
dcterms.referencesSasaki-Saito, N.; Sawada, Y.; Nakamura, M. 954 Maresin-1 inhibits IL-23 receptors via down-regulation of RORt expression and internalization in an imiquimod-induced psoriasis model mouse. J. Investig. Derm. 2018, 138, S162eng
dcterms.referencesAbdulnour, R.-E.E.; Dalli, J.; Colby, J.K.; Krishnamoorthy, N.; Timmons, J.Y.; Tan, S.H.; Colas, R.A.; Petasis, N.A.; Serhan, C.N.; Levy, B.D. Maresin 1 biosynthesis during platelet–neutrophil interactions is organ-protective. Proc. Natl. Acad. Sci. USA 2014, 111, 16526–16531.eng
dcterms.referencesSaito-Sasaki, N.; Sawada, Y.; Mashima, E.; Yamaguchi, T.; Ohmori, S.; Yoshioka, H.; Haruyama, S.; Okada, E.; Nakamura, M. Maresin-1 suppresses imiquimod-induced skin inflammation by regulating IL-23 receptor expression. Sci. Rep. 2018, 8, 5522.eng
dcterms.referencesFania, L.; Morelli, M.; Scarponi, C.; Mercurio, L.; Scopelliti, F.; Cattani, C.; Scaglione, G.L.; Tonanzi, T.; Pilla, M.A.; Pagnanelli, G.; et al. Paradoxical psoriasis induced by TNF-α blockade shows immunological features typical of the early phase of psoriasis development. J. Pathol. Clin. Res. 2020, 6, 55–68.eng
dcterms.referencesYunanto, K.A.; Waspodo, N.N.; Tabri, F.; Ilyas, F. Obesity as a comorbid factor in a boy with psoriasis vulgaris. Enfermería Clínica 2020, 30, 259–262.eng
dcterms.referencesGriffiths, C.E.; Barker, J.N. Pathogenesis and clinical features of psoriasis. Lancet 2007, 370, 263–271.eng
dcterms.referencesHugh, J.M.; Weinberg, J.M. Update on the pathophysiology of psoriasis. Cutis 2018, 102, 6–12.eng
dcterms.referencesGrän, F.; Kerstan, A.; Serfling, E.; Goebeler, M.; Muhammad, K. Current Developments in the Immunology of Psoriasis. Yale J. Biol. Med. 2020, 93, 97–110.eng
dcterms.referencesHwang, J.; Yoo, J.A.; Yoon, H.; Han, T.; Yoon, J.; An, S.; Cho, J.Y.; Lee, J. The Role of Leptin in the Association between Obesity and Psoriasis. Biomol. Ther. 2021, 29, 11–21.eng
dcterms.referencesThomas, J.; Küpper, M.; Batra, R.; Jargosch, M.; Atenhan, A.; Baghin, V.; Krause, L.; Lauffer, F.; Biedermann, T.; Theis, F.J.; et al. Is the humoral immunity dispensable for the pathogenesis of psoriasis? J. Eur. Acad. Derm. Venereol. 2019, 33, 115–122.eng
dcterms.referencesKyriakou, A.; Patsatsi, A.; Sotiriadis, D.; Goulis, D.G. Serum Leptin, Resistin, and Adiponectin Concentrations in Psoriasis: A Meta-Analysis of Observational Studies. Dermatology 2017, 233, 378–389.eng
dcterms.referencesYeung, H.; Takeshita, J.; Mehta, N.N.; Kimmel, S.E.; Ogdie, A.; Margolis, D.J.; Shin, D.B.; Attor, R.; Troxel, A.B.; Gelfand, J.M. Psoriasis Severity and the Prevalence of Major Medical Comorbidity: A Population-Based Study. JAMA Derm. 2013, 149, 1173–1179.eng
dcterms.referencesGupta, S.; Syrimi, Z.; Hughes, D.M.; Zhao, S.S. Comorbidities in psoriatic arthritis: A systematic review and meta-analysis. Rheumatol. Int. 2021, 41, 275–284.eng
dcterms.referencesHile, G.; Kahlenberg, J.M.; Gudjonsson, J.E. Recent genetic advances in innate immunity of psoriatic arthritis. Clin. Immunol. 2020, 214, 108405.eng
dcterms.referencesBen Abdallah, H.; Johansen, C.; Iversen, L. Key Signaling Pathways in Psoriasis: Recent Insights from Antipsoriatic Therapeutics. Psoriasis Targets Ther. 2021, 11, 83–97.eng
dcterms.referencesStorer, M.A.; Danesh, M.J.; Sandhu, M.E.; Pascoe, V.; Kimball, A.B. An assessment of the relative impact of hidradenitis suppurativa, psoriasis, and obesity on quality of life. Int. J. Womens Dermatol. 2018, 4, 198–202.eng
dcterms.referencesSalihbegovic, E.M.; Kurtalic, S.; Omerkic, E. Comorbidity in Men with Psoriasis. Med. Arch. 2021, 75, 31–34.eng
dcterms.referencesHiggins, E. Psoriasis. Medicine 2021, 49, 361–369.eng
dcterms.referencesShi, L.-Q.; Lian, N.; Sun, J.-T.; Liu, L.-H.; Chen, M. Association between the systemic treatment of psoriasis and cardiovascular risk. Chin. Med. J. 2021, 134, 518–520.eng
dcterms.referencesJacobi, A.; Langenbruch, A.; Purwins, S.; Augustin, M.; Radtke, M.A. Prevalence of Obesity in Patients with Psoriasis: Results of the National Study PsoHealth3. Dermatology 2015, 231, 231–238.eng
dcterms.referencesShah, S.; Nikam, B.; Kale, M.; Jamale, V.; Chavan, D. Safety and efficacy profile of oral cyclosporine vs oral methotrexate vs oral acitretin in palmoplantar psoriasis: A hospital based prospective investigator blind randomized controlled comparative study. Dermatol. Ther. 2021, 34, e14650.eng
dcterms.referencesGhoreschi, K.; Balato, A.; Enerbäck, C.; Sabat, R. Therapeutics targeting the IL-23 and IL-17 pathway in psoriasis. Lancet 2021, 397, 754–766.eng
dcterms.referencesMylonas, A.; Conrad, C. Psoriasis: Classical vs. Paradoxical. The Yin-Yang of TNF and Type I Interferon. Front. Immunol. 2018, 9, 2746.eng
dcterms.referencesLe, S.T.; Toussi, A.; Maverakis, N.; Marusina, A.I.; Barton, V.R.; Merleev, A.A.; Luxardi, G.; Raychaudhuri, S.P.; Maverakis, E. The cutaneous and intestinal microbiome in psoriatic disease. Clin. Immunol. 2020, 218, 108537.eng
dcterms.referencesGuo, Z.; Yang, Y.; Liao, Y.; Shi, Y.; Zhang, L. Emerging Roles of Adipose Tissue in the Pathogenesis of Psoriasis and Atopic Dermatitis in Obesity. JID Innov. 2022, 2, 100064.eng
dcterms.referencesHsu, D.K.; Fung, M.A.; Chen, H.-L. Role of skin and gut microbiota in the pathogenesis of psoriasis, an inflammatory skin disease. Med. Microecol. 2020, 4, 100016.eng
dcterms.referencesSzántó, M.; Dózsa, A.; Antal, D.; Szabó, K.; Kemény, L.; Bai, P. Targeting the gut-skin axis—Probiotics as new tools for skin disorder management? Exp. Derm. 2019, 28, 1210–1218.eng
dcterms.referencesThye, A.Y.-K.; Bah, Y.-R.; Law, J.W.-F.; Tan, L.T.-H.; He, Y.-W.; Wong, S.-H.; Thurairajasingam, S.; Chan, K.-G.; Lee, L.-H.; Letchumanan, V. Gut–Skin Axis: Unravelling the Connection between the Gut Microbiome and Psoriasis. Biomedicines 2022, 10, 1037.eng
dcterms.referencesChen, Y.-H.; Wu, C.-S.; Chao, Y.-H.; Lin, C.-C.; Tsai, H.-Y.; Li, Y.-R.; Chen, Y.-Z.; Tsai, W.-H.; Chen, Y.-K. Lactobacillus pentosus GMNL-77 inhibits skin lesions in imiquimod-induced psoriasis-like mice. J. Food Drug Anal. 2017, 25, 559–566.eng
dcterms.referencesThio, H.B. The Microbiome in Psoriasis and Psoriatic Arthritis: The Skin Perspective. J. Rheumatol. 2018, 94, 30–31.eng
dcterms.referencesVijayashankar, M.; Raghunath, N. Pustular psoriasis responding to Probiotics—A new insight. Our Derm. Online 2012, 3, 326–329.eng
dcterms.referencesGroeger, D.; O’Mahony, L.; Murphy, E.F.; Bourke, J.F.; Dinan, T.G.; Kiely, B.; Shanahan, F.; Quigley, E.M.M. Bifidobacterium infantis 35,624 modulates host inflammatory processes beyond the gut. Gut. Microbes. 2013, 4, 325–339.eng
dcterms.referencesLin, C.; Zeng, T.; Deng, Y.; Yang, W.; Xiong, J. [Treatment of psoriasis vulgaris using Bacteroides fragilis BF839: A single-arm, open preliminary clinical study]. Sheng Wu Gong Cheng Xue Bao 2021, 37, 3828–3835.eng
dcterms.referencesCarrara, M.; Kelly, M.T.; Roso, F.; Larroque, M.; Margout, D. Potential of Olive Oil Mill Wastewater as a Source of Polyphenols for the Treatment of Skin Disorders: A Review. J. Agric. Food Chem. 2021, 69, 7268–7284.eng
dcterms.referencesMedina, M. Tratamiento Alternativo al Uso Crónico de Esteroides e Inmunosupresores en Psoriasis Mediante Polifenoles Aislados del Aceite Del Oliva. Ph.D. Thesis, Universidad del Zulia, Maracaibo, Venezuela.spa
dcterms.referencesMedina, M.; Cano, R.; Contreras, I.; Prado, A.; Cano, C. Estudio comparativo de la eficiencia terapéutica entre imiquimod crema al 5% y una loción basada en polifenoles en el tratamiento de la queratosis actínica. Arch. Venez. Farmacol. Ter. 2015, 34, 16–20.spa
dcterms.referencesYan, D.; Blauvelt, A.; Dey, A.K.; Golpanian, R.S.; Hwang, S.T.; Mehta, N.N.; Myers, B.; Shi, Z.; Yosipovitch, G.; Bell, S.; et al. New Frontiers in Psoriatic Disease Research, Part II: Comorbidities and Targeted Therapies. J. Investig. Dermatol. 2021, 141, 2328–2337.eng
dcterms.referencesGriffiths, C.E.M.; Armstrong, A.W.; Gudjonsson, J.E.; Barker, J.N.W.N. Psoriasis. Lancet 2021, 397, 1301–1315.eng
dcterms.referencesPirro, F.; Caldarola, G.; Chiricozzi, A.; Burlando, M.; Mariani, M.; Parodi, A.; Peris, K.; De Simone, C. Impact of Body Mass Index on the Efficacy of Biological Therapies in Patients with Psoriasis: A Real-World Study. Clin. Drug Investig. 2021, 41, 917–925.eng
dcterms.referencesSu, X.; Zhang, G.; Cheng, Y.; Wang, B. Leptin in skin disease modulation. Clin. Chim. Acta 2021, 516, 8–14.eng
dcterms.referencesMadden, S.K.; Flanagan, K.L.; Jones, G. How lifestyle factors and their associated pathogenetic mechanisms impact psoriasis. Clin. Nutr. 2020, 39, 1026–1040.eng
dcterms.referencesVillarreal-Calderón, J.R.; Cuéllar, R.X.; Ramos-González, M.R.; Rubio-Infante, N.; Castillo, E.C.; Elizondo-Montemayor, L.; García-Rivas, G. Interplay between the Adaptive Immune System and Insulin Resistance in Weight Loss Induced by Bariatric Surgery. Oxidative Med. Cell. Longev. 2019, 2019, 1–14.eng
dcterms.referencesMaglio, C.; Peltonen, M.; Rudin, A.; Carlsson, L.M.S. Bariatric Surgery and the Incidence of Psoriasis and Psoriatic Arthritis in the Swedish Obese Subjects Study: Effect of Bariatric Surgery on Psoriasis. Obesity 2017, 25, 2068–2073.eng
dcterms.referencesEgeberg, A.; Sørensen, J.A.; Gislason, G.H.; Knop, F.K.; Skov, L. Incidence and Prognosis of Psoriasis and Psoriatic Arthritis in Patients Undergoing Bariatric Surgery. JAMA Surg. 2017, 152, 344.eng
dcterms.referencesFarias, M.M.; Achurra, P.; Boza, C.; Vega, A.; de la Cruz, C. Psoriasis following bariatric surgery: Clinical evolution and impact on quality of life on 10 patients. Obes. Surg. 2012, 22, 877–880.eng
dcterms.referencesAlruwaili, H.; Dehestani, B.; le Roux, C.W. Clinical Impact of Liraglutide as a Treatment of Obesity. CPAA 2021, 13, 53–60.eng
dcterms.referencesWilding, J.P.H.; Batterham, R.L.; Calanna, S.; Davies, M.; Van Gaal, L.F.; Lingvay, I.; McGowan, B.M.; Rosenstock, J.; Tran, M.T.D.; Wadden, T.A.; et al. Once-Weekly Semaglutide in Adults with Overweight or Obesity. N. Engl. J. Med. 2021, 384, 989–1002.eng
dcterms.referencesZobel, E.H.; Ripa, R.S.; von Scholten, B.J.; Rotbain Curovic, V.; Kjaer, A.; Hansen, T.W.; Rossing, P.; Størling, J. Effect of liraglutide on expression of inflammatory genes in type 2 diabetes. Sci. Rep. 2021, 11, 18522.eng
dcterms.referencesChang, G.; Chen, B.; Zhang, L. Efficacy of GLP-1rA, liraglutide, in plaque psoriasis treatment with type 2 diabetes: A systematic review and meta-analysis of prospective cohort and before-after studies. J. Dermatol. Treat. 2022, 33, 1299–1305.eng
dcterms.referencesXu, X.; Lin, L.; Chen, P.; Yu, Y.; Chen, S.; Chen, X.; Shao, Z. Treatment with liraglutide, a glucagon-like peptide-1 analogue, improves effectively the skin lesions of psoriasis patients with type 2 diabetes: A prospective cohort study. Diabetes Res. Clin. Pract. 2019, 150, 167–173.eng
dcterms.referencesLin, L.; Xu, X.; Yu, Y.; Ye, H.; He, X.; Chen, S.; Chen, X.; Shao, Z.; Chen, P. Glucagon-like peptide-1 receptor agonist liraglutide therapy for psoriasis patients with type 2 diabetes: A randomized-controlled trial. J. Dermatol. Treat. 2022, 33, 1428–1434eng
dcterms.referencesCostanzo, G.; Curatolo, S.; Busà, B.; Belfiore, A.; Gullo, D. Two birds one stone: Semaglutide is highly effective against severe psoriasis in a type 2 diabetic patient. Endocrinol. Diabetes Metab. Case Rep. 2021, 2021.eng
oaire.versioninfo:eu-repo/semantics/publishedVersioneng

Archivos

Bloque original
Mostrando 1 - 1 de 1
Cargando...
Miniatura
Nombre:
2022_MDPI_Exploring the Links between.pdf
Tamaño:
952.64 KB
Formato:
Adobe Portable Document Format
Descripción:
PDF
Bloque de licencias
Mostrando 1 - 1 de 1
No hay miniatura disponible
Nombre:
license.txt
Tamaño:
381 B
Formato:
Item-specific license agreed upon to submission
Descripción:

Colecciones