1
|
Ridha-Salman H, Shihab EM, Hasan HK, Abbas AH, Khorsheed SM, Ayad Fakhri S. Mitigative Effects of Topical Norfloxacin on an Imiquimod-Induced Murine Model of Psoriasis. ACS Pharmacol Transl Sci 2024; 7:2739-2754. [PMID: 39296262 PMCID: PMC11406690 DOI: 10.1021/acsptsci.4c00152] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 09/21/2024]
Abstract
Psoriasis is a chronic, inflammatory dermatosis characterized by thickened, reddened, and scaly skin lesions. Norfloxacin is a fluoroquinolone antibiotic with enhanced antioxidant, anti-inflammatory, and immunomodulatory bioactivities. The aim of this study was to figure out the possible impact of topical norfloxacin on an imiquimod-induced model of psoriasis in mice. Thirty albino-type mice were split into five distinct groups of six animals each. The control group included healthy mice that had not received any treatment. The induction group was given the vehicle 2 h after the topical imiquimod, once daily for 8 days. Two hours after receiving topical imiquimod, the treatment groups including calcipotriol, norfloxacin 2.5%, and norfloxacin 5% were given topical ointments containing calcipotriol 0.005%, norfloxacin 2.5%, and norfloxacin 5%, for 8 days. Topical norfloxacin ointment significantly reduced the severity of imiquimod-exacerbated psoriatic lesions including erythema, shiny-white scaling, and acanthosis and fixed histological abnormalities. Furthermore, imiquimod-subjected mice treated with a higher concentration of norfloxacin ointment exhibited dramatically lower skin levels of inflammation-related biomarkers like IFN-γ, TNF-α, IL-6, IL-17A, IL-23, and TGF-β but higher levels of IL-10. They also demonstrated a notable decrease in angiogenesis parameters such as VEGF and IL-8, a substantial reduction in oxidative indicators like MDA and MPO, and a considerable rise in antioxidant enzymes like SOD and CAT. This study offers novel evidence that norfloxacin may assist in controlling inflammatory dermatoses like psoriasis by minimizing the severity of psoriatic plaques, correcting histological alterations, and diminishing the production of inflammatory, oxidative, and angiogenetic parameters.
Collapse
Affiliation(s)
- Hayder Ridha-Salman
- Department of Pharmacology, College of Pharmacy, Al-Mustaqbal University, Hillah 51001, Babylon +964, Iraq
| | - Elaf Mahmood Shihab
- Department of Pharmacology, College of Pharmacy, Al-Esraa University, Baghdad +964, Iraq
| | - Hasanain Kamil Hasan
- Department of Pharmacology, College of Pharmacy, Al-Mustaqbal University, Hillah 51001, Babylon +964, Iraq
| | - Alaa Hamza Abbas
- Department of Pharmacology, College of Pharmacy, Al-Mustaqbal University, Hillah 51001, Babylon +964, Iraq
| | | | - Salar Ayad Fakhri
- Department of Pharmacology, College of Pharmacy, Al-Esraa University, Baghdad +964, Iraq
| |
Collapse
|
2
|
Dascălu RC, Bărbulescu AL, Stoica LE, Dinescu ȘC, Biță CE, Popoviciu HV, Ionescu RA, Vreju FA. Review: A Contemporary, Multifaced Insight into Psoriasis Pathogenesis. J Pers Med 2024; 14:535. [PMID: 38793117 PMCID: PMC11122105 DOI: 10.3390/jpm14050535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Psoriasis is a chronic recurrent inflammatory autoimmune pathology with a significant genetic component and several interferences of immunological cells and their cytokines. The complex orchestration of psoriasis pathogenesis is related to the synergic effect of immune cells, polygenic alterations, autoantigens, and several other external factors. The major act of the IL-23/IL-17 axis, strongly influencing the inflammatory pattern established during the disease activity, is visible as a continuous perpetuation of the pro-inflammatory response and keratinocyte activation and proliferation, leading to the development of psoriatic lesions. Genome-wide association studies (GWASs) offer a better view of psoriasis pathogenic pathways, with approximately one-third of psoriasis's genetic impact on psoriasis development associated with the MHC region, with genetic loci located on chromosome 6. The most eloquent genetic factor of psoriasis, PSORS1, was identified in the MHC I site. Among the several factors involved in its complex etiology, dysbiosis, due to genetic or external stimulus, induces a burst of pro-inflammatory consequences; both the cutaneous and gut microbiome get involved in the psoriasis pathogenic process. Cutting-edge research studies and comprehensive insights into psoriasis pathogenesis, fostering novel genetic, epigenetic, and immunological factors, have generated a spectacular improvement over the past decades, securing the path toward a specific and targeted immunotherapeutic approach and delayed progression to inflammatory arthritis. This review aimed to offer insight into various domains that underline the pathogenesis of psoriasis and how they influence disease development and evolution. The pathogenesis mechanism of psoriasis is multifaceted and involves an interplay of cellular and humoral immunity, which affects susceptible microbiota and the genetic background. An in-depth understanding of the role of pathogenic factors forms the basis for developing novel and individualized therapeutic targets that can improve disease management.
Collapse
Affiliation(s)
- Rucsandra Cristina Dascălu
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.C.D.); (C.E.B.); (F.A.V.)
| | - Andreea Lili Bărbulescu
- Department of Pharmacology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Loredana Elena Stoica
- Department of Dermatology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Ștefan Cristian Dinescu
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.C.D.); (C.E.B.); (F.A.V.)
| | - Cristina Elena Biță
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.C.D.); (C.E.B.); (F.A.V.)
| | - Horațiu Valeriu Popoviciu
- Department of Rheumatology, BFK and Medical Rehabilitation, University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Mures, Romania;
| | - Răzvan Adrian Ionescu
- Third Internal Medicine Department, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Florentin Ananu Vreju
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.C.D.); (C.E.B.); (F.A.V.)
| |
Collapse
|
3
|
Zhang W, Jia Q, Han M, Zhang X, Guo L, Sun S, Yin W, Bo C, Han R, Sai L. Bifidobacteria in disease: from head to toe. Folia Microbiol (Praha) 2024; 69:1-15. [PMID: 37644256 DOI: 10.1007/s12223-023-01087-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Bifidobacteria as a strictly anaerobic gram-positive bacteria, is widely distributed in the intestine, vagina and oral cavity, and is one of the first gut flora to colonize the early stages of life. Intestinal flora is closely related to health, and dysbiosis of intestinal flora, especially Bifidobacteria, has been found in a variety of diseases. Numerous studies have shown that in addition to maintaining intestinal homeostasis, Bifidobacteria may be involved in diseases covering all parts of the body, including the nervous system, respiratory system, genitourinary system and so on. This review collects evidence for the variation of Bifidobacteria in typical diseases among various systems, provides mild and effective therapeutic options for those diseases that are difficult to cure, and moves Bifidobacteria from basic research to further clinical applications.
Collapse
Affiliation(s)
- Weiliang Zhang
- Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong, China
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingming Han
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xin Zhang
- Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong, China
| | - Limin Guo
- Rongcheng Municipal Hospital of Traditional Chinese Medicine, Rongcheng, Shandong, China
| | - Shichao Sun
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Shandong University of Traditional Chinese Medicine Doctoral candidate Class of 2022, Jinan, Shandong, China
| | - Wenhui Yin
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Cunxiang Bo
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ru Han
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Linlin Sai
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
4
|
Siu PLK, Choy CT, Chan HHY, Leung RKK, Chan UK, Zhou J, Wong CH, Lee YW, Chan HW, Lo CJY, Tsui JCC, Loo SKF, Tsui SKW. A Novel Multi-Strain E3 Probiotic Formula Improved the Gastrointestinal Symptoms and Quality of Life in Chinese Psoriasis Patients. Microorganisms 2024; 12:208. [PMID: 38276193 PMCID: PMC10820679 DOI: 10.3390/microorganisms12010208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Psoriasis is a chronic immune-mediated inflammatory disease affecting the skin and other systems. Gastrointestinal disease was found to be correlated with psoriasis in previous studies and it can significantly affect the quality of life of psoriasis patients. Despite the importance of the gut microbiome in gut and skin health having already been demonstrated in many research studies, the potential effect of probiotics on GI comorbidities in psoriasis patients is unclear. To investigate the effects of probiotics on functional GI comorbidities including irritable bowel syndrome, functional constipation, and functional diarrhea in psoriasis patients, we conducted a targeted 16S rRNA sequencing and comprehensive bioinformatic analysis among southern Chinese patients to compare the gut microbiome profiles of 45 psoriasis patients over an 8-week course of novel oral probiotics. All the participants were stratified into responders and non-responders according to their improvement in GI comorbidities, which were based on their Bristol Stool Form Scale (BSFS) scores after intervention. The Dermatological Life Quality Index (DLQI) score revealed a significant improvement in quality of life within the responder group (DLQI: mean 10.4 at week 0 vs. mean 15.9 at week 8, p = 0.0366). The proportion of psoriasis patients without GI comorbidity manifestation at week 8 was significantly higher than that at week 0 (week 0: Normal 53.33%, Constipation/Diarrhea 46.67%; week 8: Normal 75.56%, Constipation/Diarrhea 24.44%, p = 0.0467). In addition, a significant difference in the gut microbiome composition between the responders and non-responders was observed according to alpha and beta diversities. Differential abundance analysis revealed that the psoriasis patients exhibited (1) an elevated relative abundance of Lactobacillus acidophilus, Parabacteroides distasonis, and Ruminococcus bromii and (2) a reduced relative abundance of Oscillibacter, Bacteroides vulgatus, Escherichia sp., and Biophila wadsworthia after the 8-week intervention. The responders also exhibited a higher relative abundance of Fusicatenibacter saccharivorans when compared to the non-responders. In summary, our study discovers the potential clinical improvement effects of the novel probiotic formula in improving GI comorbidities and quality of life in psoriasis patients. We also revealed the different gut microbiome composition as well as the gut microbial signatures in the patients who responded to probiotics. These findings could provide insight into the use of probiotics in the management of psoriasis symptoms.
Collapse
Affiliation(s)
- Pui Ling Kella Siu
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Chi Tung Choy
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Helen Hoi Yin Chan
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Ross Ka Kit Leung
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Un Kei Chan
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Junwei Zhou
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Chi Ho Wong
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Yuk Wai Lee
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Ho Wang Chan
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Claudia Jun Yi Lo
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Joseph Chi Ching Tsui
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Steven King Fan Loo
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
- Hong Kong Institute of Integrative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Dermatology Centre, CUHK Medical Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Stephen Kwok Wing Tsui
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Centre for Microbial Genomics and Proteomics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
5
|
Salman HR, Al-Zubaidy AA, Abbas AH, Zigam QA. The ameliorative effects of topical gemifloxacin alone or in combination with clobetasol propionate on imiquimod-induced model of psoriasis in mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:599-616. [PMID: 37490123 DOI: 10.1007/s00210-023-02629-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023]
Abstract
Psoriasis is a lifelong immune-driven skin condition characterized by excessive epidermal overgrowth and inflammatory cell infiltration. Gemifloxacin is a fourth-generation fluoroquinolone with improved immunomodulatory and anti-inflammatory properties that are believed to possess an attractive role in psoriasis via suppressing the production of cytokines, chemokines, and eosinophil and neutrophil chemotaxis. The aim of this research is to investigate the ameliorative effects of prolonged topical gemifloxacin (GMF) alone and combined with clobetasol propionate (CLO) on an imiquimod (IMQ)-induced mouse model of psoriasis. Forty-eight Swiss albino mice were divided into six groups of eight. All groups except the negative controls got 62.5 mg of IMQ 5% topically for 8 days. Mice in the control group (controls) got Vaseline instead. Following the induction in the IMQ 5% group, mice in treatment groups CLO 0.05, GMF 1%, GMF 3%, and CLO + GMF obtained clobetasol propionate 0.05%, GMF 1% and 3%, and a combination of both, respectively, for an additional 8 days, rendering the experiment 16 days long. Our results revealed that gemifloxacin alleviated erythematous, thickened, and scaly psoriatic lesions and inhibited the tissue level of inflammatory cytokines, including interleukin (IL)-8, IL-17A, IL-23, tumor necrosis factor-α (TNF-α), and transforming growth factor-β1 (TGF-β1). The anti-inflammatory effect also occurred by hindering nuclear factor-kappa B (NF-κB) signaling and reversing histopathological problems. Gemifloxacin acts effectively in mitigating psoriasis-associated lesions and restricting NF-κB-mediated inflammation, recommending gemifloxacin as a promising adjuvant candidate for additional studies on the long-term treatment of autoimmune and autoinflammatory dermatoses like psoriasis.
Collapse
Affiliation(s)
- Hayder Ridha Salman
- Department of Pharmacology, College of Pharmacy, Al-Mustaqbal University, 510001, Hillah, Babylon, Iraq.
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq.
| | - Adeeb Ahmed Al-Zubaidy
- Department of Pharmacology, College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
| | - Alaa Hamza Abbas
- Department of Pharmacology, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | - Qassim A Zigam
- Department of Pharmacology, College of Pharmacy, Al-Mustaqbal University, 510001, Hillah, Babylon, Iraq
| |
Collapse
|
6
|
Lai Y, Wu X, Chao E, Bloomstein JD, Wei G, Hwang ST, Shi Z. Impact of Gut Bacterial Metabolites on Psoriasis and Psoriatic Arthritis: Current Status and Future Perspectives. J Invest Dermatol 2023; 143:1657-1666. [PMID: 37422760 DOI: 10.1016/j.jid.2023.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/04/2023] [Accepted: 05/17/2023] [Indexed: 07/10/2023]
Abstract
There is growing evidence that supports a role of gut dysbiosis in the pathogenesis of psoriasis (Pso). Thus, probiotic supplementation and fecal microbiota transplantation may serve as promising preventive and therapeutic strategies for patients with Pso. One of the basic mechanisms through which the gut microbiota interacts with the host is through bacteria-derived metabolites, usually intermediate or end products produced by microbial metabolism. In this study, we provide an up-to-date review of the most recent literature on microbial-derived metabolites and highlight their roles in the immune system, with a special focus on Pso and one of its most common comorbidities, psoriatic arthritis.
Collapse
Affiliation(s)
- Yuhsien Lai
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuesong Wu
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Ellen Chao
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | | | - Grace Wei
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Sam T Hwang
- Department of Dermatology, University of California, Davis, Sacramento, California, USA
| | - Zhenrui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
7
|
Yu N, Wang J, Liu Y, Guo Y. Investigating the gut microbiota's influence on psoriasis and psoriatic arthritis risk: a Mendelian randomization analysis. PRECISION CLINICAL MEDICINE 2023; 6:pbad023. [PMID: 38025973 PMCID: PMC10680138 DOI: 10.1093/pcmedi/pbad023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/07/2023] [Indexed: 12/01/2023] Open
Abstract
Background Numerous investigations have revealed the interplay between gut microbiota (GM) and psoriasis (Ps) and psoriatic arthritis (PsA). However, the causal relationship between them remains unknown. Methods We curated a collection of genetic variants (P < 1 × 10-5) associated with GM (n = 18 340) derived from the MiBioGen study. To explore the intricate relationship between GM and Ps as well as PsA, we harnessed the comprehensive resources of the FinnGen database, encompassing a vast cohort of individuals, including 4510 Ps cases and 212 242 controls and 1637 PsA cases and 212 242 controls. Mendelian randomization (MR) was used, including an inverse variance weighting method, followed by a sensitivity analysis to verify the robustness of the results. Results For Ps, some bacterial taxa, including Lactococcus, Ruminiclostridium 5, and Eubacterium fissicatena, were identified as risk factors; but Odoribacter demonstrated a protective effect against Ps. In the case of PsA, Lactococcus, Verrucomicrobiales, Akkermansia, Coprococcus 1, and Verrucomicrobiaceae were identified as risk factors; Odoribacter and Rikenellaceae exhibited a protective effect against the development of PsA. Conclusion Our study establishes a causal link between the GM and Ps and PsA. These findings provide insights into the underlying mechanisms and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Nianzhou Yu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiayi Wang
- Xiangya School of Medicine, Central South University, Changsha 410083, China
| | - Yuancheng Liu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yeye Guo
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
8
|
Constantin C, Surcel M, Munteanu A, Neagu M. Insights into Nutritional Strategies in Psoriasis. Nutrients 2023; 15:3528. [PMID: 37630719 PMCID: PMC10458768 DOI: 10.3390/nu15163528] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/18/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Psoriasis, an autoimmune chronic inflammatory skin condition, has a high incidence in the general population, reaching 2-4%. Its pathogenesis involves an interplay of genetic factors, immune disturbances, and environmental factors. Within the environmental factors that aid the appearance of this autoimmune skin disease, the Western lifestyle and overall diet play important roles in the steady growth in psoriasis prevalence. Furthermore, psoriasis is associated with comorbidities such as psoriatic arthritis, cardiovascular disease, metabolic syndrome, and obesity. Accumulating evidence suggests that obesity is an important risk factor for psoriasis. Moreover, obesity aggravates established psoriasis, and a reduction in the body mass index can improve the clinical outcomes of psoriasis and increase the efficacy of standard psoriasis therapies. The possible connection between this autoimmune disease and obesity relies on the fact that white adipose tissue is an essential endocrine organ that secretes an array of immune mediators and inflammatory and metabolic factors with pro-inflammatory action. Thus, immune-mediated mechanisms in both psoriasis and obesity conditions are common factors. This paper describes the factors that link obesity with skin autoimmune disease and highlights the importance of the stimulatory or regulatory effects of nutrients and food in psoriasis and the possible improvement of psoriasis through nutritional strategies.
Collapse
Affiliation(s)
- Carolina Constantin
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (C.C.); (M.S.); (A.M.)
- Pathology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Mihaela Surcel
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (C.C.); (M.S.); (A.M.)
| | - Adriana Munteanu
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (C.C.); (M.S.); (A.M.)
| | - Monica Neagu
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (C.C.); (M.S.); (A.M.)
- Pathology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Doctoral School, Faculty of Biology, University of Bucharest, 050107 Bucharest, Romania
| |
Collapse
|
9
|
Bharadwaj R, Lusi CF, Mashayekh S, Nagar A, Subbarao M, Kane GI, Wodzanowski KA, Brown AR, Okuda K, Monahan A, Paik D, Nandy A, Anonick MV, Goldman WE, Kanneganti TD, Orzalli MH, Grimes CL, Atukorale PU, Silverman N. Methotrexate suppresses psoriatic skin inflammation by inhibiting muropeptide transporter SLC46A2 activity. Immunity 2023; 56:998-1012.e8. [PMID: 37116499 PMCID: PMC10195032 DOI: 10.1016/j.immuni.2023.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/04/2023] [Accepted: 04/03/2023] [Indexed: 04/30/2023]
Abstract
Cytosolic innate immune sensing is critical for protecting barrier tissues. NOD1 and NOD2 are cytosolic sensors of small peptidoglycan fragments (muropeptides) derived from the bacterial cell wall. These muropeptides enter cells, especially epithelial cells, through unclear mechanisms. We previously implicated SLC46 transporters in muropeptide transport in Drosophila immunity. Here, we focused on Slc46a2, which was highly expressed in mammalian epidermal keratinocytes, and showed that it was critical for the delivery of diaminopimelic acid (DAP)-muropeptides and activation of NOD1 in keratinocytes, whereas the related transporter Slc46a3 was critical for delivering the NOD2 ligand MDP to keratinocytes. In a mouse model, Slc46a2 and Nod1 deficiency strongly suppressed psoriatic inflammation, whereas methotrexate, a commonly used psoriasis therapeutic, inhibited Slc46a2-dependent transport of DAP-muropeptides. Collectively, these studies define SLC46A2 as a transporter of NOD1-activating muropeptides, with critical roles in the skin barrier, and identify this transporter as an important target for anti-inflammatory intervention.
Collapse
Affiliation(s)
- Ravi Bharadwaj
- Program in Innate Immunity and Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Christina F Lusi
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | | | - Abhinit Nagar
- Program in Innate Immunity and Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Malireddi Subbarao
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Griffin I Kane
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | | | - Ashley R Brown
- Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Kendi Okuda
- Program in Innate Immunity and Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Amanda Monahan
- Program in Innate Immunity and Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Donggi Paik
- Program in Innate Immunity and Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Anubhab Nandy
- Program in Innate Immunity and Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | | - William E Goldman
- Department of Microbiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | - Megan H Orzalli
- Program in Innate Immunity and Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | | - Prabhani U Atukorale
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Neal Silverman
- Program in Innate Immunity and Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
10
|
Choy CT, Chan UK, Siu PLK, Zhou J, Wong CH, Lee YW, Chan HW, Tsui JCC, Loo SKF, Tsui SKW. A Novel E3 Probiotics Formula Restored Gut Dysbiosis and Remodelled Gut Microbial Network and Microbiome Dysbiosis Index (MDI) in Southern Chinese Adult Psoriasis Patients. Int J Mol Sci 2023; 24:ijms24076571. [PMID: 37047542 PMCID: PMC10094986 DOI: 10.3390/ijms24076571] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Psoriasis is a common chronic immune-mediated inflammatory skin disease with the association of various comorbidities. Despite the introduction of highly effective biologic therapies over the past few decades, the exact trigger for an immune reaction in psoriasis is unclear. With the majority of immune cells residing in the gut, the effect of gut microbiome dysbiosis goes beyond the gastrointestinal site and may exacerbate inflammation and regulate the immune system elsewhere, including but not limited to the skin via the gut-skin axis. In order to delineate the role of the gut microbiome in Southern Chinese psoriasis patients, we performed targeted 16S rRNA sequencing and comprehensive bioinformatic analysis to compare the gut microbiome profile of 58 psoriasis patients against 49 healthy local subjects presumably with similar lifestyles. Blautia wexlerae and Parabacteroides distasonis were found to be enriched in psoriasis patients and in some of the healthy subjects, respectively. Metabolic functional pathways were predicted to be differentially abundant, with a clear shift toward SCFA synthesis in healthy subjects. The alteration of the co-occurrence network was also evident in the psoriasis group. In addition, we also profiled the gut microbiome in 52 of the 58 recruited psoriasis patients after taking 8 weeks of an orally administrated novel E3 probiotics formula (with prebiotics, probiotics and postbiotics). The Dermatological Life Quality Index (p = 0.009) and Psoriasis Area and Severity Index (p < 0.001) were significantly improved after taking 8 weeks of probiotics with no adverse effect observed. We showed that probiotics could at least partly restore gut dysbiosis via the modulation of the gut microbiome. Here, we also report the potential application of a machine learning-derived gut dysbiosis index based on a quantitative PCR panel (AUC = 0.88) to monitor gut dysbiosis in psoriasis patients. To sum up, our study suggests the gut microbial landscape differed in psoriasis patients at the genera, species, functional and network levels. Additionally, the dysbiosis index could be a cost-effective and rapid tool to monitor probiotics use in psoriasis patients.
Collapse
Affiliation(s)
- Chi Tung Choy
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China
| | - Un Kei Chan
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China
| | - Pui Ling Kella Siu
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China
| | - Junwei Zhou
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China
| | - Chi Ho Wong
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China
| | - Yuk Wai Lee
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China
| | - Ho Wang Chan
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China
| | | | - Steven King Fan Loo
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China
- Hong Kong Institute of Integrative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Dermatology Centre, CUHK Medical Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Stephen Kwok Wing Tsui
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Centre for Microbial Genomics and Proteomics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
11
|
Paradoxical Reactions to Anti-TNFα and Anti-IL-17 Treatment in Psoriasis Patients: Are Skin and/or Gut Microbiota Involved? Dermatol Ther (Heidelb) 2023; 13:911-933. [PMID: 36929119 DOI: 10.1007/s13555-023-00904-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/14/2023] [Indexed: 03/18/2023] Open
Abstract
Psoriasis is a chronic, immune-mediated, inflammatory disease primarily affecting the skin. It is currently coming to light that patients with psoriasis have disrupted intestinal barrier and often suffer from comorbidities associated with the gastrointestinal tract. Moreover, there is growing evidence of both cutaneous and intestinal paradoxical reactions during biologic treatment in patients with psoriasis. This review focuses on barrier defects and changes in immune responses in patients with psoriasis, which play an important role in the development of the disease but are also influenced by modern biological treatments targeting IL-17 and TNFα cytokines. Here, we highlight the relationship between the gut-skin axis, microbiota, psoriasis treatment, and the incidence of paradoxical reactions, such as inflammatory bowel disease in patients with psoriasis. A better understanding of the interconnection of these mechanisms could lead to a more personalized therapy and lower the incidence of treatment side effects, thereby improving the quality of life of the affected patients.
Collapse
|
12
|
Wen C, Pan Y, Gao M, Wang J, Huang K, Tu P. Altered gut microbiome composition in nontreated plaque psoriasis patients. Microb Pathog 2023; 175:105970. [PMID: 36621696 DOI: 10.1016/j.micpath.2023.105970] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Recent studies have demonstrated that dysbiosis of the gut microbiota is associated with psoriasis, but these studies showed some conflicting results. Our study examined differences in microbiome composition associated in people with psoriasis and those without. Comparing individuals with their healthy partners was a second strategy. We explored the fecal microbiota among 32 nontreated plaque psoriasis patients, 15 healthy controls and 17 healthy couples by metagenomic gene sequencing. The relative levels of intestinal microbiota of the psoriasis cohort differed from those in healthy controls and these patients' partners. However, there was no microbial diversity among these three cohorts. On the level of the phylum, Firmicutes and Bacteroidetes' relative abundances were reversed. Escherichia coli was significantly enriched in the psoriasis group compared with the healthy people and the healthy spouses. Gene functional analysis indicated that Ribosome (ko03010) was upregulated, Flagellar assembly (ko02040) and Bacterial chemotaxis (ko02030) were downregulated in the psoriasis cohort compared with the healthy individuals and the healthy spouses. The microbiota in severe psoriasis patients differed from those with milder conditions. These findings strongly support the association between intestinal flora and psoriasis. It is necessary to perform more meaningful experiments to identify whether the differences of gut microbiota are the cause or consequences of psoriasis in future.
Collapse
Affiliation(s)
- Chunmiao Wen
- Department of Dermatology and Venerology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, No.9 Beiguan Street, Tongzhou District, Beijing, 101149, China
| | - Ming Gao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Jianlei Wang
- Department of Dermatology and Venerology, Civil Aviation General Hospital, 1 Gaojing A, Chaoyang District, Beijing, 100123, China
| | - Kun Huang
- Department of Gastroenterology, Civil Aviation General Hospital, 1 Gaojing A, Chaoyang District, Beijing, 100123, China
| | - Ping Tu
- Department of Dermatology and Venerology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing, 100034, China.
| |
Collapse
|
13
|
Interrogation of the mammalian gut-brain axis using LC-MS/MS-based targeted metabolomics with in vitro bacterial and organoid cultures and in vivo gnotobiotic mouse models. Nat Protoc 2023; 18:490-529. [PMID: 36352124 DOI: 10.1038/s41596-022-00767-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 07/26/2022] [Indexed: 11/11/2022]
Abstract
Interest in the communication between the gastrointestinal tract and central nervous system, known as the gut-brain axis, has prompted the development of quantitative analytical platforms to analyze microbe- and host-derived signals. This protocol enables investigations into connections between microbial colonization and intestinal and brain neurotransmitters and contains strategies for the comprehensive evaluation of metabolites in in vitro (organoids) and in vivo mouse model systems. Here we present an optimized workflow that includes procedures for preparing these gut-brain axis model systems: (stage 1) growth of microbes in defined media; (stage 2) microinjection of intestinal organoids; and (stage 3) generation of animal models including germ-free (no microbes), specific-pathogen-free (complete gut microbiota) and specific-pathogen-free re-conventionalized (germ-free mice associated with a complete gut microbiota from a specific-pathogen-free mouse), and Bifidobacterium dentium and Bacteroides ovatus mono-associated mice (germ-free mice colonized with a single gut microbe). We describe targeted liquid chromatography-tandem mass spectrometry-based metabolomics methods for analyzing microbially derived short-chain fatty acids and neurotransmitters from these samples. Unlike other protocols that commonly examine only stool samples, this protocol includes bacterial cultures, organoid cultures and in vivo samples, in addition to monitoring the metabolite content of stool samples. The incorporation of three experimental models (microbes, organoids and animals) enhances the impact of this protocol. The protocol requires 3 weeks of murine colonization with microbes and ~1-2 weeks for liquid chromatography-tandem mass spectrometry-based instrumental and quantitative analysis, and sample post-processing and normalization.
Collapse
|
14
|
Reiss Z, Rob F, Kolar M, Schierova D, Kreisinger J, Jackova Z, Roubalova R, Coufal S, Mihula M, Thon T, Bajer L, Novakova M, Vasatko M, Kostovcikova K, Galanova N, Lukas M, Kverka M, Tresnak Hercogova J, Tlaskalova-Hogenova H, Jiraskova Zakostelska Z. Skin microbiota signature distinguishes IBD patients and reflects skin adverse events during anti-TNF therapy. Front Cell Infect Microbiol 2023; 12:1064537. [PMID: 36704107 PMCID: PMC9872723 DOI: 10.3389/fcimb.2022.1064537] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/01/2022] [Indexed: 01/11/2023] Open
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are two forms of inflammatory bowel disease (IBD), where the role of gut but not skin dysbiosis is well recognized. Inhibitors of TNF have been successful in IBD treatment, but up to a quarter of patients suffer from unpredictable skin adverse events (SkAE). For this purpose, we analyzed temporal dynamics of skin microbiota and serum markers of inflammation and epithelial barrier integrity during anti-TNF therapy and SkAE manifestation in IBD patients. We observed that the skin microbiota signature of IBD patients differs markedly from healthy subjects. In particular, the skin microbiota of CD patients differs significantly from that of UC patients and healthy subjects, mainly in the retroauricular crease. In addition, we showed that anti-TNF-related SkAE are associated with specific shifts in skin microbiota profile and with a decrease in serum levels of L-FABP and I-FABP in IBD patients. For the first time, we showed that shifts in microbial composition in IBD patients are not limited to the gut and that skin microbiota and serum markers of the epithelium barrier may be suitable markers of SkAE during anti-TNF therapy.
Collapse
Affiliation(s)
- Zuzana Reiss
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Filip Rob
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University, University Hospital Bulovka, Prague, Czechia
| | - Martin Kolar
- IBD Clinical and Research Centre ISCARE a.s., Prague, Czechia
| | - Dagmar Schierova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jakub Kreisinger
- Department of Zoology, Faculty of Science, Charles University, Prague, Czechia
| | - Zuzana Jackova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Radka Roubalova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Stepan Coufal
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Mihula
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Tomas Thon
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Lukas Bajer
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia,Department of Gastroenterology and Hepatology, Institute of Clinical and Experimental Medicine, Prague, Czechia
| | - Michaela Novakova
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University, University Hospital Bulovka, Prague, Czechia
| | - Martin Vasatko
- IBD Clinical and Research Centre ISCARE a.s., Prague, Czechia
| | - Klara Kostovcikova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Natalie Galanova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Milan Lukas
- IBD Clinical and Research Centre ISCARE a.s., Prague, Czechia,Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czechia
| | - Miloslav Kverka
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jana Tresnak Hercogova
- Department of Dermatovenerology, Second Faculty of Medicine, Charles University, University Hospital Bulovka, Prague, Czechia,Prof. Hercogova Dermatology, Prague, Czechia
| | | | - Zuzana Jiraskova Zakostelska
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia,*Correspondence: Zuzana Jiraskova Zakostelska,
| |
Collapse
|
15
|
Schwarz A, Philippsen R, Piticchio SG, Hartmann JN, Häsler R, Rose-John S, Schwarz T. Crosstalk between microbiome, regulatory T cells and HCA2 orchestrates the inflammatory response in a murine psoriasis model. Front Immunol 2023; 14:1038689. [PMID: 36891315 PMCID: PMC9986334 DOI: 10.3389/fimmu.2023.1038689] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/02/2023] [Indexed: 02/22/2023] Open
Abstract
The organ-specific microbiome plays a crucial role in tissue homeostasis, among other things by inducing regulatory T cells (Treg). This applies also to the skin and in this setting short chain fatty acids (SCFA) are relevant. It was demonstrated that topical application of SCFA controls the inflammatory response in the psoriasis-like imiquimod (IMQ)-induced murine skin inflammation model. Since SCFA signal via HCA2, a G-protein coupled receptor, and HCA2 expression is reduced in human lesional psoriatic skin, we studied the effect of HCA2 in this model. HCA2 knock-out (HCA2-KO) mice reacted to IMQ with stronger inflammation, presumably due to an impaired function of Treg. Surprisingly, injection of Treg from HCA2-KO mice even enhanced the IMQ reaction, suggesting that in the absence of HCA2 Treg switch from a suppressive into a proinflammatory type. HCA2-KO mice differed in the composition of the skin microbiome from wild type mice. Co-housing reversed the exaggerated response to IMQ and prevented the alteration of Treg, implying that the microbiome dictates the outcome of the inflammatory reaction. The switch of Treg into a proinflammatory type in HCA2-KO mice could be a downstream phenomenon. This opens the opportunity to reduce the inflammatory tendency in psoriasis by altering the skin microbiome.
Collapse
Affiliation(s)
- Agatha Schwarz
- Department of Dermatology and Allergology, University Kiel, Kiel, Germany
| | - Rebecca Philippsen
- Department of Dermatology and Allergology, University Kiel, Kiel, Germany
| | - Serena G Piticchio
- Institute of Clinical Molecular Biology (IKMB), University Kiel, Kiel, Germany.,Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain
| | - Jan N Hartmann
- Department of Dermatology and Allergology, University Kiel, Kiel, Germany
| | - Robert Häsler
- Department of Dermatology and Allergology, University Kiel, Kiel, Germany
| | | | - Thomas Schwarz
- Department of Dermatology and Allergology, University Kiel, Kiel, Germany
| |
Collapse
|
16
|
Controne I, Scoditti E, Buja A, Pacifico A, Kridin K, Fabbro MD, Garbarino S, Damiani G. Do Sleep Disorders and Western Diet Influence Psoriasis? A Scoping Review. Nutrients 2022; 14:nu14204324. [PMID: 36297008 PMCID: PMC9608488 DOI: 10.3390/nu14204324] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022] Open
Abstract
Western diet may trigger sleep disorders and vice versa, but their single and mutual effects on systemic inflammatory diseases (i.e., psoriasis) are far from being fully elucidated. At the same time, psoriatic patients display a great burden of sleep disorders and dysmetabolisms related to an unhealthy lifestyle (i.e., diet). These patients are also affected by a chronic disorder deeply modulated by environmental factors (i.e., sleep and diet) capable to influence drug-response and disease progression. Thus, we aimed to summarize the evidence in the literature that may highlight a potential link among psoriasis–diet–sleep in order to further promote a multidisciplinary approach to psoriatic patients in the scientific community.
Collapse
Affiliation(s)
- Ilaria Controne
- Clinical Dermatology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy
| | - Egeria Scoditti
- Institute of Clinical Physiology, National Research Council (CNR), 73100 Lecce, Italy
| | - Alessandra Buja
- Department of Cardiologic, Vascular and Thoracic Sciences, and Public Health, University of Padova, 35128 Padova, Italy
| | - Alessia Pacifico
- Clinical Dermatology Department, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Khalaf Kridin
- Lübeck Institute of Experimental Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Massimo Del Fabbro
- Clinical Dermatology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy
- IRCCS Fondazione Ca’ Granda IRCCS Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Sergio Garbarino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Maternal/Child Sciences (DINOGMI), University of Genoa, 16132 Genoa, Italy
| | - Giovanni Damiani
- Clinical Dermatology, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-02-662141
| |
Collapse
|
17
|
Su T, Liu F, Kou CX, Liu HB, Xu SJ, Zhang DY, Wei-Xin F, Fang C, Zhang M. Characteristic of decreased bacterial diversity in psoriasis. ARQ BRAS MED VET ZOO 2022. [DOI: 10.1590/1678-4162-12583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Disturbance of commensal intestinal microbiota is related to chronic inflammatory dermatosis. We analyzed the diversity of the gut microbiota to characterize the biological variation of psoriasis (Ps). Significant differences of gut microbiome profiles were revealed in murine model with psoriasis by sequencing 16S rRNA V3-V4 variable region. Group comparisons included the imiquimod cream (IMQ group, n=8), the imiquimod cream and antibiotics (ATB) (PC+IMQ group, n=8) and the healthy control (CTRL group, n=8). The gut microbiota existed in Ps groups including IMQ group and PC+IMQ group encompassed less diversity than controls, which were attributed to decreased presence of several taxa. The two Ps groups were characterized by significant reduction in firmicutes. In this study, microbiota of psoriasis was defined by an increase presence of Bacteroides. After treated with ATB, we found substantial increase of Lactobacillales but significant decrease of Clostridiales and Coriobacteriales. Relative lower abundance of multiple intestinal bacteria was observed in Ps groups. Although part of genera were concomitantly reduced in both IMQ and PC+IMQ conditions, we discovered the specialty of PC+IMQ group samples was that contained lower abundance of beneficial taxa. Characteristics of gut microbiota profiles in Ps mice were comparable to profiles in patients with Ps, which were related to alteration of specific inflammatory proteins in disease groups but were significantly different from control group. Thus, this study emphasizes the role of intestinal microbiota in the pathogenesis of Ps and provides new insight for investigating association between intestinal microbes and immune inflammation.
Collapse
Affiliation(s)
- Tong Su
- Nanjing Medical University, China
| | - Fang Liu
- Hospital affliated to Nanjing University, China
| | | | - Hai-Bo Liu
- Hospital affliated to Nanjing University, China
| | | | | | - Fan Wei-Xin
- Hospital of Nanjing Medical Uniuersity, China
| | | | | |
Collapse
|
18
|
Schett G, Rahman P, Ritchlin C, McInnes IB, Elewaut D, Scher JU. Psoriatic arthritis from a mechanistic perspective. Nat Rev Rheumatol 2022; 18:311-325. [PMID: 35513599 DOI: 10.1038/s41584-022-00776-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 12/13/2022]
Abstract
Psoriatic arthritis (PsA) is part of a group of closely related clinical phenotypes ('psoriatic disease') that is defined by shared molecular pathogenesis resulting in excessive, prolonged inflammation in the various tissues affected, such as the skin, the entheses or the joints. Psoriatic disease comprises a set of specific drivers that promote an aberrant immune response and the consequent development of chronic disease that necessitates therapeutic intervention. These drivers include genetic, biomechanical, metabolic and microbial factors that facilitate a robust and continuous mobilization, trafficking and homing of immune cells into the target tissues. The role of genetic variants involved in the immune response, the contribution of mechanical factors triggering an exaggerated inflammatory response (mechanoinflammation), the impact of adipose tissue and altered lipid metabolism and the influence of intestinal dysbiosis in the disease process are discussed. Furthermore, the role of key cytokines, such as IL-23, IL-17 and TNF, in orchestrating the various phases of the inflammatory disease process and as therapeutic targets in PsA is reviewed. Finally, the nature and the mechanisms of inflammatory tissue responses inherent to PsA are summarized.
Collapse
Affiliation(s)
- Georg Schett
- Department of Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitaets-klinikum Erlangen, Erlangen, Germany. .,Deutsches Zentrum Immuntherapie DZI, Friedrich Alexander University Erlangen-Nuremberg and Universitaets-klinikum Erlangen, Erlangen, Germany.
| | - Proton Rahman
- St. Clare's Mercy Hospital, St. John's, Newfoundland, Canada
| | - Christopher Ritchlin
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center Rochester, Rochester, NY, USA
| | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Dirk Elewaut
- VIB-UGent Center for Inflammation Research and Ghent University Hospital, Ghent, Belgium
| | - Jose U Scher
- Department of Medicine, Division of Rheumatology, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
19
|
Gut–Skin Axis: Unravelling the Connection between the Gut Microbiome and Psoriasis. Biomedicines 2022; 10:biomedicines10051037. [PMID: 35625774 PMCID: PMC9138548 DOI: 10.3390/biomedicines10051037] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022] Open
Abstract
Evidence has shown that gut microbiome plays a role in modulating the development of diseases beyond the gastrointestinal tract, including skin disorders such as psoriasis. The gut–skin axis refers to the bidirectional relationship between the gut microbiome and skin health. This is regulated through several mechanisms such as inflammatory mediators and the immune system. Dysregulation of microbiota has been seen in numerous inflammatory skin conditions such as atopic dermatitis, rosacea, and psoriasis. Understanding how gut microbiome are involved in regulating skin health may lead to development of novel therapies for these skin disorders through microbiome modulation, in particularly psoriasis. In this review, we will compare the microbiota between psoriasis patients and healthy control, explain the concept of gut–skin axis and the effects of gut dysbiosis on skin physiology. We will also review the current evidence on modulating gut microbiome using probiotics in psoriasis.
Collapse
|
20
|
Garabatos N, Santamaria P. Gut Microbial Antigenic Mimicry in Autoimmunity. Front Immunol 2022; 13:873607. [PMID: 35572569 PMCID: PMC9094498 DOI: 10.3389/fimmu.2022.873607] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota plays a major role in the developmental biology and homeostasis of cells belonging to the adaptive and innate arms of the immune system. Alterations in its composition, which are known to be regulated by both genetic and environmental factors, can either promote or suppress the pathogenic processes underlying the development of various autoimmune diseases, including inflammatory bowel disease, multiple sclerosis, systemic lupus erythematosus, type 1 diabetes and rheumatoid arthritis, to just name a few. Cross-recognition of gut microbial antigens by autoreactive T cells as well as gut microbe-driven alterations in the activation and homeostasis of effector and regulatory T cells have been implicated in this process. Here, we summarize our current understanding of the positive and negative associations between alterations in the composition of the gut microbiota and the development of various autoimmune disorders, with a special emphasis on antigenic mimicry.
Collapse
Affiliation(s)
- Nahir Garabatos
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Pere Santamaria
- Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Julia McFarlane Diabetes Research Centre (JMDRC), Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
21
|
Lousada MB, Lachnit T, Edelkamp J, Paus R, Bosch TCG. Hydra and the hair follicle - An unconventional comparative biology approach to exploring the human holobiont. Bioessays 2022; 44:e2100233. [PMID: 35261041 DOI: 10.1002/bies.202100233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/02/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022]
Abstract
The microbiome of human hair follicles (HFs) has emerged as an important player in different HF and skin pathologies, yet awaits in-depth exploration. This raises questions regarding the tightly linked interactions between host environment, nutrient dependency of host-associated microbes, microbial metabolism, microbe-microbe interactions and host immunity. The use of simple model systems facilitates addressing generally important questions and testing overarching, therapeutically relevant principles that likely transcend obvious interspecies differences. Here, we evaluate the potential of the freshwater polyp Hydra, to dissect fundamental principles of microbiome regulation by the host, that is the human HF. In particular, we focus on therapeutically targetable host-microbiome interactions, such as nutrient dependency, microbial interactions and host defence. Offering a new lens into the study of HF - microbiota interactions, we argue that general principles of how Hydra manages its microbiota can inform the development of novel, microbiome-targeting therapeutic interventions in human skin disease.
Collapse
Affiliation(s)
- Marta B Lousada
- Monasterium Laboratory Skin & Hair Research, Münster, Germany.,Zoological Institute, Christian-Albrechts, University of Kiel, Kiel, Germany
| | - Tim Lachnit
- Zoological Institute, Christian-Albrechts, University of Kiel, Kiel, Germany
| | - Janin Edelkamp
- Monasterium Laboratory Skin & Hair Research, Münster, Germany
| | - Ralf Paus
- Monasterium Laboratory Skin & Hair Research, Münster, Germany.,Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Thomas C G Bosch
- Zoological Institute, Christian-Albrechts, University of Kiel, Kiel, Germany
| |
Collapse
|
22
|
Pereira MS, Redanz S, Kriegel MA. Skin Deep: The Role of the Microbiota in Cutaneous Autoimmunity. J Invest Dermatol 2022; 142:834-840. [PMID: 35027173 DOI: 10.1016/j.jid.2021.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 12/16/2022]
Abstract
The skin microbiota is thought to possibly contribute to the pathogenesis of skin autoimmune diseases. The gut microbiota affects systemically the development and function of the immune system, thereby potentially influencing cutaneous autoimmunity as well. In this paper, we review the role of the gut and skin microbiota in cutaneous autoimmune diseases. Besides direct inflammatory effects at the skin barrier, microbiota may contribute to the pathogenesis of skin autoimmune diseases by metabolites, recall immune cell responses, and permeation of antigens to the subepidermal space. Skin and gut barrier dysfunction may represent a common pathophysiologic process allowing microbiota or its particles to promote autoimmune diseases at barrier surfaces.
Collapse
Affiliation(s)
- Márcia S Pereira
- Department of Translational Rheumatology and Immunology, Institute of Musculoskeletal Medicine, University of Münster, Münster, Germany
| | - Sylvio Redanz
- Department of Translational Rheumatology and Immunology, Institute of Musculoskeletal Medicine, University of Münster, Münster, Germany
| | - Martin A Kriegel
- Department of Translational Rheumatology and Immunology, Institute of Musculoskeletal Medicine, University of Münster, Münster, Germany; Section of Rheumatology and Clinical Immunology, Department of Medicine, University Hospital Münster, Münster, Germany; Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
23
|
Ezeji JC, Sarikonda DK, Hopperton A, Erkkila HL, Cohen DE, Martinez SP, Cominelli F, Kuwahara T, Dichosa AEK, Good CE, Jacobs MR, Khoretonenko M, Veloo A, Rodriguez-Palacios A. Parabacteroides distasonis: intriguing aerotolerant gut anaerobe with emerging antimicrobial resistance and pathogenic and probiotic roles in human health. Gut Microbes 2022; 13:1922241. [PMID: 34196581 PMCID: PMC8253142 DOI: 10.1080/19490976.2021.1922241] [Citation(s) in RCA: 138] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Parabacteroides distasonis is the type strain for the genus Parabacteroides, a group of gram-negative anaerobic bacteria that commonly colonize the gastrointestinal tract of numerous species. First isolated in the 1930s from a clinical specimen as Bacteroides distasonis, the strain was re-classified to form the new genus Parabacteroides in 2006. Currently, the genus consists of 15 species, 10 of which are listed as 'validly named' (P. acidifaciens, P. chartae, P. chinchillae, P. chongii, P. distasonis, P. faecis, P. goldsteinii, P. gordonii, P. johnsonii, and P. merdae) and 5 'not validly named' (P. bouchesdurhonensis, P. massiliensis, P. pacaensis, P. provencensis, and P. timonensis) by the List of Prokaryotic names with Standing in Nomenclature. The Parabacteroides genus has been associated with reports of both beneficial and pathogenic effects in human health. Herein, we review the literature on the history, ecology, diseases, antimicrobial resistance, and genetics of this bacterium, illustrating the effects of P. distasonis on human and animal health.
Collapse
Affiliation(s)
- Jessica C. Ezeji
- Digestive Diseases Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Division of Gastroenterology and Liver Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Daven K. Sarikonda
- Digestive Diseases Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Division of Gastroenterology and Liver Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Austin Hopperton
- Digestive Diseases Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Division of Gastroenterology and Liver Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - Hailey L. Erkkila
- Digestive Diseases Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Division of Gastroenterology and Liver Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Daniel E. Cohen
- Digestive Diseases Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Division of Gastroenterology and Liver Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | - Fabio Cominelli
- Digestive Diseases Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Division of Gastroenterology and Liver Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA,Germ-Free and Gut Microbiome Core, Case Western Reserve University, Cleveland, OH, United States
| | - Tomomi Kuwahara
- Department of Microbiology, Faculty of Medicine, Kagawa University, Miki, Kagawa, Japan
| | - Armand E. K. Dichosa
- B-10 Biosecurity and Public Health, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Caryn E. Good
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Michael R. Jacobs
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | | | - Alida Veloo
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Alexander Rodriguez-Palacios
- Digestive Diseases Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Division of Gastroenterology and Liver Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA,Germ-Free and Gut Microbiome Core, Case Western Reserve University, Cleveland, OH, United States,University Hospitals Research and Education Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA,CONTACT Alexander Rodriguez-Palacios Digestive Diseases Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
24
|
Mouse Models of Psoriasis: A Comprehensive Review. J Invest Dermatol 2021; 142:884-897. [PMID: 34953514 DOI: 10.1016/j.jid.2021.06.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/26/2021] [Accepted: 06/15/2021] [Indexed: 01/20/2023]
Abstract
The use of preclinical animal models of psoriasis has significantly increased over the last three decades, with each model having unique strengths and limitations. Some models translate better to human disease, and many have provided unique insight into psoriasis disease pathogenesis. In this comprehensive review, we present a comparative description and discussion of genetic mouse models, xenograft approaches, and elicited methods using cytokine injections into and topical imiquimod onto mice. We provide an inclusive list of genetically modified animals that have had imiquimod applied to or cytokines injected into their skin and describe the outcomes of these manipulations. This review will provide a valuable resource for those interested in working with psoriasis animal models.
Collapse
|
25
|
Chen HL, Zeng YB, Zhang ZY, Kong CY, Zhang SL, Li ZM, Huang JT, Xu YY, Mao YQ, Cai PR, Han B, Wang WQ, Wang LS. Gut and Cutaneous Microbiome Featuring Abundance of Lactobacillus reuteri Protected Against Psoriasis-Like Inflammation in Mice. J Inflamm Res 2021; 14:6175-6190. [PMID: 34853526 PMCID: PMC8627893 DOI: 10.2147/jir.s337031] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/06/2021] [Indexed: 11/23/2022] Open
Abstract
Background Psoriasis is a chronic autoinflammatory skin disease, and its aetiology remains incompletely understood. Recently, gut microbial dysbiosis is found to be tightly associated with psoriasis. Objective We sought to reveal the causal role of gut microbiota dysbiosis in psoriasis pathogenesis and investigate the protective effect of healthy commensal bacteria against imiquimod -induced psoriasis-like skin response. Methods By using fecal microbial transplantation (FMT), 16S rRNA gene-based taxonomic profiling and Lactobacillus supplement, we have assessed the effect of FMT from healthy individuals on psoriasis-like skin inflammation and associated immune disorders in imiquimod-induced psoriasis mice. Results Here, by using psoriasis mice humanized with the stools from healthy donors and psoriasis patients, the imiquimod-induced psoriasis in mice with psoriasis patient stool was found to be significantly aggravated as compared to the mice with healthy donor stools. Further analysis showed fecal microbiota of healthy individuals protected against Treg/Th17 imbalance in psoriasis. Moreover, we found the gut and skin microbiome in mice receipted with gut microbiota of healthy individuals (HD) differed from those of mice receipted with gut microbiota of psoriasis patients (PSD). 16S rRNA sequencing revealed that Lactobacillus reuteri was greatly enriched in fecal and cutaneous microbiome of HD mice as compared to PSD mice. Intriguingly, supplement with Lactobacillus reuteri was sufficient to increase the expression of anti-inflammatory gene IL-10, reduce Th17 cells counts and confer resistance to imiquimod-induced inflammation on the mice with gut microbiota dysbiosis. Conclusion Our results suggested that the gut microbiota dysbiosis is the potential causal factor for psoriasis and the gut microbiota may serve as promising therapy target for psoriasis patients.
Collapse
Affiliation(s)
- Hui-Ling Chen
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Yi-Bin Zeng
- Dermatological Department, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Zheng-Yan Zhang
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Chao-Yue Kong
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Shi-Long Zhang
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Zhan-Ming Li
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Jia-Ting Huang
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Ya-Yun Xu
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Yu-Qin Mao
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Pei-Ran Cai
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Bing Han
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Wu-Qing Wang
- Dermatological Department, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| | - Li-Shun Wang
- Center for Traditional Chinese Medicine and Gut Microbiota, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China.,Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, 201100, People's Republic of China
| |
Collapse
|
26
|
Fernández-Gallego N, Sánchez-Madrid F, Cibrian D. Role of AHR Ligands in Skin Homeostasis and Cutaneous Inflammation. Cells 2021; 10:cells10113176. [PMID: 34831399 PMCID: PMC8622815 DOI: 10.3390/cells10113176] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 02/07/2023] Open
Abstract
Aryl hydrocarbon receptor (AHR) is an important regulator of skin barrier function. It also controls immune-mediated skin responses. The AHR modulates various physiological functions by acting as a sensor that mediates environment–cell interactions, particularly during immune and inflammatory responses. Diverse experimental systems have been used to assess the AHR’s role in skin inflammation, including in vitro assays of keratinocyte stimulation and murine models of psoriasis and atopic dermatitis. Similar approaches have addressed the role of AHR ligands, e.g., TCDD, FICZ, and microbiota-derived metabolites, in skin homeostasis and pathology. Tapinarof is a novel AHR-modulating agent that inhibits skin inflammation and enhances skin barrier function. The topical application of tapinarof is being evaluated in clinical trials to treat psoriasis and atopic dermatitis. In the present review, we summarize the effects of natural and synthetic AHR ligands in keratinocytes and inflammatory cells, and their relevance in normal skin homeostasis and cutaneous inflammatory diseases.
Collapse
Affiliation(s)
- Nieves Fernández-Gallego
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), 28006 Madrid, Spain;
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Francisco Sánchez-Madrid
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), 28006 Madrid, Spain;
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (F.S.-M.); (D.C.)
| | - Danay Cibrian
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa (IIS-IP), 28006 Madrid, Spain;
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (F.S.-M.); (D.C.)
| |
Collapse
|
27
|
Li CY, Dai YX, Chang YT, Bai YM, Tsai SJ, Chen TJ, Chen MH. Proton Pump Inhibitors Are Associated with Increased Risk of Psoriasis: A Nationwide Nested Case-Control Study. Dermatology 2021; 237:884-890. [PMID: 34474411 DOI: 10.1159/000517515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/31/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Proton pump inhibitors (PPIs) are among the most widely used drugs. Little is known about the association between PPI use and risk of psoriasis. OBJECTIVE To investigate the association between PPI use and subsequent psoriasis risk. METHODS We included participants from the Taiwan National Health Insurance Research Database. Patients with PPI use and an incidence of psoriasis (n = 5,756) were assigned to the case cohort and 1:1 matched to controls. PPI use was defined as >30 cumulative defined daily doses (cDDDs); PPI nonuse was defined as ≤30 cDDDs. Logistic regression was used for the analyses. RESULTS There was a significant association between PPI use and psoriasis risk. The confounder-adjusted odd ratios (95% confidence interval [CI]) were 1.52 (1.31-1.76) and 1.54 (1.22-1.93) for patients with 120-365 cDDDs and >365 cDDDs, respectively, compared with PPI nonusers. Stratified analyses based on PPI type showed that exposure to lansoprazole (OR, 1.25; 95% CI, 1.11-1.41) was associated with subsequent psoriasis risk. CONCLUSIONS PPI use might be associated with an increased risk of developing psoriasis or as an epiphenomenon. Further prospective studies are needed to elucidate the association and underlying mechanisms.
Collapse
Affiliation(s)
- Cheng-Yuan Li
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Xiu Dai
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yun-Ting Chang
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ya-Mei Bai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Jen Tsai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzeng-Ji Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Hospital and Health Care Administration, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mu-Hong Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
28
|
Potential Role of Probiotics in Ameliorating Psoriasis by Modulating Gut Microbiota in Imiquimod-Induced Psoriasis-Like Mice. Nutrients 2021; 13:nu13062010. [PMID: 34207960 PMCID: PMC8230682 DOI: 10.3390/nu13062010] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/02/2021] [Accepted: 06/06/2021] [Indexed: 12/14/2022] Open
Abstract
Psoriasis is an immune-mediated systemic disease that may be treated with probiotics. In this study, probiotic strains that could or could not decrease interleukin (IL)-17 levels were applied to imiquimod (IMQ)-induced psoriasis-like mice via oral administration. Bifidobacterium adolescentis CCFM667, B. breve CCFM1078, Lacticaseibacillus paracasei CCFM1074, and Limosilactobacillus reuteri CCFM1132 ameliorated psoriasis-like pathological characteristics and suppressed the release of IL-23/T helper cell 17 (Th17) axis-related inflammatory cytokines, whereas B. animalis CCFM1148, L. paracasei CCFM1147, and L. reuteri CCFM1040 neither alleviated the pathological characteristics nor reduced the levels of inflammatory cytokines. All effective strains increased the contents of short-chain fatty acids, which were negatively correlated with the levels of inflammatory cytokines. By performing 16S rRNA gene sequencing, the diversity of gut microbiota in psoriasis-like mice was found to decrease, but all effective strains made some specific changes to the composition of gut microbiota compared to the ineffective strains. Furthermore, except for B. breve CCFM1078, all other effective strains decreased the abundance of the family Rikenellaceae, which was positively correlated with psoriasis-like pathological characteristics and was negatively correlated with propionate levels. These findings demonstrated effects of strain-specificity, and how probiotics ameliorated psoriasis and provide new possibilities for the treatment of psoriasis.
Collapse
|
29
|
Shinno-Hashimoto H, Hashimoto Y, Wei Y, Chang L, Fujita Y, Ishima T, Matsue H, Hashimoto K. Abnormal composition of microbiota in the gut and skin of imiquimod-treated mice. Sci Rep 2021; 11:11265. [PMID: 34050205 PMCID: PMC8163751 DOI: 10.1038/s41598-021-90480-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/12/2021] [Indexed: 02/07/2023] Open
Abstract
Psoriasis is a chronic, inflammatory skin disease. Although the precise etiology of psoriasis remains unclear, gut-microbiota axis might play a role in the pathogenesis of the disease. Here we investigated whether the composition of microbiota in the intestine and skin is altered in the imiquimod (IMQ)-treated mouse model of psoriasis. Topical application of IMQ to back skin caused significant changes in the composition of microbiota in the intestine and skin of IMQ-treated mice compared to control mice. The LEfSe algorithm identified the species Staphylococcus lentus as potential skin microbial marker for IMQ group. Furthermore, there were correlations for several microbes between the intestine and skin, suggesting a role of skin-gut-microbiota in IMQ-treated mice. Levels of succinic acid and lactic acid in feces from IMQ-treated mice were significantly higher than control mice. Moreover, the predictive functional analysis of the microbiota in the intestine and skin showed that IMQ caused alterations in several KEGG pathways. In conclusion, the current data indicated that topical application with IMQ to skin alters the composition of the microbiota in the gut and skin of host. It is likely that skin-gut microbiota axis plays a role in pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Hiroyo Shinno-Hashimoto
- Department of Dermatology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Yaeko Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
- Department of Respirology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
| | - Yan Wei
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lijia Chang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Yuko Fujita
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Tamaki Ishima
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan
| | - Hiroyuki Matsue
- Department of Dermatology, Chiba University Graduate School of Medicine, Chiba, 260-8670, Japan
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
30
|
Ogawa C, Inoue R, Yonejima Y, Hisa K, Yamamoto Y, Suzuki T. Supplemental Leuconostoc mesenteroides strain NTM048 attenuates imiquimod-induced psoriasis in mice. J Appl Microbiol 2021; 131:3043-3055. [PMID: 34028144 DOI: 10.1111/jam.15161] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/05/2021] [Accepted: 05/19/2021] [Indexed: 12/12/2022]
Abstract
AIMS Psoriasis, a chronic inflammatory skin disease, is associated with altered intestinal microbiota. Here, we investigated the ameliorative effect of Leuconostoc mesenteroides NTM048 strain in imiquimod (IMQ)-induced psoriasis in mice. METHODS AND RESULTS Mice were administered NTM048 for 21 days alongside the topical application of IMQ on the dorsal skin for 6 consecutive days. IMQ induced psoriatic symptoms such as erythema and scaling and also upregulated interleukin (IL)-17, a key effector cytokine of psoriasis, in the skin. Supplemental NTM048 suppressed these abnormalities, increased the levels of plasma deoxycholic acid (DCA), a secondary bile acid and altered the faecal microbiota composition, as indicated by the increased abundance of Akkermansia and decreased abundance of Staphylococcus and Streptococcus. Notably, DCA treatment of murine splenocytes reduced IL-17 production. CONCLUSIONS The NTM048-mediated reduction of psoriasis was shown to involve the downregulation of IL-17 in mouse skin, which was possibly associated with the plasma DCA derived from intestinal microbiota. SIGNIFICANCE AND IMPACT OF THE STUDY Our findings propose not only a novel approach for psoriasis reduction but also a crosstalk between the skin and intestine in psoriasis.
Collapse
Affiliation(s)
- C Ogawa
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - R Inoue
- Faculty of Agriculture, Setsunan University, Hirakata, Japan
| | | | | | - Y Yamamoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - T Suzuki
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan.,Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima, Japan
| |
Collapse
|
31
|
Contribution of Infectious Agents to the Development of Celiac Disease. Microorganisms 2021; 9:microorganisms9030547. [PMID: 33800833 PMCID: PMC8001938 DOI: 10.3390/microorganisms9030547] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
The ingestion of wheat gliadin (alcohol-soluble proteins, an integral part of wheat gluten) and related proteins induce, in genetically predisposed individuals, celiac disease (CD), which is characterized by immune-mediated impairment of the small intestinal mucosa. The lifelong omission of gluten and related grain proteins, i.e., a gluten-free diet (GFD), is at present the only therapy for CD. Although a GFD usually reduces CD symptoms, it does not entirely restore the small intestinal mucosa to a fully healthy state. Recently, the participation of microbial components in pathogenetic mechanisms of celiac disease was suggested. The present review provides information on infectious diseases associated with CD and the putative role of infections in CD development. Moreover, the involvement of the microbiota as a factor contributing to pathological changes in the intestine is discussed. Attention is paid to the mechanisms by which microbes and their components affect mucosal immunity, including tolerance to food antigens. Modulation of microbiota composition and function and the potential beneficial effects of probiotics in celiac disease are discussed.
Collapse
|
32
|
Afonina IS, Van Nuffel E, Beyaert R. Immune responses and therapeutic options in psoriasis. Cell Mol Life Sci 2021; 78:2709-2727. [PMID: 33386888 PMCID: PMC11072277 DOI: 10.1007/s00018-020-03726-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 11/23/2020] [Accepted: 11/28/2020] [Indexed: 02/07/2023]
Abstract
Psoriasis is a chronic inflammatory disease of the skin that affects about 2-3% of the population and greatly impairs the quality of life of affected individuals. Psoriatic skin is characterized by excessive proliferation and aberrant differentiation of keratinocytes, as well as redness caused by increased dilation of the dermal blood vessels and infiltration of immune cells. Although the pathogenesis of psoriasis has not yet been completely elucidated, it is generally believed to arise from a complex interplay between hyperproliferating keratinocytes and infiltrating, activated immune cells. So far, the exact triggers that elicit this disease are still enigmatic, yet, it is clear that genetic predisposition significantly contributes to the development of psoriasis. In this review, we summarize current knowledge of important cellular and molecular mechanisms driving the initiation and amplification stages of psoriasis development, with a particular focus on cytokines and emerging evidence illustrating keratinocyte-intrinsic defects as key drivers of inflammation. We also discuss mouse models that have contributed to a better understanding of psoriasis pathogenesis and the preclinical development of novel therapeutics, including monoclonal antibodies against specific cytokines or cytokine receptors that have revolutionized the treatment of psoriasis. Future perspectives that may have the potential to push basic research and open up new avenues for therapeutic intervention are provided.
Collapse
Affiliation(s)
- Inna S Afonina
- Unit of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, Ghent University - VIB, Technologiepark 71, B-9052, Ghent, Belgium
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 71, B-9052, Ghent, Belgium
| | - Elien Van Nuffel
- Unit of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, Ghent University - VIB, Technologiepark 71, B-9052, Ghent, Belgium
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 71, B-9052, Ghent, Belgium
| | - Rudi Beyaert
- Unit of Molecular Signal Transduction in Inflammation, Center for Inflammation Research, Ghent University - VIB, Technologiepark 71, B-9052, Ghent, Belgium.
- Department for Biomedical Molecular Biology, Ghent University, Technologiepark 71, B-9052, Ghent, Belgium.
| |
Collapse
|
33
|
Shi Z, Wu X, Santos Rocha C, Rolston M, Garcia-Melchor E, Huynh M, Nguyen M, Law T, Haas KN, Yamada D, Millar NL, Wan YJY, Dandekar S, Hwang ST. Short-Term Western Diet Intake Promotes IL-23‒Mediated Skin and Joint Inflammation Accompanied by Changes to the Gut Microbiota in Mice. J Invest Dermatol 2021; 141:1780-1791. [PMID: 33485880 DOI: 10.1016/j.jid.2020.11.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 02/08/2023]
Abstract
We previously showed that exposure to a high-sugar and moderate-fat diet (i.e., Western diet [WD]) in mice induces appreciable skin inflammation and enhances the susceptibility to imiquimod-induced psoriasiform dermatitis, suggesting that dietary components may render the skin susceptible to psoriatic inflammation. In this study, utilizing an IL-23 minicircle-based model with features of both psoriasiform dermatitis and psoriatic arthritis, we showed that intake of WD for 10 weeks predisposed mice not only to skin but also to joint inflammation. Both WD-induced skin and joint injuries were associated with an expansion of IL-17A‒producing γδ T cells and increased expression of T helper type 17 cytokines. After IL-23 minicircle delivery, WD-fed mice had reduced microbial diversity and pronounced dysbiosis. Treatment with broad-spectrum antibiotics suppressed IL-23‒mediated skin and joint inflammation in the WD-fed mice. Strikingly, reduced skin and joint inflammation with a partial reversion of the gut microbiota were noted when mice switched from a WD to a standard diet after IL-23 minicircle delivery. These findings reveal that a short-term WD intake‒induced dysbiosis is accompanied by enhanced psoriasis-like skin and joint inflammation. Modifications toward a healthier dietary pattern should be considered in patients with psoriatic skin and/or joint disease.
Collapse
Affiliation(s)
- Zhenrui Shi
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA; Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuesong Wu
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
| | - Clarissa Santos Rocha
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Sacramento, California, USA
| | - Matthew Rolston
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Sacramento, California, USA
| | - Emma Garcia-Melchor
- Institute of Infection, Immunity & Inflammation, College of Medicine, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mindy Huynh
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
| | - Mimi Nguyen
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
| | - Timothy Law
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
| | - Kelly N Haas
- Biology Department, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Daisuke Yamada
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
| | - Neal L Millar
- Institute of Infection, Immunity & Inflammation, College of Medicine, Veterinary & Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Yu-Jui Yvonne Wan
- Department of Medical Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, California, USA
| | - Satya Dandekar
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Sacramento, California, USA
| | - Samuel T Hwang
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA.
| |
Collapse
|
34
|
Yegorov S, Babenko D, Kozhakhmetov S, Akhmaltdinova L, Kadyrova I, Nurgozhina A, Nurgaziyev M, Good SV, Hortelano GH, Yermekbayeva B, Kushugulova A. Psoriasis Is Associated With Elevated Gut IL-1α and Intestinal Microbiome Alterations. Front Immunol 2020; 11:571319. [PMID: 33117362 PMCID: PMC7559734 DOI: 10.3389/fimmu.2020.571319] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/02/2020] [Indexed: 12/20/2022] Open
Abstract
Background Psoriasis is a chronic inflammatory condition that predominantly affects the skin and is associated with extracutaneous disorders, such as inflammatory bowel disease and arthritis. Changes in gut immunology and microbiota are important drivers of proinflammatory disorders and could play a role in the pathogenesis of psoriasis. Therefore, we explored whether psoriasis in a Central Asian cohort is associated with alterations in select immunological markers and/or microbiota of the gut. Methods We undertook a case-control study of stool samples collected from outpatients, aged 30-45 years, of a dermatology clinic in Kazakhstan presenting with plaque, guttate, or palmoplantar psoriasis (n = 20), and age-sex matched subjects without psoriasis (n = 20). Stool supernatant was subjected to multiplex ELISA to assess the concentration of 47 cytokines and immunoglobulins and to 16S rRNA gene sequencing to characterize microbial diversity in both psoriasis participants and controls. Results The psoriasis group tended to have higher concentrations of most analytes in stool (29/47 = 61.7%) and gut IL-1α was significantly elevated (4.19-fold, p = 0.007) compared to controls. Levels of gut IL-1α in the psoriasis participants remained significantly unaltered up to 3 months after the first sampling (p = 0.430). Psoriasis was associated with alterations in gut Firmicutes, including elevated Faecalibacterium and decreased Oscillibacter and Roseburia abundance, but no association was observed between gut microbial diversity or Firmicutes/Bacteroidetes ratios and disease status. Conclusions Psoriasis may be associated with gut inflammation and dysbiosis. Studies are warranted to explore the use of gut microbiome-focused therapies in the management of psoriasis in this under-studied population.
Collapse
Affiliation(s)
- Sergey Yegorov
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan.,Faculty of Education and Humanities, Suleyman Demirel University, Almaty, Kazakhstan
| | - Dmitriy Babenko
- Karaganda Medical University Research Centre, Karaganda, Kazakhstan.,Art Science LLP Innovative Center, Nur-Sultan, Kazakhstan
| | - Samat Kozhakhmetov
- Laboratory of Human Microbiome and Longevity, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | | | - Irina Kadyrova
- Karaganda Medical University Research Centre, Karaganda, Kazakhstan
| | - Ayaulym Nurgozhina
- Laboratory of Human Microbiome and Longevity, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Madiyar Nurgaziyev
- Laboratory of Human Microbiome and Longevity, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Sara V Good
- Department of Biology, The University of Winnipeg, Winnipeg, MB, Canada
| | - Gonzalo H Hortelano
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | | | - Almagul Kushugulova
- Laboratory of Human Microbiome and Longevity, National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
35
|
Owczarczyk-Saczonek A, Purzycka-Bohdan D, Nedoszytko B, Reich A, Szczerkowska-Dobosz A, Bartosiñska J, Batycka-Baran A, Czajkowski R, Dobrucki IT, Dobrucki LW, Górecka-Sokołowska M, Janaszak-Jasiecka A, Kalinowski L, Krasowska D, Radulska A, Reszka E, Samotij D, Słominski A, Słominski R, Sobalska-Kwapis M, Stawczyk-Macieja M, Strapagiel D, Szczêch J, Żmijewski M, Nowicki RJ. Pathogenesis of psoriasis in the "omic" era. Part III. Metabolic disorders, metabolomics, nutrigenomics in psoriasis. Postepy Dermatol Alergol 2020; 37:452-467. [PMID: 32994764 PMCID: PMC7507147 DOI: 10.5114/ada.2020.98284] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/28/2020] [Indexed: 12/15/2022] Open
Abstract
Psoriasis is a systemic disease that is strictly connected with metabolic disorders (insulin resistance, atherogenic dyslipidemia, arterial hypertension, and cardiovascular diseases). It occurs more often in patients with a more severe course of the disease. Obesity is specially an independent risk factor and it is associated with a worse treatment outcome because of the high inflammatory activity of visceral fatty tissue and the production of inflammatory mediators involved in the development of both psoriasis and metabolic disorders. However, in psoriasis the activation of the Th17/IL-17 and the abnormalities in the Th17/Treg balance axis are observed, but this pathomechanism does not fully explain the frequent occurrence of metabolic disorders. Therefore, there is a need to look for better biomarkers in the diagnosis, prognosis and monitoring of concomitant disorders and therapeutic effects in psoriasis. In addition, the education on the use of a proper diet as a prophylaxis for the development of the above disorders is an important element of holistic care for a patient with psoriasis. Diet may affect gene expression due to epigenetic modification which encompasses interactions of environment, nutrition and diseases. Patients with psoriasis should be advised to adopt proper diet and dietician support.
Collapse
Affiliation(s)
- Agnieszka Owczarczyk-Saczonek
- Chair and Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Dorota Purzycka-Bohdan
- Chair and Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Bogusław Nedoszytko
- Chair and Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Adam Reich
- Department of Dermatology, University of Rzeszow, Rzeszow, Poland
| | - Aneta Szczerkowska-Dobosz
- Chair and Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Joanna Bartosiñska
- Chair and Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Aleksandra Batycka-Baran
- Chair and Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Rafał Czajkowski
- Chair and Department of Dermatology, Sexually Transmitted Diseases and Immunodermatology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Iwona T. Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
| | - Lawrence W. Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdansk, Poland
| | | | - Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, Lodz, Poland
- Gdansk University of Technology, Gdansk, Poland
| | - Dorota Krasowska
- Chair and Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Adrianna Radulska
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Edyta Reszka
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Dominik Samotij
- Department of Dermatology, University of Rzeszow, Rzeszow, Poland
| | - Andrzej Słominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, USA
| | - Radomir Słominski
- Department of Medicine and Microbiology, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, USA
| | | | - Marta Stawczyk-Macieja
- Chair and Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | | | - Justyna Szczêch
- Department of Dermatology, University of Rzeszow, Rzeszow, Poland
| | - Michał Żmijewski
- Department of Histology, Medical University of Gdansk, Gdansk, Poland
| | - Roman J. Nowicki
- Chair and Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
36
|
Metagenomic analysis of gut microbiota in non-treated plaque psoriasis patients stratified by disease severity: development of a new Psoriasis-Microbiome Index. Sci Rep 2020; 10:12754. [PMID: 32728075 PMCID: PMC7391695 DOI: 10.1038/s41598-020-69537-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
Psoriasis is an immune-mediated skin disorder. Imbalance of gut microbial populations has been implicated in many diseases. We aimed to investigate whether there were differences in gut microbiota in psoriasis patients vs non-psoriasis controls and between psoriasis severity groups. 55 psoriasis patients and 27 controls were included. V3–V4 regions of the 16S rRNA gene of fecal samples were analyzed using Illumina MiSeq. Bioinformatic analysis was performed. We found changes in gut microbiome composition depending on their psoriasis status as determined by weighted unifrac (p < 0.05), in particular an increase in Firmicutes and depletion of Bacteroidetes in psoriasis patients. Additionally, the Faecalibacterium and Blautia genus were higher in psoriasis patients while Bacteroides and Paraprevotella in non-psoriasis controls (p < 0.05, LDA score > 2). Moderate-to-severe psoriasis patients had lower biodiversity than mild psoriatic patients (p = 0.049). No differences for beta-diversity were found. We developed a Psoriasis-Microbiota Index (PMI), which discriminated among psoriasis patients and controls with sensitivity: 0.78 and specificity: 0.79. Furthermore, we performed a meta-analysis with published data to validate this index. We demonstrated gut dysbiosis in psoriasis patients, suggesting a role in psoriasis pathophysiology. Furthermore, we developed a PMI with the potential to discriminate between psoriasis patients and controls across different populations, which could be used as a biomarker in the clinical practice.
Collapse
|
37
|
Sikora M, Stec A, Chrabaszcz M, Knot A, Waskiel-Burnat A, Rakowska A, Olszewska M, Rudnicka L. Gut Microbiome in Psoriasis: An Updated Review. Pathogens 2020; 9:pathogens9060463. [PMID: 32545459 PMCID: PMC7350295 DOI: 10.3390/pathogens9060463] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/29/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
(1) Background: A growing body of evidence highlights that intestinal dysbiosis is associated with the development of psoriasis. The gut–skin axis is the novel concept of the interaction between skin diseases and microbiome through inflammatory mediators, metabolites and the intestinal barrier. The objective of this study was to synthesize current data on the gut microbial composition in psoriasis. (2) Methods: We conducted a systematic review of studies investigating intestinal microbiome in psoriasis, using the PRISMA checklist. We searched MEDLINE, EMBASE, and Web of Science databases for relevant published articles (2000–2020). (3) Results: All of the 10 retrieved studies reported alterations in the gut microbiome in patients with psoriasis. Eight studies assessed alpha- and beta-diversity. Four of them reported a lack of change in alpha-diversity, but all confirmed significant changes in beta-diversity. At the phylum-level, at least two or more studies reported a lower relative abundance of Bacteroidetes, and higher Firmicutes in psoriasis patients versus healthy controls. (4) Conclusions: There is a significant association between alterations in gut microbial composition and psoriasis; however, there is high heterogeneity between studies. More unified methodological standards in large-scale studies are needed to understand microbiota’s contribution to psoriasis pathogenesis and its modulation as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Mariusz Sikora
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
- Correspondence:
| | - Albert Stec
- Student Research Committee, Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.S.); (A.K.)
| | - Magdalena Chrabaszcz
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Aleksandra Knot
- Student Research Committee, Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.S.); (A.K.)
| | - Anna Waskiel-Burnat
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Adriana Rakowska
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Malgorzata Olszewska
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.C.); (A.W.-B.); (A.R.); (M.O.); (L.R.)
| |
Collapse
|
38
|
Abstract
BACKGROUND Psoriasis is a frequent chronic inflammatory systemic disease. It is associated with changes in the microbiome, which may trigger psoriasis and influence the course of the disease. OBJECTIVE Current methods for detection and the potential role of the microbiome in the pathogenesis of psoriasis are described. MATERIAL AND METHODS A literature search was conducted using the databases Medline and PubMed as well as a general internet and book research. RESULTS Both skin and gut microbiota are involved in the immunopathogenesis and may substantially modulate psoriasis. Antimicrobial peptides may serve as a link between the microbiome and the immunological mechanisms in psoriasis by regulating the microbiome at interfaces and can trigger psoriasis as antigens. Recent innovative methods, such as 16S rRNA next-generation sequencing significantly facilitate microbiome analysis. CONCLUSION The analysis of the microbiome in patients with psoriasis before, during and after treatment provides a basis for the identification of potential biomarkers for predicting individual treatment responses and facilitating the decision for a certain treatment.
Collapse
|