1
|
Jimenez-Sanchez M, Celiberto LS, Yang H, Sham HP, Vallance BA. The gut-skin axis: a bi-directional, microbiota-driven relationship with therapeutic potential. Gut Microbes 2025; 17:2473524. [PMID: 40050613 PMCID: PMC11901370 DOI: 10.1080/19490976.2025.2473524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
This review explores the emerging term "gut-skin axis" (GSA), describing the bidirectional signaling that occurs between the skin and the gastrointestinal tract under both homeostatic and disease conditions. Central to GSA communication are the gut and skin microbiota, the microbial communities that colonize these barrier surfaces. By influencing diverse host pathways, including innate immune, vitamin D receptor, and Aryl hydrocarbon receptor signaling, a balanced microbiota contributes to both tissue homeostasis and host defense. In contrast, microbiota imbalance, or dysbiosis at one site, can lead to local barrier dysfunction, resulting in the activation of signaling pathways that can disrupt tissue homeostasis at the other site, potentially leading to inflammatory skin conditions such as atopic dermatitis and psoriasis, or gut diseases like Inflammatory Bowel Disease. To date, most research on the GSA has examined the impact of the gut microbiota and diet on skin health, but recent studies show that exposing the skin to ultraviolet B-light can beneficially modulate both the gut microbiome and intestinal health. Thus, despite the traditional focus of clinicians and researchers on these organ systems as distinct, the GSA offers new opportunities to better understand the pathogenesis of cutaneous and gastrointestinal diseases and promote health at both sites.
Collapse
Affiliation(s)
- Maira Jimenez-Sanchez
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Larissa S. Celiberto
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Hyungjun Yang
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Ho Pan Sham
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Bruce A. Vallance
- Department of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| |
Collapse
|
2
|
Polonio CM, McHale KA, Sherr DH, Rubenstein D, Quintana FJ. The aryl hydrocarbon receptor: a rehabilitated target for therapeutic immune modulation. Nat Rev Drug Discov 2025:10.1038/s41573-025-01172-x. [PMID: 40247142 DOI: 10.1038/s41573-025-01172-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2025] [Indexed: 04/19/2025]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor originally identified as the target mediating the toxic effects of environmental pollutants including polycyclic aromatic hydrocarbons (PAHs), polychlorinated biphenyls (PCBs) and dioxins. For years, AHR activation was actively avoided during drug development. However, the AHR was later identified as an important physiological regulator of the immune response. These findings triggered a paradigm shift that resulted in identification of the AHR as a regulator of both innate and adaptive immunity and outlined a pathway for its modulation by the diet, commensal flora and metabolism in the context of autoimmunity, cancer and infection. Moreover, the AHR was revealed as a candidate target for the therapeutic modulation of the immune response. Indeed, the first AHR-activating drug (tapinarof) was recently approved for the treatment of psoriasis. Clinical trials are underway to evaluate the effects of tapinarof and other AHR-targeting therapeutics in inflammatory diseases, cancer and infections. This Review outlines the molecular mechanism of AHR action, and describes how it regulates the immune response. We also discuss links to disease and AHR-targeting therapeutics that have been tested in past and ongoing clinical trials.
Collapse
Affiliation(s)
- Carolina M Polonio
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - David H Sherr
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | | | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Kulig P, Brazauskas P, Suffiotti M, Raoult E, Babilonski U, Renault B, Grieder U, Vezzali E, Blattmann P, Martinic MM, Murphy MJ. Efficacy of IDOR-1117-2520, a novel, orally available CCR6 antagonist in preclinical models of skin dermatitis. Br J Pharmacol 2025. [PMID: 40156059 DOI: 10.1111/bph.70025] [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: 10/18/2024] [Revised: 01/22/2025] [Accepted: 02/11/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND AND PURPOSE The chemokine receptor CCR6 guides pathogenic T17 cells, implicated in autoimmune diseases including psoriasis, to sites of inflammation via the chemokine CCL20. Therefor, pharmacological inhibition of CCR6+ immune cell migration provides a novel therapeutic approach. Translatability of such an intervention has not yet been assessed in detail. We evaluated the translatability of the Aldara® mouse model induced skin inflammation to psoriasis, with particular focus on immune cell trafficking and assessed the efficacy of IDOR-1117-2520, a highly selective, potent and orally available CCR6 small inhibitor. EXPERIMENTAL APPROACH Effects of IDOR-1117-2520 were investigated in the Aldara® and IL23 mouse models of skin inflammation using flow cytometry, RNA sequencing and transcriptome-based cell type deconvolution approaches to characterise immune cell migration patterns. These results were compared to human psoriasis transcriptomics data. KEY RESULTS IDOR-1117-2520 dose dependently reduced infiltration of CCR6+ immune cells into inflamed skin, and was equally efficacious as IL-17 and IL-23 inhibition in models of skin inflammation. Pathway analysis showed molecular similarities in the immune response between human psoriasis and the Aldara® mouse model. IL-17/IL-23 pathway genes were expressed in both human psoriasis and the mouse model. CCR6 inhibition modulated multiple pathways associated with inflammation beyond the proximal IL-17/IL-23 pathway. A chemokine-chemokine receptor interaction map implicated CCL20-CCR6 as the dominant axis in recruiting pathogenic T17 cells in both the model and in human psoriasis. CONCLUSION AND IMPLICATIONS IDOR-1117-2520 could provide a promising novel targeted approach to treating psoriasis and, potentially, other autoimmune diseases involving the CCR6/CCL20 axis and the IL-17/IL-23 pathway. IDOR-1117-2520 is currently being evaluated in a clinical phase 1 trial (ISRCTN28892128).
Collapse
Affiliation(s)
- Paulina Kulig
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Pijus Brazauskas
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Madeleine Suffiotti
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Emilie Raoult
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Ulrike Babilonski
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Bérengère Renault
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Ursula Grieder
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Enrico Vezzali
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Peter Blattmann
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Marianne M Martinic
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| | - Mark J Murphy
- Department of Translational and Pharmacological Science, Idorsia Pharmaceuticals Ltd, Allschwil, Switzerland
| |
Collapse
|
4
|
Huang L, Zhou Y, Xiao H, Li Y, Zhou Z, Xiao Z, Tong Y, Hu K, Kuang Y, Shen M, Xiao Y, Chen X. Emerging Contaminants: An Important But Ignored Risk Factor for Psoriasis. Clin Rev Allergy Immunol 2025; 68:33. [PMID: 40121604 DOI: 10.1007/s12016-025-09043-4] [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] [Accepted: 03/03/2025] [Indexed: 03/25/2025]
Abstract
Industrialization and modernization have changed the environment. A group of emerging contaminants (ECs) has been defined recently. Psoriasis, whose incidence has increased in recent years, is a relapsing immune-mediated disease carrying a heavy disease burden. The erythematous scaly plaque is a typical symptom and occurs on several parts of the body. In addition, psoriasis has many comorbidities, such as psoriatic arthritis, diabetes, and depression, damaging the quality of life of patients. IL-17, IL-12, IL-23, and TNF-alpha are important related cytokines. ECs can influence psoriasis through the immune system and inflammatory responses. Specific mechanisms include increasing pro-inflammatory cytokines such as TNF-α and IL-17, and activating immune cells such as macrophages. And for psoriasis patients, it is suggested to reduce the exposure of most ECs. However, the complex mechanisms involved have not been discussed together and concluded. In this review, we summarize the relationship between ECs and psoriasis, focusing on the immune system, especially the immune cells and cytokines. These results can help guide clinical treatment and long-term management of psoriasis.
Collapse
Affiliation(s)
- Leyi Huang
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, China
| | - Yinli Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, China
| | - Hui Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, China
| | - Yajia Li
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, China
| | - Zhiru Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, China
| | - Ziyi Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, China
| | - Yixuan Tong
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, China
| | - Kun Hu
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, China
| | - Yehong Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, China
| | - Minxue Shen
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha, 410008, China
| | - Yi Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, China.
| |
Collapse
|
5
|
Bartley B, Pierce C, Hivnor C, Valdes-Rodriguez R. Topical Medications for Chronic Itch in Older Patients: Navigating a Pressing Need. Drugs Aging 2025; 42:213-233. [PMID: 39883297 DOI: 10.1007/s40266-024-01174-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 01/31/2025]
Abstract
Chronic itch in older patients is a common problem, with a significant impact on quality of life. Chronic itch in the older population may be attributable to several causes, such as age-related changes, skin conditions, systemic conditions, medications, and psychological conditions. Given the complexity of itch in this population, comorbidities, and polypharmacy in most geriatric patients, treating chronic itch can be challenging for healthcare providers. Therefore, optimized topical treatment regimens are paramount to help these patients and prevent side effects.
Collapse
Affiliation(s)
- Brooke Bartley
- Department of Internal Medicine, Texas Health Presbyterian Dallas, Dallas, TX, USA
| | - Christina Pierce
- University of Texas Health Science Center at San Antonio Long School of Medicine, San Antonio, TX, USA
| | - Chad Hivnor
- Department of Dermatology, University of Texas Health Science Center, 7979 Wurzbach Rd, Grossman 3rd FL, San Antonio, TX, 78229, USA
| | - Rodrigo Valdes-Rodriguez
- Department of Dermatology, University of Texas Health Science Center, 7979 Wurzbach Rd, Grossman 3rd FL, San Antonio, TX, 78229, USA.
| |
Collapse
|
6
|
Chen Z, Dragan M, Sun P, Haensel D, Vu R, Cui L, Zhu P, Yang N, Shi Y, Dai X. The AhR-Ovol1-Id1 regulatory axis in keratinocytes promotes epidermal and immune homeostasis in atopic dermatitis-like skin inflammation. Cell Mol Immunol 2025; 22:300-315. [PMID: 39939818 PMCID: PMC11868582 DOI: 10.1038/s41423-025-01264-z] [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] [Received: 05/04/2024] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
The skin is our outer permeability and immune defense barrier against myriad external assaults. Aryl hydrocarbon receptor (AhR) senses environmental factors and regulates barrier robustness and immune homeostasis. AhR agonists have been approved by the FDA for psoriasis treatment and are in clinical trials for the treatment of atopic dermatitis (AD), but the underlying mechanism of action remains poorly defined. Here, we report that OVOL1/Ovol1 is a conserved and direct transcriptional target of AhR in epidermal keratinocytes. We show that OVOL1/Ovol1 influences AhR-mediated regulation of keratinocyte gene expression and that OVOL1/Ovol1 ablation in keratinocytes impairs the barrier-promoting function of AhR, exacerbating AD-like inflammation. Mechanistically, we have identified Ovol1's direct downstream targets genome-wide and provided in vivo evidence supporting the role of Id1 as a functional target in barrier maintenance, disease suppression, and neutrophil accumulation. Furthermore, our findings reveal that an IL-1/dermal γδT cell axis exacerbates type 2 and 3 immune responses downstream of barrier perturbation in Ovol1-deficient AD skin. Finally, we present data suggesting the clinical relevance of OVOL1 and ID1 functions in human AD skin. Our study highlights a keratinocyte-intrinsic AhR-Ovol1-Id1 regulatory axis that promotes both epidermal and immune homeostasis in the context of skin inflammation, identifying new therapeutic targets.
Collapse
Affiliation(s)
- Zeyu Chen
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Morgan Dragan
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Peng Sun
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Daniel Haensel
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Remy Vu
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Lian Cui
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Peiyao Zhu
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Nan Yang
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Yuling Shi
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China.
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA.
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA.
- Department of Dermatology, School of Medicine, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
7
|
Dand N, Stuart PE, Bowes J, Ellinghaus D, Nititham J, Saklatvala JR, Teder-Laving M, Thomas LF, Traks T, Uebe S, Assmann G, Baudry D, Behrens F, Billi AC, Brown MA, Burkhardt H, Capon F, Chung R, Curtis CJ, Duckworth M, Ellinghaus E, FitzGerald O, Gerdes S, Griffiths CEM, Gulliver S, Helliwell PS, Ho P, Hoffmann P, Holmen OL, Huang ZM, Hveem K, Jadon D, Köhm M, Kraus C, Lamacchia C, Lee SH, Ma F, Mahil SK, McHugh N, McManus R, Modalsli EH, Nissen MJ, Nöthen M, Oji V, Oksenberg JR, Patrick MT, Perez White BE, Ramming A, Rech J, Rosen C, Sarkar MK, Schett G, Schmidt B, Tejasvi T, Traupe H, Voorhees JJ, Wacker EM, Warren RB, Wasikowski R, Weidinger S, Wen X, Zhang Z, Barton A, Chandran V, Esko T, Foerster J, Franke A, Gladman DD, Gudjonsson JE, Gulliver W, Hüffmeier U, Kingo K, Kõks S, Liao W, Løset M, Mägi R, Nair RP, Rahman P, Reis A, Smith CH, Di Meglio P, Barker JN, Tsoi LC, Simpson MA, Elder JT. GWAS meta-analysis of psoriasis identifies new susceptibility alleles impacting disease mechanisms and therapeutic targets. Nat Commun 2025; 16:2051. [PMID: 40021644 PMCID: PMC11871359 DOI: 10.1038/s41467-025-56719-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/28/2025] [Indexed: 03/03/2025] Open
Abstract
Psoriasis is a common, debilitating immune-mediated skin disease. Genetic studies have identified biological mechanisms of psoriasis risk, including those targeted by effective therapies. However, the genetic liability to psoriasis is not fully explained by variation at robustly identified risk loci. To refine the genetic map of psoriasis susceptibility we meta-analysed 18 GWAS comprising 36,466 cases and 458,078 controls and identified 109 distinct psoriasis susceptibility loci, including 46 that have not been previously reported. These include susceptibility variants at loci in which the therapeutic targets IL17RA and AHR are encoded, and deleterious coding variants supporting potential new drug targets (including in STAP2, CPVL and POU2F3). We conducted a transcriptome-wide association study to identify regulatory effects of psoriasis susceptibility variants and cross-referenced these against single cell expression profiles in psoriasis-affected skin, highlighting roles for the transcriptional regulation of haematopoietic cell development and epigenetic modulation of interferon signalling in psoriasis pathobiology.
Collapse
Grants
- R01 ES033634 NIEHS NIH HHS
- R01AR050511, R01AR054966, R01AR063611, R01AR065183 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- BRC_1215_20006, NIHR302258, NIHR203308, BRC-1215-20014 DH | National Institute for Health Research (NIHR)
- 980 Maudsley Charity
- RG2/10, ST1/19, ST3/20 Psoriasis Association
- EXC 2167-390884018, CRC1181-2/project A05 Deutsche Forschungsgemeinschaft (German Research Foundation)
- STR130505 Guy's and St Thomas' Charity
- K01 AR072129, P30 AR075043, UC2 AR081033, R01AR042742 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- K08 AR078251 NIAMS NIH HHS
- P30 AR075043 NIAMS NIH HHS
- K01 AR072129 NIAMS NIH HHS
- 814364 National Psoriasis Foundation (NPF)
- R01 AR042742 NIAMS NIH HHS
- PUT1465, PRG1189, PRG1911, PRG1291 Eesti Teadusagentuur (Estonian Research Council)
- 2014-2020.4.01.15-0012 EC | European Regional Development Fund (Europski Fond za Regionalni Razvoj)
- U01AI119125 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- LF-OC-22-001033 LEO Pharma Research Foundation
- 821511 Innovative Medicines Initiative (IMI)
- RG-1611-26299 National Multiple Sclerosis Society (National MS Society)
- MR/S003126/1 RCUK | Medical Research Council (MRC)
- U01 AI119125 NIAID NIH HHS
- R01ES033634, R35GM138121, K08 AR078251, R01AR065174 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 AR054966 NIAMS NIH HHS
- R01 AR050511 NIAMS NIH HHS
- R01 AR065174 NIAMS NIH HHS
- R35 GM138121 NIGMS NIH HHS
- 01EC1407A, 01EC1401C Bundesministerium für Bildung und Forschung (Federal Ministry of Education and Research)
- SI 236/8-1, SI236/9-1, ER 155/6-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- UC2 AR081033 NIAMS NIH HHS
- R01 AR065183 NIAMS NIH HHS
- R01 AR063611 NIAMS NIH HHS
- U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- Versus Arthritis - grant reference number 21754 Additional funding support from the following bodies are also acknowledged, as detailed in the funding section of the manuscript: Ann Arbor Veterans Hospital; Babcock Memorial Trust; Cambridge Arthritis Research Endeavour (CARE); Dermatology Foundation; Faculty of Medicine and Health Sciences, NTNU; German Centre for Neurodegenerative Disorders (DZNE), Bonn; German Ministry of Education and Science; Heinz Nixdorf Foundation (Germany); Joint Research Committee between St Olav’s Hospital and the Faculty of Medicine and Health Sciences, NTNU; Krembil Foundation; Liaison Committee for Education, Research, and Innovation in Central Norway; The Michael J. Fox Foundation; MSWA; National Institutes of Health; Perron Institute for Neurological and Translational Science; Pfizer Chair Research Award in Rheumatology; Research Council of Norway; Shake It Up Australia; Stiftelsen Kristian Gerhard Jebsen; Taubman Medical Research Institute; University of Michigan
Collapse
Affiliation(s)
- Nick Dand
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Health Data Research UK, London, UK
| | - Philip E Stuart
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - John Bowes
- Centre for Genetics and Genomics Versus Arthritis, The University of Manchester, Manchester, UK
- National Institute for Health and Care Research (NIHR) Manchester Biomedical Research Centre, The University of Manchester, Manchester, UK
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Joanne Nititham
- Deparment of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Jake R Saklatvala
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | | | - Laurent F Thomas
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- BioCore - Bioinformatics Core Facility, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tanel Traks
- Department of Dermatology and Venereology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Steffen Uebe
- Institute of Human Genetics, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Gunter Assmann
- RUB University Hospital JWK Minden, Department of Rheumatology, Minden, Germany
- Jose-Carreras Centrum for Immuno- and Gene Therapy, University of Saarland Medical School, Homburg, Germany
| | - David Baudry
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Frank Behrens
- Division of Translational Rheumatology, Immunology - Inflammation Medicine, University Hospital, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Allison C Billi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Matthew A Brown
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Genomics England, Canary Wharf, London, UK
| | - Harald Burkhardt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Francesca Capon
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Raymond Chung
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Denmark Hill, Camberwell, London, UK
- National Institute for Health and Care Research (NIHR) Biomedical Research Centre, South London and Maudsley Hospital, London, UK
| | - Charles J Curtis
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Denmark Hill, Camberwell, London, UK
- National Institute for Health and Care Research (NIHR) Biomedical Research Centre, South London and Maudsley Hospital, London, UK
| | - Michael Duckworth
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Eva Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Oliver FitzGerald
- UCD School of Medicine and Medical Sciences and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Sascha Gerdes
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Christopher E M Griffiths
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Centre for Dermatology Research, University of Manchester, NIHR Manchester Biomedical Research Centre, Manchester, UK
- Department of Dermatology, King's College Hospital NHS Foundation Trust, London, UK
| | | | - Philip S Helliwell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Pauline Ho
- Centre for Genetics and Genomics Versus Arthritis, The University of Manchester, Manchester, UK
- National Institute for Health and Care Research (NIHR) Manchester Biomedical Research Centre, The University of Manchester, Manchester, UK
- The Kellgren Centre for Rheumatology, Manchester University NHS Foundation Trust, Manchester, UK
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Oddgeir L Holmen
- HUNT Research Centre, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Levanger, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Zhi-Ming Huang
- Deparment of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Levanger, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Deepak Jadon
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Michaela Köhm
- Division of Translational Rheumatology, Immunology - Inflammation Medicine, University Hospital, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Cornelia Kraus
- Institute of Human Genetics, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Céline Lamacchia
- Division of Rheumatology, Geneva University Hospital, Geneva, Switzerland
| | - Sang Hyuck Lee
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Denmark Hill, Camberwell, London, UK
- National Institute for Health and Care Research (NIHR) Biomedical Research Centre, South London and Maudsley Hospital, London, UK
| | - Feiyang Ma
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Satveer K Mahil
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- St John's Institute of Dermatology, Guy's and St Thomas' National Health Service (NHS) Foundation Trust, London, UK
| | - Neil McHugh
- Department of Life Sciences, University of Bath, Bath, UK
| | - Ross McManus
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Ellen H Modalsli
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Dermatology, Clinic of Orthopedy, Rheumatology and Dermatology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Michael J Nissen
- Division of Rheumatology, Geneva University Hospital, Geneva, Switzerland
| | - Markus Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Vinzenz Oji
- Department of Dermatology, University of Münster, Münster, Germany
| | - Jorge R Oksenberg
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew T Patrick
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Andreas Ramming
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jürgen Rech
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Cheryl Rosen
- Division of Dermatology, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Mrinal K Sarkar
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Börge Schmidt
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Trilokraj Tejasvi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Ann Arbor Veterans Affairs Hospital, Ann Arbor, MI, USA
| | - Heiko Traupe
- Department of Dermatology, University of Münster, Münster, Germany
| | - John J Voorhees
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eike Matthias Wacker
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Richard B Warren
- Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Centre for Dermatology Research, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M6 8HD, UK
| | - Rachael Wasikowski
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Stephan Weidinger
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Xiaoquan Wen
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Zhaolin Zhang
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, The University of Manchester, Manchester, UK
- National Institute for Health and Care Research (NIHR) Manchester Biomedical Research Centre, The University of Manchester, Manchester, UK
- The Kellgren Centre for Rheumatology, Manchester University NHS Foundation Trust, Manchester, UK
| | - Vinod Chandran
- Schroeder Arthritis Institute, Krembil Research Institute and Toronto Western Hospital, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Tõnu Esko
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - John Foerster
- College of Medicine, Dentistry, and Nursing, University of Dundee, Dundee, UK
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Dafna D Gladman
- Schroeder Arthritis Institute, Krembil Research Institute and Toronto Western Hospital, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Johann E Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Wayne Gulliver
- Newlab Clinical Research Inc, St. John's, NL, Canada
- Department of Dermatology, Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Ulrike Hüffmeier
- Institute of Human Genetics, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Külli Kingo
- Department of Dermatology and Venereology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA, 6150, Australia
| | - Wilson Liao
- Deparment of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Mari Løset
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Dermatology, Clinic of Orthopedy, Rheumatology and Dermatology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Rajan P Nair
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Proton Rahman
- Memorial University of Newfoundland, St. John's, NL, Canada
| | - André Reis
- Institute of Human Genetics, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Catherine H Smith
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- St John's Institute of Dermatology, Guy's and St Thomas' National Health Service (NHS) Foundation Trust, London, UK
| | - Paola Di Meglio
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Jonathan N Barker
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- St John's Institute of Dermatology, Guy's and St Thomas' National Health Service (NHS) Foundation Trust, London, UK
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Michael A Simpson
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK.
| | - James T Elder
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Ann Arbor Veterans Affairs Hospital, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Liu K, Deng S, Zhou Y, Xu B, Zhang Y, Li W, Liu X, Yao X. Crosstalk Between the Skin Environment and Microbial Community in Immune-Related Skin Diseases. Clin Rev Allergy Immunol 2025; 68:16. [PMID: 39954089 DOI: 10.1007/s12016-025-09029-2] [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] [Accepted: 02/04/2025] [Indexed: 02/17/2025]
Abstract
The skin surface hosts diverse skin microbiota, including bacteria, fungi, and viruses. Intricate interactions between the skin microenvironment and microbial community are crucial for maintaining cutaneous homeostasis. This review explores the bidirectional relationship between the skin ecosystem and its microbiota. The skin microenvironment is shaped by a combination of intrinsic factors, dominated by sweat glands and pilosebaceous units, and external factors, such as UV radiation and personal care products, which create distinct niches that influence microbial colonization patterns across different skin regions. The skin microbiome, in turn, modulates the physical, chemical, immunological, and microbial barriers of the skin. We also discuss the alterations in this crosstalk in various immune-related skin conditions such as atopic dermatitis, psoriasis, rosacea, hidradenitis suppurativa, skin cancer, and aging. Understanding these interactions is vital for developing targeted microbiome-based therapies for various skin disorders. Further researches are needed to deepen insights into the microbial roles and their therapeutic potentials in skin health and disease.
Collapse
Affiliation(s)
- Kecheng Liu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Shuting Deng
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Yuan Zhou
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Beilei Xu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Yu Zhang
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Wei Li
- Department of Dermatology, Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, Shanghai, 200040, China
| | - Xiaochun Liu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
| | - Xu Yao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
| |
Collapse
|
9
|
Dawe HR, Di Meglio P. The Aryl Hydrocarbon Receptor (AHR): Peacekeeper of the Skin. Int J Mol Sci 2025; 26:1618. [PMID: 40004095 PMCID: PMC11855870 DOI: 10.3390/ijms26041618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
In the last decade, the aryl hydrocarbon receptor (AHR) has emerged as a critical peacekeeper for the maintenance of healthy skin. The evolutionary conservation of AHR implied physiological functions for this receptor, beyond the detoxification of man-made compounds, a notion further supported by the existence of physiological AHR ligands, notably derivates of tryptophan by the host and host microbiome. The UV light-derived ligand, 6-formylindolo[3,2-b]carbazole (FICZ), anticipated a role for AHR in skin, a UV light-exposed organ, where physiological AHR activation promotes a healthy skin barrier and constrains inflammation. The clinical development of tapinarof, the first topical AHR modulating drug for inflammatory skin disease, approved by the FDA for mild-to-moderate psoriasis and poised for approval in atopic dermatitis, supports the therapeutic targeting of the AHR pathway to harness its beneficial effect in skin inflammation. Here, we describe how a tightly controlled, physiological activation of the AHR pathway maintains skin homeostasis, and discuss how the pathway is dysregulated in psoriasis and atopic dermatitis, identifying areas offering opportunities for alternative therapeutic approaches, for further investigation.
Collapse
Affiliation(s)
- Hannah R. Dawe
- St John’s Institute of Dermatology, King’s College London, London SE1 9RT, UK;
- KHP Centre for Translational Medicine, London SE1 9RT, UK
| | - Paola Di Meglio
- St John’s Institute of Dermatology, King’s College London, London SE1 9RT, UK;
- KHP Centre for Translational Medicine, London SE1 9RT, UK
| |
Collapse
|
10
|
Zhu BJ, Yao LY, Qiu SL, Wu YD, Kang M, Zhao LY, Qiu SX. Stilbene-enriched extract from the leaves of Cajanus cajan attenuates psoriasis in imiquimod-induced psoriatic mice by targeting aryl hydrocarbon receptor and chemokines. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119109. [PMID: 39547364 DOI: 10.1016/j.jep.2024.119109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The leaves of Cajanus cajan (L.) Millsp., an Asian traditional folkloric medicine, have been used to treat inflammatory conditions since ancient times. In Southern China, these leaves have been employed to alleviate the symptoms associated with various skin diseases. However, the therapeutic effects and the underlying mechanisms of Cajanus cajan leaves in the treatment of psoriasis remain poorly understood. AIM OF THE STUDY This study aims to investigate the efficacy of stilbene-enriched extract from C. cajan leaves (termed as "EXT") in treating imiquimod (IMQ)-induced psoriatic mice and to elucidate its possible underlying mechanism in psoriasis treatment. MATERIALS AND METHODS The coumpounds of EXT was analyzed through a UPLC-MS system, the MS survey scan was conducted across the mass range of m/z 100-1000 Da. The activation of aryl hydrocarbon receptor (AhR), a potential therapeutic target, by EXT in HaCaT cells was assessed using RT-qPCR and immunofluorescence. Subsequently, EXT was administrated to IMQ-induced psoriatic mice once daily for 10 days. The efficacy of EXT in treating psoriasis was evaluated through pathological analysis including change of weight, PASI score, Baker score, epidermal thickness, and H&E staining of lesion skin. Additionally, transcriptomic analysis of lesion skins was conducted to identify the potential therapeutic targets and possible mechanisms of EXT in psoriasis treatment. RESULTS It was identified that the primary stilbenes present in EXT were 3.10% pinosylvin monomethyl ether (PME), 12.32 % cajaninstilbene (CSA), 4.54 % ongistylin A (LGA) and 2.43 % longistylin C (LGC). In cellular tests, the addition of 2.5 μg/mL EXT to HaCaT cells enhanced the expression of AhR and its nuclear translocation. In vivo tests of EXT in IMQ-induced psoriasis mouse model, 50 mg 1.0 % EXT reduced PASI and Baker score of lesion skin to 2.67 and 4.5, respectively. In addition, the epidermis thickness of lesion skin induced by IMQ returned to normal following the application of 50 mg 1.0 % EXT in psoriatic mice. Transcriptomic profiling revealed significant downregulation of numerous chemokines (Ccl2, Ccl20, and Cxc5, etc.), pro-inflammatory cytokines (Il17a, Il19, Il22, and Il23, etc.), and genes associated with keratinocyte differentiation (Lce and Sprr family genes). Conversely, AhR and genes of the cytochrome P450 family were activated. CONCLUSIONS This study is the first to demonstrate that the ethyl acetate (EtOAc) extract enriched with stilbenes from Cajanus cajan leaves (EXT) effectively alleviates symptoms in IMQ-induced psoriatic mice. The mechanism involves the activation of the aryl hydrocarbon receptor (AhR) and a subsequent reduction in the production of various inflammatory chemokines and cytokines. These findings suggest that EXT holds significant potential as a plant-derived therapeutic agent for the treatment of psoriasis.
Collapse
Affiliation(s)
- Bao-Jun Zhu
- Program for Natural Product Chemical Biology, State Key Laboratory of Plant Diversity and Specialty Crops & Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China; South China National Botanical Garden, Guangzhou, 510650, PR China
| | - Li-Yuan Yao
- Program for Natural Product Chemical Biology, State Key Laboratory of Plant Diversity and Specialty Crops & Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China; South China National Botanical Garden, Guangzhou, 510650, PR China; University of the Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Si-Lin Qiu
- Program for Natural Product Chemical Biology, State Key Laboratory of Plant Diversity and Specialty Crops & Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China; South China National Botanical Garden, Guangzhou, 510650, PR China
| | - Yao-Dan Wu
- Program for Natural Product Chemical Biology, State Key Laboratory of Plant Diversity and Specialty Crops & Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China; South China National Botanical Garden, Guangzhou, 510650, PR China
| | - Ming Kang
- Program for Natural Product Chemical Biology, State Key Laboratory of Plant Diversity and Specialty Crops & Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China; South China National Botanical Garden, Guangzhou, 510650, PR China
| | - Li-Yun Zhao
- Program for Natural Product Chemical Biology, State Key Laboratory of Plant Diversity and Specialty Crops & Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China; South China National Botanical Garden, Guangzhou, 510650, PR China.
| | - Sheng-Xiang Qiu
- Program for Natural Product Chemical Biology, State Key Laboratory of Plant Diversity and Specialty Crops & Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, PR China; South China National Botanical Garden, Guangzhou, 510650, PR China.
| |
Collapse
|
11
|
Yang J, Qiao P, Wang G, Dang E. The Role of Aryl Hydrocarbon Receptor in Skin Homeostasis: Implications for Therapeutic Strategies in Skin Disorders. Cell Biochem Funct 2025; 43:e70047. [PMID: 39866071 DOI: 10.1002/cbf.70047] [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] [Received: 10/08/2024] [Revised: 12/26/2024] [Accepted: 01/15/2025] [Indexed: 01/28/2025]
Abstract
The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, is extensively expressed in diverse human organs and plays a pivotal role in mediating the onset, progression, and severity of numerous diseases. Recent research has explored the substantial impact of AhR on skin homeostasis and related pathologies. As a multi-layered organ, the skin comprises multiple cell populations that express AhR. In this review, we introduce the role of AhR in various skin cells and its impact on skin barrier function. Furthermore, we explore the involvement of AhR in the development of various skin diseases, highlighting its potential as a therapeutic target for skin disorders. By targeting AhR, we may open new avenues for the development of novel and efficient skin disease treatments.
Collapse
Affiliation(s)
- Jundan Yang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pei Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
12
|
Zhu S, Cheng L, Chen T, Liu X, Zhang C, Aji A, Guo W, Zhu J, Chu Y, Guo D, Li F. Bergapten Ameliorates Psoriatic Skin Lesions and IL-17A-Induced Activation of the NF-κB Signaling Pathway via the Downregulation of CYP1B1. Phytother Res 2025; 39:661-675. [PMID: 39638770 DOI: 10.1002/ptr.8399] [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] [Received: 07/23/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 12/07/2024]
Abstract
Bergapten (BP) is a plant-derived furocoumarin that has a wide range of pharmacological effects. BP serves as a candidate amplifier in phototherapy against skin inflammation, such as psoriasis and atopic dermatitis. However, the anti-inflammatory role of BP remains elusive. We utilized IL-17A-stimulated keratinocyte line and imiquimod-challenged BALB/c mice to imitate psoriasis-like inflammation. Inflammatory phenotypes were determined by expressions of inflammatory genes and cytokines, histopathological changes and activities of nuclear factor-κB (NF-κB) pathway. An RNA-seq analysis of rodent skin was performed to explore possible mechanism lying behind. SiRNAs and antagonist (TMS) against cytochrome P450 family 1 subfamily B member 1 (CYP1B1) were subsequently used to determine the role of CYP1B1 in psoriasis pathogenesis in vitro and in vivo. Overexpression of CYP1B1 with lentivirus further validate therapeutic effect of BP. BP significantly suppressed activation of the NF-κB pathway by inhibiting p65 phosphorylation and improved the inflammatory phenotype both in vitro and in vivo. We revealed the key role of CYP1B1 in regulating the activation of the NF-κB signaling pathway. Knock-down with siRNAs significantly reduce the expression of inflammatory genes and cytokines. An intraperitoneal injection of TMS partially remediated IMQ-induced inflammation, mainly in terms of skin thickness. Overexpression of Cyp1b1 led to increased expression of the CYP1B1 protein and rescued the therapeutic effect of BP in vitro. This study revealed that BP suppressed expression of Cyp1b1 in keratinocytes and inhibited the activation of NF-κB signaling pathway by blocking the phosphorylation of p65.
Collapse
Affiliation(s)
- Shengjie Zhu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linyan Cheng
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Teng Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin Liu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Abudula Aji
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wanjun Guo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianyong Zhu
- Department of Pharmacy Research, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuxia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dongjie Guo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fulun Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
13
|
Kantheti U, Forward TS, Lucas ED, Schafer JB, Tamburini PJ, Burchill MA, Tamburini BAJ. PD-L1-CD80 interactions are required for intracellular signaling necessary for dendritic cell migration. SCIENCE ADVANCES 2025; 11:eadt3044. [PMID: 39879305 PMCID: PMC11777207 DOI: 10.1126/sciadv.adt3044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025]
Abstract
Programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) interactions are targets for immunotherapies aimed to reinvigorate T cell function. Recently, it was documented that PD-L1 regulates dendritic cell (DC) migration through intracellular signaling events. In this study, we find that both preclinical murine and clinically available human PD-L1 antibodies limit DC migration. We show that cis interactions between PD-L1 and CD80 are critical for promoting migration and define specific regions within these proteins necessary for migration. Furthermore, we demonstrate that αPD-L1 significantly impedes DC migration in a B16 melanoma tumor model. Last, we outline how blocking cis PD-L1:CD80 interactions or mutation of the intracellular domain of PD-L1, in an imiquimod-induced murine model of psoriasis, limits DC migration to the lymph node, decreases interleukin-17 production by CD4+ T cells in the lymph node, and reduces epidermal thickening. Therefore, PD-L1 and CD80 interactions are important regulators of DC migration to the draining lymph node.
Collapse
Affiliation(s)
- Uma Kantheti
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Tadg S. Forward
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Erin D. Lucas
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Johnathon B. Schafer
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Pierce J. Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Matthew A. Burchill
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Beth Ann Jirón Tamburini
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Aurora, CO, USA
- Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
14
|
Gan YL, Lee YH. Indoleamine 2,3-Dioxygenase 1/Aryl Hydrocarbon Receptor Feedback Loop Mediates Anti-inflammation in lipopolysaccharide-stimulated Astrocytes to Dampen Inflammatory Neurotoxicity. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2025; 68:1-10. [PMID: 39846297 DOI: 10.4103/ejpi.ejpi-d-24-00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/12/2024] [Indexed: 01/24/2025]
Abstract
ABSTRACT Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that regulates cell immune responses in a cell type-specific and ligand-dependent manner. In the central nervous system, astrocytic AhR plays important roles in regulating neuroinflammation by mediating responses to endogenous ligands generated from the inflammation-induced indoleamine 2,3-dioxygenase 1 (IDO1)/kynurenine (KYN) pathway. We previously demonstrated that reduction of AhR expression decreases lipopolysaccharide (LPS)-induced pro-inflammatory responses in microglia. However, the role of AhR in the astrocytic immune responses and its subsequent effects on microglial activation and neurotoxicity remain unclear. In this study, we used LPS-induced neuroinflammation in rat cortical glia-neuron (GN) mix cultures, which increased the expression of tumor necrosis factor-α and interleukin-6 and microglial activation. These proinflammatory responses were attenuated by a specific AhR agonist 6-formylindolo [3,2-b] carbazole (FICZ), but not by the AhR antagonist CH223191. CH223191, which inhibits LPS- and FICZ-induced AhR activation, enhanced neurotoxicity induced by LPS-glutamate co-treatment in GN mix cultures. Furthermore, inhibition of AhR expression and activation enhanced LPS-induced proinflammatory responses, and LPS-induced AhR activation was abrogated by the inhibition of IDO1 expression in astrocytes. Notably, AhR knockdown inhibited the anti-inflammatory effects of KYN while enhancing LPS-induced IDO1 expression in astrocytes, suggesting that AhR mediates the anti-inflammatory effect of KYN and the negative feedback regulation of IDO1 expression. Finally, we examined the role of astrocytic AhR in inflammatory astrogliosis-induced neurotoxicity by treating primary cortical neurons with LPS-treated astrocyte-conditioned medium (ACM). The results revealed that ACM derived from siAhR-transfected astrocytes increased neurotoxicity. In conclusion, inflammation-activated AhR mediates the anti-inflammatory effects and negative feedback regulation of the IDO1/KYN pathway in astrocytes, thereby dampening inflammatory astrogliosis-induced neurotoxicity.
Collapse
Affiliation(s)
- Yu-Ling Gan
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
15
|
Eichenfield LF, Silverberg JI, Hebert AA, Boguniewicz M. Targeting the aryl hydrocarbon receptor as a strategy to expand the therapeutic armamentarium in atopic dermatitis. J DERMATOL TREAT 2024; 35:2300354. [PMID: 38213229 DOI: 10.1080/09546634.2023.2300354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/23/2023] [Indexed: 01/13/2024]
Affiliation(s)
| | - Jonathan I Silverberg
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Adelaide A Hebert
- UTHealth McGovern School of Medicine and Children's Memorial Hermann Hospital, Houston, TX, USA
| | - Mark Boguniewicz
- National Jewish Health and University of Colorado School of Medicine, Denver, CO, USA
| |
Collapse
|
16
|
Elias AE, McBain AJ, Aldehalan FA, Taylor G, O'Neill CA. Activation of the aryl hydrocarbon receptor via indole derivatives is a common feature in skin bacterial isolates. J Appl Microbiol 2024; 135:lxae273. [PMID: 39444068 DOI: 10.1093/jambio/lxae273] [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] [Received: 08/13/2024] [Revised: 10/04/2024] [Accepted: 10/22/2024] [Indexed: 10/25/2024]
Abstract
AIMS The aryl hydrocarbon receptor (AhR) is a ligand-activated receptor implicated in many inflammatory disorders. The skin microbiota plays a crucial role in maintaining epidermal barrier integrity and is thought to modulate skin homeostasis partly through the production of AhR ligands, including metabolites of microbial tryptophan metabolism such as indole derivatives. Here, we report the skin microbiota that activate AhR and their unique tryptophan metabolite profiles. METHODS AND RESULTS Of the bacteria isolated from healthy human skin and screened for the ability to metabolize tryptophan (18 species, five genera), 14 were positive. The tryptophan metabolites of 10 positive and two negative bacteria were then characterized using liquid chromatography-mass spectrometry. Whole genome sequencing confirmed the presence of key genes involved in the indole-3-pyruvic acid pathway within the genomes of indole-3-acetaldehyde, indole-3-acetic acid, and indole-3-aldehyde-producing organisms. A cell-based luciferase reporter gene assay identified functional agonist activity against human AhR in the culture supernatants of 12 of the 18 species tested. High indole derivative-producing organisms induced potent AhR activation. CONCLUSIONS These data demonstrate the relationship between skin microbiota, tryptophan metabolites, and AhR activation.
Collapse
Affiliation(s)
- Abigail E Elias
- Division of Musculoskeletal and Dermatological Sciences, Manchester M13 9PL, United Kingdom
| | - Andrew J McBain
- Division of Pharmacy and Optometry, Manchester M13 9PL, United Kingdom
| | - Faye A Aldehalan
- Division of Musculoskeletal and Dermatological Sciences, Manchester M13 9PL, United Kingdom
| | - George Taylor
- Biological Mass Spectrometry Facility, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Catherine A O'Neill
- Division of Musculoskeletal and Dermatological Sciences, Manchester M13 9PL, United Kingdom
| |
Collapse
|
17
|
Yu S, Li Z, Zhang X, Zhang Q, Zhang L, Zhao L, Liu P, Guo J, Chen J, Zhang C, Liu X, Yu M, Jin D, Wang X, Li G, Cao Y, Ren F, Wang R. Skin Rejuvenation in Aged Mice by Fecal Transplantation Microbiota from Young Mice Feces. ENGINEERING 2024; 42:26-38. [DOI: 10.1016/j.eng.2024.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
18
|
Lin Y, Yang L, Wang D, Lei H, Zhang Y, Sun W, Liu J. Indigo alleviates psoriasis through the AhR/NF-κB signaling pathway: an in vitro and in vivo study. PeerJ 2024; 12:e18326. [PMID: 39465158 PMCID: PMC11505883 DOI: 10.7717/peerj.18326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
Background Psoriasis is a chronic inflammatory skin disease. A strong association between the AhR/ NFκB axis and the inflammatory response in psoriasis. Indigo (IDG) has demonstrated significant anti-inflammatory properties. This study aimed to assess the anti-psoriatic efficacy of IDG while investigating the underlying mechanisms involved. Methods In the in vitro experiments, cell viability was assessed using the CCK-8. qRT-PCR was employed to measure the mRNA levels of NF-κB, TNF-α, IL-1β, AhR, and CYP1A1. Western blotting was conducted to examine alterations in cytoplasmic and nuclear AhR protein levels. Additionally, an IDG nanoemulsion (NE) cream was prepared for the in vivo experiments. A psoriasis-like skin lesion mice model was induced using IMQ (62.5 mg/day for 7 days). The severity of psoriasis was evaluated using PASI, and skin lesions were scored while epidermal thickness was assessed via HE staining. The expression of inflammatory markers, including IL-6, IL-13, IL-17A, MCP-1, and TNF-α, was detected in skin lesions using Luminex. The levels of CYP1A1, p65, and p-p65 proteins were determined by Western blotting. Results LPS stimulation significantly elevated TNF-α, IL-6, and NF-κB mRNA levels, which were notably reduced by IDG treatment. Additionally, IDG significantly enhanced the expression of AhR and CYP1A1 mRNA. Further investigation revealed that IDG facilitated AhR translocation from the cytoplasm to the nucleus. In the IMQ-induced psoriasis-like mouse model, IDG NE substantially ameliorated the severity of skin lesions. Moreover, IDG NE treatment reduced the upregulation of inflammatory cytokines such as IL-6, IL-17A, MCP-1, and TNF-α in IMQ-induced skin lesions. It was also observed that IDG NE treatment increased CYP1A1 protein expression while inhibiting p65 and p-p65 protein expression. Conclusion IDG emerges as a promising treatment for psoriasis, demonstrating effective therapeutic outcomes. Its mechanism of action is likely linked to the modulation of the AhR/NFκB signaling pathway.
Collapse
Affiliation(s)
- Yu Lin
- Department of Dermatology, the First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Dermatology, the First Affliated Hospital of Jinan University, Guangzhou, China
| | - Lihong Yang
- Department of Dermatology, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Dongxiang Wang
- Department of Dermatology, the First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haiqing Lei
- Department of Dermatology, the First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuelin Zhang
- Department of Dermatology, the First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wen Sun
- Department of Dermatology, Jingmen Central Hospital, Jingmen, China
| | - Jing Liu
- Department of Dermatology, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| |
Collapse
|
19
|
Dai W, Yin S, Wang F, Kuang T, Xiao H, Kang W, Yun C, Wang F, Luo L, Ao S, Zhou J, Yang X, Fan C, Li W, He D, Jin H, Tang W, Liu L, Wang R, Liang H, Zhu J. Punicalagin as a novel selective aryl hydrocarbon receptor (AhR) modulator upregulates AhR expression through the PDK1/p90RSK/AP-1 pathway to promote the anti-inflammatory response and bactericidal activity of macrophages. Cell Commun Signal 2024; 22:473. [PMID: 39363344 PMCID: PMC11448010 DOI: 10.1186/s12964-024-01847-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024] Open
Abstract
Aryl hydrocarbon receptor (AhR) plays an important role in inflammation and immunity as a new therapeutic target for infectious disease and sepsis. Punicalagin (PUN) is a Chinese herbal monomer extract of pomegranate peel that has beneficial anti-inflammatory, antioxidant and anti-infective effects. However, whether PUN is a ligand of AhR, its effect on AhR expression, and its signaling pathway remain poorly understood. In this study, we found that PUN was a unique polyphenolic compound that upregulated AhR expression at the transcriptional level, and regulated the AhR nongenomic pathway. AhR expression in lipopolysaccharide-induced macrophages was upregulated by PUN in vitro and in vivo in a time- and dose-dependent manner. Using specific inhibitors and siRNA, induction of AhR by PUN depended on sequential phosphorylation of 90-kDa ribosomal S6 kinase (p90RSK), which was activated by the mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) and phosphoinositide-dependent protein kinase (PDK)1 pathways. PUN promoted p90RSK-mediated activator protein-1 (AP-1) activation. AhR knockout or inhibitors reversed suppression of interleukin (IL)-6 and IL-1β expression by PUN. PUN decreased Listeria load and increased macrophage survival via AhR upregulation. In conclusion, we identified PUN as a novel selective AhR modulator involved in AhR expression via the MEK/ERK and PDK1 pathways targeting p90RSK/AP-1 in inflammatory macrophages, which inhibited macrophage inflammation and promoted bactericidal activity.
Collapse
Affiliation(s)
- Weihong Dai
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Emergency of The Second Affiliated Hospital of Hainan Medical University, Haikou, 571100, China
| | - Shuangqin Yin
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Fangjie Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Tianyin Kuang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hongyan Xiao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Wenyuan Kang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education & Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Caihong Yun
- Emergency of The Second Affiliated Hospital of Hainan Medical University, Haikou, 571100, China
| | - Fei Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Emergency of The Second Affiliated Hospital of Hainan Medical University, Haikou, 571100, China
| | - Li Luo
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shengxiang Ao
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jing Zhou
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xue Yang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Chao Fan
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Wei Li
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Dongmei He
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - He Jin
- Department of Cardiothoracic Surgery, 926th Hospital of Joint Logistics Support Force of PLA, Kaiyuan, 661600, China
| | - Wanqi Tang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lizhu Liu
- Emergency of The Second Affiliated Hospital of Hainan Medical University, Haikou, 571100, China
| | - Rixing Wang
- Emergency of The Second Affiliated Hospital of Hainan Medical University, Haikou, 571100, China.
| | - Huaping Liang
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Junyu Zhu
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
20
|
Stockinger B, Diaz OE, Wincent E. The influence of AHR on immune and tissue biology. EMBO Mol Med 2024; 16:2290-2298. [PMID: 39242971 PMCID: PMC11473696 DOI: 10.1038/s44321-024-00135-w] [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] [Received: 05/03/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024] Open
Abstract
The aryl hydrocarbon receptor is a ligand dependent transcription factor which functions as an environmental sensor. Originally discovered as the sensor for man made pollutants such as 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) it has recently gained prominence as an important mediator for environmental triggers via the diet or microbiota which influences many physiological functions in different cell types and tissues across the body. Notably AHR activity contributes to prevent excessive inflammation following tissue damage in barrier organs such as skin, lung or gut which has received wide attention in the past decade. In this review we will focus on emerging common AHR functions across cell types and tissues and discuss ongoing issues that confound the understanding of AHR physiology. Furthermore, we will discuss the need for deeper molecular understanding of the functional activity of AHR in different contexts with respect to development of potential therapeutic applications.
Collapse
Affiliation(s)
| | - Oscar E Diaz
- The Francis Crick Institute, London, United Kingdom
| | - Emma Wincent
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
21
|
Badawy AAB, Dawood S. Molecular Insights into the Interaction of Tryptophan Metabolites with the Human Aryl Hydrocarbon Receptor in Silico: Tryptophan as Antagonist and no Direct Involvement of Kynurenine. FRONT BIOSCI-LANDMRK 2024; 29:333. [PMID: 39344334 DOI: 10.31083/j.fbl2909333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND A direct link between the tryptophan (Trp) metabolite kynurenine (Kyn) and the aryl hydrocarbon receptor (AhR) is not supported by metabolic considerations and by studies demonstrating the failure of Kyn concentrations of up to 100 μM to activate the receptor in cell culture systems using the proxy system of cytochrome P-450-dependent metabolism. The Kyn metabolite kynurenic acid (KA) activates the AhR and may mediate the Kyn link. Recent studies demonstrated down regulation and antagonism of activation of the AhR by Trp. We have addressed the link between Kyn and the AhR by looking at their direct molecular interaction in silico. METHODS Molecular docking of Kyn, KA, Trp and a range of Trp metabolites to the crystal structure of the human AhR was performed under appropriate docking conditions. RESULTS Trp and 30 of its metabolites docked to the AhR to various degrees, whereas Kyn and 3-hydroxykynurenine did not. The strongest docking was observed with the Trp metabolite and photooxidation product 6-Formylindolo[3,2-b]carbazole (FICZ), cinnabarinic acid, 5-hydroxytryptophan, N-acetyl serotonin and indol-3-yllactic acid. Strong docking was also observed with other 5-hydroxyindoles. CONCLUSIONS We propose that the Kyn-AhR link is mediated by KA. The strong docking of Trp and its recently reported down regulation of the receptor suggest that Trp is an AhR antagonist and may thus play important roles in body homeostasis beyond known properties or simply being the precursor of biologically active metabolites. Differences in AhR activation reported in the literature are discussed.
Collapse
Affiliation(s)
- Abdulla A-B Badawy
- Formerly School of Health Sciences, Cardiff Metropolitan University, CF5 2YB Wales, UK
| | - Shazia Dawood
- Pharmacy and Allied Health Sciences, Iqra University, 7580 Karachi, Pakistan
| |
Collapse
|
22
|
Le S, Wu X, Dou Y, Song T, Fu H, Luo H, Zhang F, Cao Y. Promising strategies in natural products treatments of psoriasis-update. Front Med (Lausanne) 2024; 11:1386783. [PMID: 39296901 PMCID: PMC11408484 DOI: 10.3389/fmed.2024.1386783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/31/2024] [Indexed: 09/21/2024] Open
Abstract
Psoriasis is a chronic, relapsing, inflammatory skin disease and has been increasing year by year. It is linked to other serious illnesses, such as psoriatic arthritis, cardiometabolic syndrome, and depression, resulting in a notable decrease in the quality of life for patients. Existing therapies merely alleviate symptoms, rather than providing a cure. An in-depth under-standing of the pathogenesis of psoriasis is helpful to discover new therapeutic targets and develop effective novel therapeutic agents, so it has important clinical significance. This article reviews the new progress in the study of pathogenesis and natural products of psoriasis in recent years. These natural products were summarized, mainly classified as terpenoids, polyphenols and alkaloids. However, the translation of experimental results to the clinic takes a long way to go.
Collapse
Affiliation(s)
- Sihua Le
- Ningbo Medical Center LiHuiLi Hosptial, Ningbo, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuan Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuan Dou
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tianhao Song
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongyang Fu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Hongbin Luo
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Fan Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Yi Cao
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| |
Collapse
|
23
|
Smits JPH, Qu J, Pardow F, van den Brink NJM, Rodijk-Olthuis D, van Vlijmen-Willems IMJJ, van Heeringen SJ, Zeeuwen PLJM, Schalkwijk J, Zhou H, van den Bogaard EH. The Aryl Hydrocarbon Receptor Regulates Epidermal Differentiation through Transient Activation of TFAP2A. J Invest Dermatol 2024; 144:2013-2028.e2. [PMID: 38401701 DOI: 10.1016/j.jid.2024.01.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/26/2024]
Abstract
The aryl hydrocarbon receptor (AHR) is an evolutionary conserved environmental sensor identified as an indispensable regulator of epithelial homeostasis and barrier organ function. Molecular signaling cascade and target genes upon AHR activation and their contribution to cell and tissue function are however not fully understood. Multiomics analyses using human skin keratinocytes revealed that upon ligand activation, AHR binds open chromatin to induce expression of transcription factors, for example, TFAP2A, as a swift response to environmental stimuli. The terminal differentiation program, including upregulation of barrier genes, FLG and keratins, was mediated by TFAP2A as a secondary response to AHR activation. The role of AHR-TFAP2A axis in controlling keratinocyte terminal differentiation for proper barrier formation was further confirmed using CRISPR/Cas9 in human epidermal equivalents. Overall, the study provides additional insights into the molecular mechanism behind AHR-mediated barrier function and identifies potential targets for the treatment of skin barrier diseases.
Collapse
Affiliation(s)
- Jos P H Smits
- Department of Dermatology, Radboud Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands; Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Jieqiong Qu
- Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Felicitas Pardow
- Department of Dermatology, Radboud Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands; Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Noa J M van den Brink
- Department of Dermatology, Radboud Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands
| | - Diana Rodijk-Olthuis
- Department of Dermatology, Radboud Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands
| | | | - Simon J van Heeringen
- Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Patrick L J M Zeeuwen
- Department of Dermatology, Radboud Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands
| | - Joost Schalkwijk
- Department of Dermatology, Radboud Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands
| | - Huiqing Zhou
- Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands; Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands.
| | - Ellen H van den Bogaard
- Department of Dermatology, Radboud Research Institute for Medical Innovation, Radboudumc, Nijmegen, The Netherlands.
| |
Collapse
|
24
|
Başar Kılıç Ş, Taheri S, Mehmetbeyoğlu Duman E, Öksüm Solak E, Yılmaz Şükranlı Z, Rassoulzadegan M, Borlu M. Psoriatic skin transcript phenotype: androgen/estrogen and cortisone/cortisol imbalance with increasing DNA damage response. Mol Biol Rep 2024; 51:933. [PMID: 39180588 DOI: 10.1007/s11033-024-09782-1] [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] [Received: 03/30/2024] [Accepted: 07/04/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Patients prone to psoriasis suffer after a breakdown of the epidermal barrier and develop poorly healing lesions with abnormal proliferation of keratinocytes. Strong inflammatory reactions with genotoxicity (short telomeres) suggest impaired immune defenses with DNA damage repair response (DDR) in patients with psoriasis. Recent evidence indicates the existence of crosstalk mechanisms linking the DDR machinery and hormonal signaling pathways that cooperate to influence both progressions of many diseases and responses to treatment. The aim of this study was to clarify whether steroid biosynthesis and genomic stability markers are altered in parallel during the formation of psoriatic skin. Understanding the interaction of the steroid pathway and DNA damage response is crucial to addressing underlying fundamental issues and managing resulting epidermal barrier disruption in psoriasis. METHODS Skin (Lesional, non-lesional) and blood samples from twenty psoriasis patients and fifteen healthy volunteers were collected. Real-Time-PCR study was performed to assess levels of known transcripts such as: estrogen (ESR1, ESR2), androgen (AR), glucocorticoid/mineralocorticoid receptors (NR3C1, NR3C2), HSD11B1/HSD11B2, and DNA damage sensors (SMC1A, TREX1, TREX2, SSBP3, RAD1, RAD18, EXO1, POLH, HUS1). RESULTS We found that ESR1, ESR2, HSD11B1, NR3C1, NR3C2, POLH, and SMC1A transcripts were significantly decreased and AR, TREX1, RAD1, and SSBP3 transcripts were increased dramatically in the lesional skin compared to skin samples of controls. CONCLUSION We found that the regulation of the steroidogenic pathway was disrupted in the lesional tissue of psoriasis patients and that a sufficient glucocorticoid and mineralocorticoid response did not form and the estrogen/androgen balance was altered in favour of androgens. We suggest that an increased androgen response in the presence of DDR increases the risk of developing psoriasis. Although this situation may be the cause or the consequence of a disruption of the epidermal barrier, our data suggest developing new therapeutic strategies.
Collapse
Affiliation(s)
- Şeyma Başar Kılıç
- Dermatology and Venereology Department, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| | - Serpil Taheri
- Medical Biology Department, Faculty of Medicine, Erciyes University, Kayseri, Turkey
- Betul Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Ecmel Mehmetbeyoğlu Duman
- Betul Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
- Department of Cancer and Genetics, Cardiff University, Cardiff, CF14 4XN, UK
| | - Eda Öksüm Solak
- Dermatology and Venereology Department, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | | | - Minoo Rassoulzadegan
- Betul Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Murat Borlu
- Dermatology and Venereology Department, Faculty of Medicine, Erciyes University, Kayseri, Turkey.
| |
Collapse
|
25
|
Wang JC, Crosson T, Nikpoor AR, Gupta S, Rafei M, Talbot S. NOCICEPTOR NEURONS CONTROL POLLUTION-MEDIATED NEUTROPHILIC ASTHMA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609202. [PMID: 39229121 PMCID: PMC11370576 DOI: 10.1101/2024.08.22.609202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The immune and sensory nervous systems, having evolved together, use a shared language of receptors and transmitters to maintain homeostasis by responding to external and internal disruptions. Although beneficial in many cases, neurons can exacerbate inflammation during allergic reactions, such as asthma. Our research modeled asthma aggravated by pollution, exposing mice to ambient PM2.5 particles and ovalbumin. This exposure significantly increased bronchoalveolar lavage fluid neutrophils and γδ T cells compared to exposure to ovalbumin alone. We normalized airway inflammation and lung neutrophil levels by silencing nociceptor neurons at inflammation's peak using intranasal QX-314 or ablating TRPV1-expressing neurons. Additionally, we observed heightened sensitivity in chemical-sensing TRPA1 channels in neurons from pollution-exacerbated asthmatic mice. Elevated levels of artemin were detected in the bronchoalveolar lavage fluid from pollution-exposed mice, with artemin levels normalizing in mice with ablated nociceptor neurons. Upon exposure PM2.5 particles, alveolar macrophages expressing pollution-sensing aryl hydrocarbon receptors, were identified as the source of artemin. This molecule enhanced TRPA1 responsiveness and increased neutrophil influx, providing a novel mechanism by which lung-innervating neurons respond to air pollution and suggesting a potential therapeutic target for controlling neutrophilic airway inflammation in asthma, a clinically intractable condition.
Collapse
Affiliation(s)
- Jo-Chiao Wang
- Department of Pharmacology and Physiology, University de Montreal, Canada
| | - Theo Crosson
- Department of Pharmacology and Physiology, University de Montreal, Canada
| | - Amin Reza Nikpoor
- Department of Physiology and Pharmacology, Karolinska Institutet, Sweden
- Department of Biomedical and Molecular Sciences, Queen’s University, Canada
| | - Surbhi Gupta
- Department of Biomedical and Molecular Sciences, Queen’s University, Canada
| | - Moutih Rafei
- Department of Pharmacology and Physiology, University de Montreal, Canada
| | - Sebastien Talbot
- Department of Physiology and Pharmacology, Karolinska Institutet, Sweden
- Department of Biomedical and Molecular Sciences, Queen’s University, Canada
| |
Collapse
|
26
|
Bahman F, Choudhry K, Al-Rashed F, Al-Mulla F, Sindhu S, Ahmad R. Aryl hydrocarbon receptor: current perspectives on key signaling partners and immunoregulatory role in inflammatory diseases. Front Immunol 2024; 15:1421346. [PMID: 39211042 PMCID: PMC11358079 DOI: 10.3389/fimmu.2024.1421346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a versatile environmental sensor and transcription factor found throughout the body, responding to a wide range of small molecules originating from the environment, our diets, host microbiomes, and internal metabolic processes. Increasing evidence highlights AhR's role as a critical regulator of numerous biological functions, such as cellular differentiation, immune response, metabolism, and even tumor formation. Typically located in the cytoplasm, AhR moves to the nucleus upon activation by an agonist where it partners with either the aryl hydrocarbon receptor nuclear translocator (ARNT) or hypoxia-inducible factor 1β (HIF-1β). This complex then interacts with xenobiotic response elements (XREs) to control the expression of key genes. AhR is notably present in various crucial immune cells, and recent research underscores its significant impact on both innate and adaptive immunity. This review delves into the latest insights on AhR's structure, activating ligands, and its multifaceted roles. We explore the sophisticated molecular pathways through which AhR influences immune and lymphoid cells, emphasizing its emerging importance in managing inflammatory diseases. Furthermore, we discuss the exciting potential of developing targeted therapies that modulate AhR activity, opening new avenues for medical intervention in immune-related conditions.
Collapse
Affiliation(s)
- Fatemah Bahman
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Khubaib Choudhry
- Department of Human Biology, University of Toronto, Toronto, ON, Canada
| | - Fatema Al-Rashed
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Fahd Al-Mulla
- Department of Translational Research, Dasman Diabetes Institute, Dasman, Kuwait
| | - Sardar Sindhu
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
- Animal & Imaging Core Facilities, Dasman Diabetes Institute, Dasman, Kuwait
| | - Rasheed Ahmad
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| |
Collapse
|
27
|
Chen Z, Li Y, Tan X, Nie S, Chen B, Mei X, Wu Z. Dysregulated tryptophan metabolism and AhR pathway contributed to CXCL10 upregulation in stable non-segmental vitiligo. J Dermatol Sci 2024; 115:33-41. [PMID: 38955622 DOI: 10.1016/j.jdermsci.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/21/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Tryptophan metabolism dysregulation has been observed in vitiligo. However, drawing a mechanistic linkage between this metabolic disturbance and vitiligo pathogenesis remains challenging. OBJECTIVE Aim to reveal the characterization of tryptophan metabolism in vitiligo and investigate the role of tryptophan metabolites in vitiligo pathophysiology. METHODS LC-MS/MS, dual-luciferase reporter assay, ELISA, qRT-PCR, small interfering RNA, western blotting, and immunohistochemistry were employed. RESULTS Kynurenine pathway activation and KYAT enzyme-associated deviation to kynurenic acid (KYNA) in the plasma of stable non-segmental vitiligo were determined. Using a public microarray dataset, we next validated the activation of kynurenine pathway was related with inflammatory-related genes expression in skin of vitiligo patients. Furthermore, we found that KYNA induced CXCL10 upregulation in keratinocytes via AhR activation. Moreover, the total activity of AhR agonist was increased while the AhR concentration per se was decreased in the plasma of vitiligo patients. Finally, higher KYAT, CXCL10, CYP1A1 and lower AhR expression in vitiligo lesional skin were observed by immunohistochemistry staining. CONCLUSION This study depicts the metabolic and genetic characterizations of tryptophan metabolism in vitiligo and proposes that KYNA, a tryptophan-derived AhR ligand, can enhance CXCL10 expression in keratinocytes.
Collapse
Affiliation(s)
- Zile Chen
- Department of Dermatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiting Li
- Department of Dermatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Tan
- Department of Dermatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Nie
- Department of Dermatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Chen
- Department of Dermatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingyu Mei
- Department of Dermatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhouwei Wu
- Department of Dermatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
28
|
Silverberg JI, Boguniewicz M, Quintana FJ, Clark RA, Gross L, Hirano I, Tallman AM, Brown PM, Fredericks D, Rubenstein DS, McHale KA. Tapinarof validates the aryl hydrocarbon receptor as a therapeutic target: A clinical review. J Allergy Clin Immunol 2024; 154:1-10. [PMID: 38154665 DOI: 10.1016/j.jaci.2023.12.013] [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] [Received: 08/10/2023] [Revised: 11/09/2023] [Accepted: 12/08/2023] [Indexed: 12/30/2023]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-dependent transcription factor that has wide-ranging roles, including regulation of inflammation and homeostasis. AhR is not a cell surface receptor; rather, it exists in a cytoplasmic complex that responds to a wide variety of structurally dissimilar endogenous, microbial, and environmental ligands. The ubiquitous expression of AhR, its ability to be activated by a wide range of ligands, and its capacity to act as a master regulator for gene expression and homeostasis make it a promising new therapeutic target. Clinical trials of tapinarof cream have now validated AhR agonism as a therapeutic approach that can deliver significant efficacy for treating inflammatory skin diseases, including psoriasis and atopic dermatitis. Tapinarof 1% cream is a first-in-class, nonsteroidal, topical, AhR agonist with a pharmacokinetic profile that results in localized exposure at sites of disease, avoiding systemic safety concerns, drug interactions, or off-target effects. Psoriasis and atopic dermatitis both involve epidermal inflammation, cellular immune responses, dysregulation of skin barrier protein expression, and oxidative stress. On the basis of the clinical effectiveness of tapinarof cream for treating inflammatory skin diseases, we review how targeting AhR may offer a significant opportunity in other conditions that share key aspects of pathogenesis, including asthma, inflammatory bowel disease, eosinophilic esophagitis, ophthalmic, and nervous system diseases.
Collapse
Affiliation(s)
| | - Mark Boguniewicz
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, Denver, Colo
| | - Francisco J Quintana
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | | | - Lara Gross
- Dallas Allergy and Asthma Center, and the Allergy and Immunology Division, Baylor University Medical Center, Dallas, Tex
| | - Ikuo Hirano
- Northwestern University Feinberg School of Medicine, Chicago, Ill
| | | | | | | | | | | |
Collapse
|
29
|
Guarnieri T. Light Sensing beyond Vision: Focusing on a Possible Role for the FICZ/AhR Complex in Skin Optotransduction. Cells 2024; 13:1082. [PMID: 38994936 PMCID: PMC11240502 DOI: 10.3390/cells13131082] [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] [Received: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Although our skin is not the primary visual organ in humans, it acts as a light sensor, playing a significant role in maintaining our health and overall well-being. Thanks to the presence of a complex and sophisticated optotransduction system, the skin interacts with the visible part of the electromagnetic spectrum and with ultraviolet (UV) radiation. Following a brief overview describing the main photosensitive molecules that detect specific electromagnetic radiation and their associated cell pathways, we analyze their impact on physiological functions such as melanogenesis, immune response, circadian rhythms, and mood regulation. In this paper, we focus on 6-formylindolo[3,2-b]carbazole (FICZ), a photo oxidation derivative of the essential amino acid tryptophan (Trp). This molecule is the best endogenous agonist of the Aryl hydrocarbon Receptor (AhR), an evolutionarily conserved transcription factor, traditionally recognized as a signal transducer of both exogenous and endogenous chemical signals. Increasing evidence indicates that AhR is also involved in light sensing within the skin, primarily due to its ligand FICZ, which acts as both a chromophore and a photosensitizer. The biochemical reactions triggered by their interaction impact diverse functions and convey crucial data to our body, thus adding a piece to the complex puzzle of pathways that allow us to decode and elaborate environmental stimuli.
Collapse
Affiliation(s)
- Tiziana Guarnieri
- Cell Physiology Laboratory, Department of Biological, Geological, and Environmental Sciences, Alma Mater Studiorum Università di Bologna, Via Francesco Selmi 3, 40126 Bologna, Italy;
- Interdepartmental Center for Industrial Research in Life Sciences and Technologies, University of Bologna, 40126 Bologna, Italy
- Consiglio Nazionale delle Ricerche, Istituto per le Applicazioni del Calcolo “Mauro Picone”, Via dei Taurini 19, 00185 Roma, Italy
| |
Collapse
|
30
|
Hemida AS, Hammam MA, Swilam AA, Shehata WA. Evaluation of OVOL1 and Filaggrin immunohistochemical expression and clinical relevance in psoriasis. Diagn Pathol 2024; 19:84. [PMID: 38907248 PMCID: PMC11191297 DOI: 10.1186/s13000-024-01491-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/26/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Psoriasis is a disease of overactive immune system. OVOL1 and Filaggrin have been associated with many inflammatory skin lesions. To the best of our knowledge, the correlation between OVOL1 and Filaggrin in psoriasis was not previously investigated. This work aims to search the immunohistochemical expression and correlation between OVOL1 and Filaggrin in psoriasis. MATERIALS AND METHODS Slides cut from paraffin blocks of 30 psoriasis cases and 30 control subjects were stained with OVOL1 and Filaggrin. Clinicopathological data were correlated with the results of staining. RESULTS OVOL1 and Filaggrin expression in epidermis showed a significant gradual reduction from normal skin to peri-lesional and psoriasis biopsies (P < 0.001). In contrast, psoriasis dermis showed a significant overexpression of OVOL1 in inflammatory cells in relation to peri-lesional biopsies (P < 0.002). OVOL1 demonstrated a significant direct correlation with Filaggrin expression in psoriasis (r = 0.568, P < 0.004). OVOL1 and Filaggrin expression in psoriasis skin epidermis demonstrated a statistically significant negative correlation with PASI score. CONCLUSION OVOL1 and Filaggrin might be involved in psoriasis-associated inflammation and skin hyperproliferation. OVOL1 might have a protective barrier function in the skin and could be used to stratify progressive disease. Filaggrin may play a role in progression of psoriasis. OVOL1 inhibition could be considered in suppression of Filaggrin function. OVOL1 agonists may be beneficial in psoriasis treatment.
Collapse
Affiliation(s)
- Aiat Shaban Hemida
- Pathology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Shebin El Kom, Egypt.
| | - Mostafa Ahmed Hammam
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| | - Aya Ahmed Swilam
- Department of Dermatology, Andrology and Venereology, Shebin El Kom Teaching Hospital, Menoufia, Egypt
| | - Wafaa Ahmed Shehata
- Department of Dermatology, Andrology and Venereology, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| |
Collapse
|
31
|
Liao Z, Zeng X, Guo X, Shi Q, Tang Z, Li P, Chen C, Chen M, Chen J, Xu J, Cai Y. Targeting the aryl hydrocarbon receptor with FICZ regulates IL-2 and immune infiltration to alleviate Hashimoto's thyroiditis in mice. Eur J Pharmacol 2024; 973:176588. [PMID: 38621508 DOI: 10.1016/j.ejphar.2024.176588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/25/2024] [Accepted: 04/11/2024] [Indexed: 04/17/2024]
Abstract
Hashimoto's thyroiditis (HT) is the most frequent autoimmune disorder. Growing work points to the involvement of aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor, in the regulation of immune homeostasis. However, the roles of AhR and its ligands in HT remains unclear. In this study, we leveraged public human database analyses to postulate that the AhR expression was predominantly in thyroid follicular cells, correlating significantly with the thyroid infiltration levels of multiple immune cells in HT patients. Using a thyroglobulin-induced HT mouse model and in vitro thyroid follicular epithelial cell cultures, we found a significant downregulation of AhR expression in thyrocytes both in vivo and in vitro. Conversely, activating AhR by FICZ, a natural AhR ligand, mitigated inflammation and apoptosis in thyrocytes in vitro and conferred protection against HT in mice. RNA sequencing (RNA-seq) of thyroid tissues indicated that AhR activation moderated HT-associated immune or inflammatory signatures. Further, immunoinfiltration analysis indicated that AhR activation regulated immune cell infiltration in the thyroid of HT mice, such as suppressing cytotoxic CD8+ T cell infiltration and promoting anti-inflammatory M2 macrophage polarization. Concomitantly, the expression levels of interleukin-2 (IL-2), a lymphokine that downregulates immune responses, were typically decreased in HT but restored upon AhR activation. In silico validation substantiated the binding interaction between AhR and IL-2. In conclusion, targeting the AhR with FICZ regulates IL-2 and immune infiltration to alleviate experimental HT, shedding new light on the therapeutic intervention of this prevalent disease.
Collapse
Affiliation(s)
- Zhengzheng Liao
- Department of Pharmacy, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Xianzhong Zeng
- Department of Endocrinology, Ganzhou People's Hospital, Ganzhou, 341000, People's Republic of China
| | - Xiaoling Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Qing Shi
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Ziyun Tang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China
| | - Ping Li
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, 330006, People's Republic of China; Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, 330006, People's Republic of China
| | - Cuiyun Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, 330006, People's Republic of China; Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, 330006, People's Republic of China
| | - Mengxia Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, 330006, People's Republic of China; Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, 330006, People's Republic of China
| | - Jianrong Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, 330006, People's Republic of China; Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, 330006, People's Republic of China
| | - Jixiong Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, 330006, People's Republic of China; Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, 330006, People's Republic of China.
| | - Yaojun Cai
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People's Republic of China; Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, 330006, People's Republic of China; Jiangxi Branch of National Clinical Research Center for Metabolic Disease, Nanchang, 330006, People's Republic of China.
| |
Collapse
|
32
|
Heine S, Alessandrini F, Grosch J, Graß C, Heldner A, Schnautz B, Grosch J, Buters J, Slusarenko BO, Krappmann D, Fallarino F, Ohnmacht C, Schmidt-Weber CB, Blank S. Activation of the aryl hydrocarbon receptor improves allergen-specific immunotherapy of murine allergic airway inflammation: a novel adjuvant option? Front Immunol 2024; 15:1397072. [PMID: 38915403 PMCID: PMC11194380 DOI: 10.3389/fimmu.2024.1397072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/29/2024] [Indexed: 06/26/2024] Open
Abstract
Background Allergen-specific immunotherapy (AIT) is able to restore immune tolerance to allergens in allergic patients. However, some patients do not or only poorly respond to current treatment protocols. Therefore, there is a need for deeper mechanistic insights and further improvement of treatment strategies. The relevance of the aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor, has been investigated in several inflammatory diseases, including allergic asthma. However, its potential role in AIT still needs to be addressed. Methods A murine model of AIT in ovalbumin-induced allergic airway inflammation was performed in AhR-deficient (AhR-/-) and wild-type mice. Furthermore, AIT was combined with the application of the high-affinity AhR agonist 10-chloro-7H-benzimidazo[2,1-a]benzo[de]iso-quinolin-7-one (10-Cl-BBQ) as an adjuvant to investigate the effects of AhR activation on therapeutic outcome. Results Although AhR-/- mice suffer stronger allergic responses than wild-type mice, experimental AIT is comparably effective in both. Nevertheless, combining AIT with the administration of 10-Cl-BBQ improved therapeutic effects by an AhR-dependent mechanism, resulting in decreased cell counts in the bronchoalveolar fluid, decreased pulmonary Th2 and Th17 cell levels, and lower sIgE levels. Conclusion This study demonstrates that the success of AIT is not dependent on the AhR. However, targeting the AhR during AIT can help to dampen inflammation and improve tolerogenic vaccination. Therefore, AhR ligands might represent promising candidates as immunomodulators to enhance the efficacy of AIT.
Collapse
Affiliation(s)
- Sonja Heine
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Francesca Alessandrini
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Johannes Grosch
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Carina Graß
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutic Center, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Alexander Heldner
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Benjamin Schnautz
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Johanna Grosch
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Jeroen Buters
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Benjamin O. Slusarenko
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Daniel Krappmann
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutic Center, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | | | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Carsten B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Health & Helmholtz Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, Munich, Germany
| |
Collapse
|
33
|
Lee YG, Jung Y, Choi HK, Lee JI, Lim TG, Lee J. Natural Product-Derived Compounds Targeting Keratinocytes and Molecular Pathways in Psoriasis Therapeutics. Int J Mol Sci 2024; 25:6068. [PMID: 38892253 PMCID: PMC11172960 DOI: 10.3390/ijms25116068] [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] [Received: 04/17/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Psoriasis is a chronic autoimmune inflammatory skin disorder that affects approximately 2-3% of the global population due to significant genetic predisposition. It is characterized by an uncontrolled growth and differentiation of keratinocytes, leading to the formation of scaly erythematous plaques. Psoriasis extends beyond dermatological manifestations to impact joints and nails and is often associated with systemic disorders. Although traditional treatments provide relief, their use is limited by potential side effects and the chronic nature of the disease. This review aims to discuss the therapeutic potential of keratinocyte-targeting natural products in psoriasis and highlight their efficacy and safety in comparison with conventional treatments. This review comprehensively examines psoriasis pathogenesis within keratinocytes and the various related signaling pathways (such as JAK-STAT and NF-κB) and cytokines. It presents molecular targets such as high-mobility group box-1 (HMGB1), dual-specificity phosphatase-1 (DUSP1), and the aryl hydrocarbon receptor (AhR) for treating psoriasis. It evaluates the ability of natural compounds such as luteolin, piperine, and glycyrrhizin to modulate psoriasis-related pathways. Finally, it offers insights into alternative and sustainable treatment options with fewer side effects.
Collapse
Affiliation(s)
- Yu Geon Lee
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.G.L.); (Y.J.); (H.-K.C.); (J.-I.L.)
| | - Younjung Jung
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.G.L.); (Y.J.); (H.-K.C.); (J.-I.L.)
| | - Hyo-Kyoung Choi
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.G.L.); (Y.J.); (H.-K.C.); (J.-I.L.)
| | - Jae-In Lee
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.G.L.); (Y.J.); (H.-K.C.); (J.-I.L.)
| | - Tae-Gyu Lim
- Department of Food Science & Biotechnology, College of Life Sciences, Sejong University, Seoul 05006, Republic of Korea;
- Carbohydrate Bioproduct Research Center, Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Jangho Lee
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (Y.G.L.); (Y.J.); (H.-K.C.); (J.-I.L.)
| |
Collapse
|
34
|
Lin X, Meng X, Lin J. The Role of Aryl Hydrocarbon Receptor in the Pathogenesis and Treatment of Psoriasis. J Cutan Med Surg 2024; 28:276-286. [PMID: 38497283 DOI: 10.1177/12034754241239050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The pathogenesis of psoriasis is complex. Aryl hydrocarbon receptor (AhR) is a transcription factor that can be bound and activated by structurally diverse ligands and plays an important role in a range of biological processes and in the pathogenesis of different diseases. Recently, the role of AhR in psoriasis has attracted attention. AhR has toxicological functions and physiological functions. The overexpression and activation of AhR induced by the environmental pollutant and exogenous AhR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can drive the development of psoriasis. This TCDD-mediated toxicological response disrupts the physiological functions of AhR resulting in skin barrier disorders and the release of inflammatory cytokines, 2 of the pivotal factors of psoriasis. In addition, highly upregulated kynureninase in psoriasis decreases endogenous AhR agonists, thereby weakening the physiological functions of AhR. Activating AhR physiological signalling should be useful in the treatment of psoriasis. Studies have demonstrated that physiological activation of AhR can dampen the severity of psoriasis. The oldest and effective treatment for psoriasis coal tar works by activating AhR, and both new anti-psoriasis drugs tapinarof and benvitimod are formulations of AhR agonist, supporting that activation of AhR can be used as a new strategy for the treatment of psoriasis. Preclinical and preliminary clinical studies have revealed the anti-psoriasis effects of a number of AhR agonists, providing potential candidates for the development of new drugs for the treatment of psoriasis.
Collapse
Affiliation(s)
- Xiran Lin
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xianmin Meng
- Department of Pathology and Laboratory Medicine, Axia Women's Health, Oaks, PA, USA
| | - Jingrong Lin
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
35
|
Cai H, Wen H, Li J, Lu L, Zhao W, Jiang X, Bai R. Small-molecule agents for treating skin diseases. Eur J Med Chem 2024; 268:116269. [PMID: 38422702 DOI: 10.1016/j.ejmech.2024.116269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/16/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024]
Abstract
Skin diseases are a class of common and frequently occurring diseases that significantly impact daily lives. Currently, the limited effective therapeutic drugs are far from meeting the clinical needs; most drugs typically only provide symptomatic relief rather than a cure. Developing small-molecule drugs with improved efficacy holds paramount importance for treating skin diseases. This review aimed to systematically introduce the pathogenesis of common skin diseases in daily life, list related drugs applied in the clinic, and summarize the clinical research status of candidate drugs and the latest research progress of candidate compounds in the drug discovery stage. Also, it statistically analyzed the number of publications and global attention trends for the involved skin diseases. This review might provide practical information for researchers engaged in dermatological drugs and further increase research attention to this disease area.
Collapse
Affiliation(s)
- Hong Cai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Hao Wen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Junjie Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Liuxin Lu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Wenxuan Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Xiaoying Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China.
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China.
| |
Collapse
|
36
|
Zhang Y, Tu S, Ji X, Wu J, Meng J, Gao J, Shao X, Shi S, Wang G, Qiu J, Zhang Z, Hua C, Zhang Z, Chen S, Zhang L, Zhu SJ. Dubosiella newyorkensis modulates immune tolerance in colitis via the L-lysine-activated AhR-IDO1-Kyn pathway. Nat Commun 2024; 15:1333. [PMID: 38351003 PMCID: PMC10864277 DOI: 10.1038/s41467-024-45636-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
Commensal bacteria generate immensely diverse active metabolites to maintain gut homeostasis, however their fundamental role in establishing an immunotolerogenic microenvironment in the intestinal tract remains obscure. Here, we demonstrate that an understudied murine commensal bacterium, Dubosiella newyorkensis, and its human homologue Clostridium innocuum, have a probiotic immunomodulatory effect on dextran sulfate sodium-induced colitis using conventional, antibiotic-treated and germ-free mouse models. We identify an important role for the D. newyorkensis in rebalancing Treg/Th17 responses and ameliorating mucosal barrier injury by producing short-chain fatty acids, especially propionate and L-Lysine (Lys). We further show that Lys induces the immune tolerance ability of dendritic cells (DCs) by enhancing Trp catabolism towards the kynurenine (Kyn) pathway through activation of the metabolic enzyme indoleamine-2,3-dioxygenase 1 (IDO1) in an aryl hydrocarbon receptor (AhR)-dependent manner. This study identifies a previously unrecognized metabolic communication by which Lys-producing commensal bacteria exert their immunoregulatory capacity to establish a Treg-mediated immunosuppressive microenvironment by activating AhR-IDO1-Kyn metabolic circuitry in DCs. This metabolic circuit represents a potential therapeutic target for the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Yanan Zhang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Shuyu Tu
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510080, PR China
| | - Xingwei Ji
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Jianan Wu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, PR China
| | - Jinxin Meng
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, 266109, PR China
| | - Jinsong Gao
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Xian Shao
- Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, Zhejiang, 312000, PR China
| | - Shuai Shi
- Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, Zhejiang, 312000, PR China
| | - Gan Wang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Jingjing Qiu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, PR China
| | - Zhuobiao Zhang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Chengang Hua
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Ziyi Zhang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Shuxian Chen
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, 510080, PR China
| | - Shu Jeffrey Zhu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, PR China.
- Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, Zhejiang, 312000, PR China.
| |
Collapse
|
37
|
Dec M, Arasiewicz H. Aryl hydrocarbon receptor role in chronic inflammatory skin diseases: a narrative review. Postepy Dermatol Alergol 2024; 41:9-19. [PMID: 38533374 PMCID: PMC10962376 DOI: 10.5114/ada.2023.135617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/05/2023] [Indexed: 03/28/2024] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that has gained increasing attention in the field of dermatology due to its multifaceted role in skin health and disease. This review provides a comprehensive overview of the current state of knowledge regarding the AHR and its implications in dermatological conditions. The AHR is well known for its involvement in xenobiotic metabolism, particularly in response to polycyclic aromatic hydrocarbons and dioxins. However, recent research has unveiled its pivotal role in the skin immune response, barrier function, and homeostasis. The AHR signalling pathway is intricately linked to various dermatological disorders, including psoriasis, atopic dermatitis, acne and hidradenitis suppurativa. In this review, we delve into the molecular mechanisms through which AHR activation influences skin physiology and highlight how dysregulation can lead to pathological conditions. Moreover, we discuss the emerging therapeutic potential of AHR modulators in the treatment of skin diseases. In conclusion, the AHR is a pivotal player in dermatology, with a multifaceted role in skin physiology and pathology. Understanding the intricacies of AHR signalling in the skin offers promising avenues for the development of novel therapies and preventive strategies for various dermatological conditions. Further research is warranted to elucidate the full scope of AHR's contributions to dermatology and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Michał Dec
- Department of Dermatology and Vascular Anomalies, John Paul II Centre of Paediatrics, Sosnowiec, Poland
| | - Hubert Arasiewicz
- Department of Dermatology and Vascular Anomalies, John Paul II Centre of Paediatrics, Sosnowiec, Poland
| |
Collapse
|
38
|
Furuya H, Nguyen CT, Chan T, Marusina AI, Merleev AA, Garcia-Hernandez MDLL, Hsieh SL, Tsokos GC, Ritchlin CT, Tagkopoulos I, Maverakis E, Adamopoulos IE. IL-23 induces CLEC5A + IL-17A + neutrophils and elicit skin inflammation associated with psoriatic arthritis. J Autoimmun 2024; 143:103167. [PMID: 38301504 PMCID: PMC10981569 DOI: 10.1016/j.jaut.2024.103167] [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: 10/04/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 02/03/2024]
Abstract
IL-23-activation of IL-17 producing T cells is involved in many rheumatic diseases. Herein, we investigate the role of IL-23 in the activation of myeloid cell subsets that contribute to skin inflammation in mice and man. IL-23 gene transfer in WT, IL-23RGFP reporter mice and subsequent analysis with spectral cytometry show that IL-23 regulates early innate immune events by inducing the expansion of a myeloid MDL1+CD11b+Ly6G+ population that dictates epidermal hyperplasia, acanthosis, and parakeratosis; hallmark pathologic features of psoriasis. Genetic ablation of MDL-1, a major PU.1 transcriptional target during myeloid differentiation exclusively expressed in myeloid cells, completely prevents IL-23-pathology. Moreover, we show that IL-23-induced myeloid subsets are also capable of producing IL-17A and IL-23R+MDL1+ cells are present in the involved skin of psoriasis patients and gene expression correlations between IL-23 and MDL-1 have been validated in multiple patient cohorts. Collectively, our data demonstrate a novel role of IL-23 in MDL-1-myelopoiesis that is responsible for skin inflammation and related pathologies. Our data open a new avenue of investigations regarding the role of IL-23 in the activation of myeloid immunoreceptors and their role in autoimmunity.
Collapse
Affiliation(s)
- Hiroki Furuya
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Cuong Thach Nguyen
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, USA
| | - Trevor Chan
- Department of Computer Science, University of California, Davis, CA, USA; Genome Center, University of California, Davis, CA, USA
| | - Alina I Marusina
- Department of Dermatology, University of California, Davis, Sacramento, USA
| | | | | | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, Nankang, Taipei, Taiwan
| | - George C Tsokos
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, USA
| | - Christopher T Ritchlin
- Division of Allergy, Immunology & Rheumatology, University of Rochester Medical School, NY, USA
| | - Ilias Tagkopoulos
- Department of Computer Science, University of California, Davis, CA, USA; Process Integration and Predictive Analytics, PIPA LLC, CA, USA
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, USA
| | - Iannis E Adamopoulos
- Department of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA; Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, USA.
| |
Collapse
|
39
|
Chen Z, Dragan M, Sun P, Haensel D, Vu R, Cui L, Shi Y, Dai X. An AhR-Ovol1-Id1 regulatory axis in keratinocytes promotes skin homeostasis against atopic dermatitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.29.577821. [PMID: 38352592 PMCID: PMC10862726 DOI: 10.1101/2024.01.29.577821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Skin is our outer permeability and immune defense barrier against myriad external assaults. Aryl hydrocarbon receptor (AhR) senses environmental factors and regulates barrier robustness and immune homeostasis. AhR agonist is in clinical trial for atopic dermatitis (AD) treatment, but the underlying mechanism of action remains ill-defined. Here we report OVOL1/Ovol1 as a conserved and direct transcriptional target of AhR in epidermal keratinocytes. We show that OVOL1/Ovol1 impacts AhR regulation of keratinocyte gene expression, and Ovol1 deletion in keratinocytes hampers AhR's barrier promotion function and worsens AD-like inflammation. Mechanistically, we identify Ovol1's direct downstream targets genome-wide, and provide in vivo evidence for Id1's critical role in barrier maintenance and disease suppression. Furthermore, our findings reveal an IL-1/dermal γδT cell axis exacerbating both type 2 and type 3 immune responses downstream of barrier perturbation in Ovol1 -deficient AD skin. Finally, we present data suggesting the clinical relevance of OVOL1 and ID1 function in human AD. Our study highlights a keratinocyte-intrinsic AhR-Ovol1-Id1 regulatory axis that promotes both epidermal and immune homeostasis against AD-like inflammation, implicating new therapeutic targets for AD.
Collapse
|
40
|
Lu YW, Dong RJ, Yang LH, Liu J, Yang T, Xiao YH, Chen YJ, Wang RR, Li YY. Identification of gene signatures and molecular mechanisms underlying the mutual exclusion between psoriasis and leprosy. Sci Rep 2024; 14:2199. [PMID: 38273053 PMCID: PMC10810956 DOI: 10.1038/s41598-024-52783-0] [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] [Received: 06/30/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024] Open
Abstract
Leprosy and psoriasis rarely coexist, the specific molecular mechanisms underlying their mutual exclusion have not been extensively investigated. This study aimed to reveal the underlying mechanism responsible for the mutual exclusion between psoriasis and leprosy. We obtained leprosy and psoriasis data from ArrayExpress and GEO database. Differential expression analysis was conducted separately on the leprosy and psoriasis using DEseq2. Differentially expressed genes (DEGs) with opposite expression patterns in psoriasis and leprosy were identified, which could potentially involve in their mutual exclusion. Enrichment analysis was performed on these candidate mutually exclusive genes, and a protein-protein interaction (PPI) network was constructed to identify hub genes. The expression of these hub genes was further validated in an external dataset to obtain the critical mutually exclusive genes. Additionally, immune cell infiltration in psoriasis and leprosy was analyzed using single-sample gene set enrichment analysis (ssGSEA), and the correlation between critical mutually exclusive genes and immune cells was also examined. Finally, the expression pattern of critical mutually exclusive genes was evaluated in a single-cell transcriptome dataset. We identified 1098 DEGs in the leprosy dataset and 3839 DEGs in the psoriasis dataset. 48 candidate mutually exclusive genes were identified by taking the intersection. Enrichment analysis revealed that these genes were involved in cholesterol metabolism pathways. Through PPI network analysis, we identified APOE, CYP27A1, FADS1, and SOAT1 as hub genes. APOE, CYP27A1, and SOAT1 were subsequently validated as critical mutually exclusive genes on both internal and external datasets. Analysis of immune cell infiltration indicated higher abundance of 16 immune cell types in psoriasis and leprosy compared to normal controls. The abundance of 6 immune cell types in psoriasis and leprosy positively correlated with the expression levels of APOE and CYP27A1. Single-cell data analysis demonstrated that critical mutually exclusive genes were predominantly expressed in Schwann cells and fibroblasts. This study identified APOE, CYP27A1, and SOAT1 as critical mutually exclusive genes. Cholesterol metabolism pathway illustrated the possible mechanism of the inverse association of psoriasis and leprosy. The findings of this study provide a basis for identifying mechanisms and therapeutic targets for psoriasis.
Collapse
Affiliation(s)
- You-Wang Lu
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Kidney Diseases, Medical College, Hubei Polytechnic University, Huangshi, China
| | - Rong-Jing Dong
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Kidney Diseases, Medical College, Hubei Polytechnic University, Huangshi, China
| | - Lu-Hui Yang
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Jiang Liu
- Department of Reproduction and Genetics, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Ting Yang
- Department of Dermatology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| | - Yong-Hong Xiao
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yong-Jun Chen
- Department of Dermatology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China.
| | - Rui-Rui Wang
- College of Pharmaceutical Sciences, Yunnan University of Traditional Chinese Medicine, Kunming, 650500, China.
| | - Yu-Ye Li
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
41
|
Huang Y, Chen L, Liu F, Xiong X, Ouyang Y, Deng Y. Tryptophan, an important link in regulating the complex network of skin immunology response in atopic dermatitis. Front Immunol 2024; 14:1300378. [PMID: 38318507 PMCID: PMC10839033 DOI: 10.3389/fimmu.2023.1300378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/31/2023] [Indexed: 02/07/2024] Open
Abstract
Atopic dermatitis (AD) is a common chronic relapsing inflammatory skin disease, of which the pathogenesis is a complex interplay between genetics and environment. Although the exact mechanisms of the disease pathogenesis remain unclear, the immune dysregulation primarily involving the Th2 inflammatory pathway and accompanied with an imbalance of multiple immune cells is considered as one of the critical etiologies of AD. Tryptophan metabolism has long been firmly established as a key regulator of immune cells and then affect the occurrence and development of many immune and inflammatory diseases. But the relationship between tryptophan metabolism and the pathogenesis of AD has not been profoundly discussed throughout the literatures. Therefore, this review is conducted to discuss the relationship between tryptophan metabolism and the complex network of skin inflammatory response in AD, which is important to elucidate its complex pathophysiological mechanisms, and then lead to the development of new therapeutic strategies and drugs for the treatment of this frequently relapsing disease.
Collapse
Affiliation(s)
- Yaxin Huang
- Department of Dermatology & Sexually Transmitted Disease (STD), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Lingna Chen
- Department of Dermatology & Sexually Transmitted Disease (STD), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Fuming Liu
- Department of Dermatology & Sexually Transmitted Disease (STD), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xia Xiong
- Department of Dermatology & Sexually Transmitted Disease (STD), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yongliang Ouyang
- Department of Dermatology & Sexually Transmitted Disease (STD), Chengdu First People’s Hospital, Chengdu, Sichuan, China
- Health Management Center, Luzhou People’s Hospital, Luzhou, China
| | - Yongqiong Deng
- Department of Dermatology & Sexually Transmitted Disease (STD), the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Dermatology & Sexually Transmitted Disease (STD), Chengdu First People’s Hospital, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Kim HR, Lee SY, You GE, Park CW, Kim HO, Chung BY. Exosomes released by environmental pollutant-stimulated Keratinocytes/PBMCs can trigger psoriatic inflammation in recipient cells via the AhR signaling pathway. Front Mol Biosci 2024; 10:1324692. [PMID: 38288335 PMCID: PMC10822922 DOI: 10.3389/fmolb.2023.1324692] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 11/29/2023] [Indexed: 01/31/2024] Open
Abstract
Introduction: Exosomes, pivotal in intercellular communication during skin disease pathogenesis, have garnered substantial attention. However, the impact of environmental pollutants, such as benzo[a]pyrene (BaP) and 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD), on exosome release amid inflammatory skin diseases remains unexplored. This study addresses this gap by examining the influence of BaP and TCDD on exosome function, specifically focusing on immune-related pathway alterations in normal recipient keratinocytes and peripheral blood mononuclear cells (PBMCs). Methods: HaCaT cells were treated with exosomes from BaP- or TCDD-treated keratinocytes. Proinflammatory cytokines and chemokines, including TNF-α, IL-1β, IL-6, IL-8, CXCL1, and CXCL5, were assessed. The involvement of the p65NF-κB/p38MAPK/ERK signaling pathway in recipient keratinocytes was investigated. Aryl hydrocarbon receptor (AhR) silencing was employed to elucidate its role in mediating the proinflammatory response induced by exosomes from BaP- or TCDD-treated keratinocytes. Results and discussion: Treatment with exosomes from BaP- or TCDD-treated keratinocytes induced a significant increase in proinflammatory cytokines and chemokines in HaCaT cells. The upregulation implicated the p65NF-κB/p38MAPK/ERK signaling pathway. AhR silencing attenuated this response, suggesting a role for AhR in mediating this response. In PBMCs from healthy controls, exosomes from BaP-stimulated PBMCs of psoriatic patients led to increased expression of proinflammatory cytokines and modulation of Th1/Th17 cell distribution via AhR activation. These findings unveil a novel dimension in the interplay between environmental xenobiotic agents (BaP and TCDD) and exosomal functions. The study establishes their influence on psoriatic inflammatory responses, shedding light on the underlying mechanisms mediated through the AhR signaling pathway in recipient keratinocytes and PBMCs.
Collapse
Affiliation(s)
- Hye Ran Kim
- Department of Dermatology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - So Yeon Lee
- Department of Dermatology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Ga Eun You
- Research and Development Institute, Biosolution, Seoul, Republic of Korea
| | - Chun Wook Park
- Department of Dermatology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Hye One Kim
- Department of Dermatology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Bo Young Chung
- Department of Dermatology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
43
|
Zhu X, Han R, Tian X, Hochgerner M, Li H, Wang J, Xia J. The opposite effect of tapinarof between IMQ and IL-23 induced psoriasis mouse models. Exp Dermatol 2024; 33:e14862. [PMID: 37350230 DOI: 10.1111/exd.14862] [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] [Received: 02/20/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023]
Abstract
Tapinarof is an aryl hydrocarbon receptor (AHR) ligand which is used to treat plaque psoriasis in adults. However, the underlying mechanism is not yet fully understood. In this study, we applied two of the most studied psoriasis mouse models: topical application of imiquimod (IMQ) and subcutaneous injection of IL-23. Although both models successfully induced psoriasis-like lesions in mice, tapinarof had a completely opposite effect on the two models. Tapinarof decreased the expression of multiple essential cytokines involved in the pathological IL-23/IL-17/IL-22 axis and ameliorated IMQ-induced psoriatic dermatitis, inhibiting keratinocyte proliferation and abnormal differentiation. However, in the IL-23-injection-model, tapinarof instead aggravated the disease. Here, tapinarof increased epidermal thickness and differentiated epidermal dysplasia in mice. Our data suggest that tapinarof may have different effects on varied types of psoriasis.
Collapse
Affiliation(s)
- Xingyu Zhu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Institute for Six-Sector Economy, Fudan University, Shanghai, China
| | - Ruomei Han
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaoxue Tian
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China
| | - Mathias Hochgerner
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China
| | - Hui Li
- MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai, China
| | - Jingjing Xia
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
44
|
Rios-Carlos M, Cervantes-García D, Córdova-Dávalos LE, Bermúdez-Humarán LG, Salinas E. Unraveling the gut-skin axis in atopic dermatitis: exploiting insights for therapeutic strategies. Gut Microbes 2024; 16:2430420. [PMID: 39601281 PMCID: PMC11610564 DOI: 10.1080/19490976.2024.2430420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/14/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Gut microbiota exert functions of high importance in the intestine. Furthermore, there is increasing evidence for its role in immune regulation and maintenance of homeostasis in many physiological processes taking place in distant tissues. In particular, in this review, we explore the impact of metabolites produced by the gut microbiota on the development of atopic dermatitis (AD). Probiotics and prebiotics balance the microbiota and promote the generation of bacterial metabolites, such as short-chain fatty acids and tryptophan derivates, which promote the regulation of the exacerbated AD immune response through regulatory T cells and IL-10 and TGF-β cytokines. Metabolites also have a direct action on keratinocytes once they reach the bloodstream. Besides, probiotics decrease the levels of metabolites associated with AD onset, such as phenols. Understanding all these crosstalk processes between the gut and the skin reveals a number of possibilities, mainly through the manipulation of the gut microbiome, which may represent therapeutic strategies that can contribute to the standard treatments of AD patients to improve their quality of life.
Collapse
Affiliation(s)
- Marcela Rios-Carlos
- Department of Microbiology, Universidad Autónoma de Aguascalientes. Av. Universidad 940, Aguascalientes, Mexico
| | - Daniel Cervantes-García
- Department of Microbiology, Universidad Autónoma de Aguascalientes. Av. Universidad 940, Aguascalientes, Mexico
- Consejo Nacional de Humanidades Ciencias y Tecnologías, Ciudad de México, México
| | - Laura E. Córdova-Dávalos
- Department of Microbiology, Universidad Autónoma de Aguascalientes. Av. Universidad 940, Aguascalientes, Mexico
| | | | - Eva Salinas
- Department of Microbiology, Universidad Autónoma de Aguascalientes. Av. Universidad 940, Aguascalientes, Mexico
| |
Collapse
|
45
|
Larsen MC, Rondelli CM, Almeldin A, Song YS, N’Jai A, Alexander DL, Forsberg EC, Sheibani N, Jefcoate CR. AhR and CYP1B1 Control Oxygen Effects on Bone Marrow Progenitor Cells: The Enrichment of Multiple Olfactory Receptors as Potential Microbiome Sensors. Int J Mol Sci 2023; 24:16884. [PMID: 38069208 PMCID: PMC10706615 DOI: 10.3390/ijms242316884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Polycyclic aromatic hydrocarbon (PAH) pollutants and microbiome products converge on the aryl hydrocarbon receptor (AhR) to redirect selective rapid adherence of isolated bone marrow (BM) cells. In young adult mice, Cyp1b1-deficiency and AhR activation by PAH, particularly when prolonged by Cyp1a1 deletion, produce matching gene stimulations in these BM cells. Vascular expression of Cyp1b1 lowers reactive oxygen species (ROS), suppressing NF-κB/RelA signaling. PAH and allelic selectivity support a non-canonical AhR participation, possibly through RelA. Genes stimulated by Cyp1b1 deficiency were further resolved according to the effects of Cyp1b1 and Cyp1a1 dual deletions (DKO). The adherent BM cells show a cluster of novel stimulations, including select developmental markers; multiple re-purposed olfactory receptors (OLFR); and α-Defensin, a microbial disruptor. Each one connects to an enhanced specific expression of the catalytic RNA Pol2 A subunit, among 12 different subunits. Mesenchymal progenitor BMS2 cells retain these features. Cyp1b1-deficiency removes lymphocytes from adherent assemblies as BM-derived mesenchymal stromal cells (BM-MSC) expand. Cyp1b1 effects were cell-type specific. In vivo, BM-MSC Cyp1b1 expression mediated PAH suppression of lymphocyte progenitors. In vitro, OP9-MSC sustained these progenitors, while Csf1 induced monocyte progenitor expansion to macrophages. Targeted Cyp1b1 deletion (Cdh5-Cre; Cyp1b1fl/fl) established endothelium control of ROS that directs AhR-mediated suppression of B cell progenitors. Monocyte Cyp1b1 deletion (Lyz2-Cre; Cyp1b1fl/fl) selectively attenuated M1 polarization of expanded macrophages, but did not enhance effects on basal M2 polarization. Thus, specific sources of Cyp1b1 link to AhR and to an OLFR network to provide BM inflammatory modulation via diverse microbiome products.
Collapse
Affiliation(s)
- Michele C. Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (A.A.)
| | | | - Ahmed Almeldin
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (A.A.)
| | - Yong-Seok Song
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Alhaji N’Jai
- Department of Pathobiological Sciences, University of Wisconsin, Madison, WI 53706, USA;
| | - David L. Alexander
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA 95064, USA; (D.L.A.); (E.C.F.)
| | - E. Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA 95064, USA; (D.L.A.); (E.C.F.)
| | - Nader Sheibani
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (A.A.)
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Colin R. Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (A.A.)
| |
Collapse
|
46
|
Guo J, Zhang H, Lin W, Lu L, Su J, Chen X. Signaling pathways and targeted therapies for psoriasis. Signal Transduct Target Ther 2023; 8:437. [PMID: 38008779 PMCID: PMC10679229 DOI: 10.1038/s41392-023-01655-6] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 11/28/2023] Open
Abstract
Psoriasis is a common, chronic, and inflammatory skin disease with a high burden on individuals, health systems, and society worldwide. With the immunological pathologies and pathogenesis of psoriasis becoming gradually revealed, the therapeutic approaches for this disease have gained revolutionary progress. Nevertheless, the mechanisms of less common forms of psoriasis remain elusive. Furthermore, severe adverse effects and the recurrence of disease upon treatment cessation should be noted and addressed during the treatment, which, however, has been rarely explored with the integration of preliminary findings. Therefore, it is crucial to have a comprehensive understanding of the mechanisms behind psoriasis pathogenesis, which might offer new insights for research and lead to more substantive progress in therapeutic approaches and expand clinical options for psoriasis treatment. In this review, we looked to briefly introduce the epidemiology, clinical subtypes, pathophysiology, and comorbidities of psoriasis and systematically discuss the signaling pathways involving extracellular cytokines and intracellular transmission, as well as the cross-talk between them. In the discussion, we also paid more attention to the potential metabolic and epigenetic mechanisms of psoriasis and the molecular mechanistic cascades related to its comorbidities. This review also outlined current treatment for psoriasis, especially targeted therapies and novel therapeutic strategies, as well as the potential mechanism of disease recurrence.
Collapse
Affiliation(s)
- Jia Guo
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Hanyi Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Wenrui Lin
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Lixia Lu
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Juan Su
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China.
| |
Collapse
|
47
|
Wojciech L, Png CW, Koh EY, Kioh DYQ, Deng L, Wang Z, Wu L, Hamidinia M, Tung DWH, Zhang W, Pettersson S, Chan ECY, Zhang Y, Tan KSW, Gascoigne NRJ. A tryptophan metabolite made by a gut microbiome eukaryote induces pro-inflammatory T cells. EMBO J 2023; 42:e112963. [PMID: 37743772 PMCID: PMC10620759 DOI: 10.15252/embj.2022112963] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 08/11/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023] Open
Abstract
The large intestine harbors microorganisms playing unique roles in host physiology. The beneficial or detrimental outcome of host-microbiome coexistence depends largely on the balance between regulators and responder intestinal CD4+ T cells. We found that ulcerative colitis-like changes in the large intestine after infection with the protist Blastocystis ST7 in a mouse model are associated with reduction of anti-inflammatory Treg cells and simultaneous expansion of pro-inflammatory Th17 responders. These alterations in CD4+ T cells depended on the tryptophan metabolite indole-3-acetaldehyde (I3AA) produced by this single-cell eukaryote. I3AA reduced the Treg subset in vivo and iTreg development in vitro by modifying their sensing of TGFβ, concomitantly affecting recognition of self-flora antigens by conventional CD4+ T cells. Parasite-derived I3AA also induces over-exuberant TCR signaling, manifested by increased CD69 expression and downregulation of co-inhibitor PD-1. We have thus identified a new mechanism dictating CD4+ fate decisions. The findings thus shine a new light on the ability of the protist microbiome and tryptophan metabolites, derived from them or other sources, to modulate the adaptive immune compartment, particularly in the context of gut inflammatory disorders.
Collapse
Affiliation(s)
- Lukasz Wojciech
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Chin Wen Png
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Immunology Programme, Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Eileen Y Koh
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Dorinda Yan Qin Kioh
- Department of Pharmacy, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Lei Deng
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Ziteng Wang
- Department of Pharmacy, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Liang‐zhe Wu
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Maryam Hamidinia
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Desmond WH Tung
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Wei Zhang
- ASEAN Microbiome Nutrition CentreNational Neuroscience InstituteSingaporeSingapore
| | - Sven Pettersson
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- ASEAN Microbiome Nutrition CentreNational Neuroscience InstituteSingaporeSingapore
- Faculty of Medical SciencesSunway UniversitySubang JayaMalaysia
- Department of OdontologyKarolinska InstitutetStockholmSweden
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Yongliang Zhang
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Immunology Programme, Life Sciences InstituteNational University of SingaporeSingaporeSingapore
| | - Kevin SW Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Nicholas RJ Gascoigne
- Immunology Translational Research Programme, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- ASEAN Microbiome Nutrition CentreNational Neuroscience InstituteSingaporeSingapore
| |
Collapse
|
48
|
Spencer RK, Jin JQ, Elhage KG, Davis MS, Liao W, Bhutani T. Management of Plaque Psoriasis in Adults: Clinical Utility of Tapinarof Cream. PSORIASIS (AUCKLAND, N.Z.) 2023; 13:59-69. [PMID: 37905185 PMCID: PMC10613418 DOI: 10.2147/ptt.s393997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 10/16/2023] [Indexed: 11/02/2023]
Abstract
Topical medications represent the most commonly used drugs in the treatment of psoriasis. However, topical steroids are mainly limited to short-term or intermittent use, and traditional non-steroidal topicals such as vitamin D analogues, topical calcineurin inhibitors, and topical retinoids are limited by low efficacy and poor local skin tolerability. Tapinarof (GSK2894512, DMVT-505) is a novel, topical aryl hydrocarbon receptor (AHR) agonist, which was recently approved by the FDA for the treatment of plaque psoriasis in adults. Tapinarof acts to improve psoriasis through diminished IL-17A production by CD4+ T cells, increased barrier gene expression in keratinocytes, and reduced production of reactive oxygen species. Both short-term and long-term efficacy and safety have been evaluated in two Phase II and two Phase III (PSOARING 1 and 2) clinical trials in addition to a long-term extension study (PSOARING 3). Overall, the drug has shown beneficial effects in achieving clear skin in adults with moderate-to-severe psoriasis, good local tolerability, and also a long duration of effect even after discontinuation of the drug. Therefore, this therapy provides a new, highly effective and safe non-steroidal option to add to our psoriasis treatment toolbox for both initial clearance and long-term maintenance of disease.
Collapse
Affiliation(s)
- Riley K Spencer
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, USA
| | - Joy Q Jin
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, USA
- School of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Kareem G Elhage
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, USA
| | - Mitchell S Davis
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, USA
| | - Wilson Liao
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, USA
| | - Tina Bhutani
- Department of Dermatology, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
49
|
Hou JJ, Ma AH, Qin YH. Activation of the aryl hydrocarbon receptor in inflammatory bowel disease: insights from gut microbiota. Front Cell Infect Microbiol 2023; 13:1279172. [PMID: 37942478 PMCID: PMC10628454 DOI: 10.3389/fcimb.2023.1279172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disease that affects more than 3.5 million people, with rising prevalence. It deeply affects patients' daily life, increasing the burden on patients, families, and society. Presently, the etiology of IBD remains incompletely clarified, while emerging evidence has demonstrated that altered gut microbiota and decreased aryl hydrocarbon receptor (AHR) activity are closely associated with IBD. Furthermore, microbial metabolites are capable of AHR activation as AHR ligands, while the AHR, in turn, affects the microbiota through various pathways. In light of the complex connection among gut microbiota, the AHR, and IBD, it is urgent to review the latest research progress in this field. In this review, we describe the role of gut microbiota and AHR activation in IBD and discussed the crosstalk between gut microbiota and the AHR in the context of IBD. Taken as a whole, we propose new therapeutic strategies targeting the AHR-microbiota axis for IBD, even for other related diseases caused by AHR-microbiota dysbiosis.
Collapse
Affiliation(s)
| | | | - Yue-Hua Qin
- Department of Gastroenterology, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
50
|
Bouchard KV, Costin GE. Promoting New Approach Methodologies (NAMs) for research on skin color changes in response to environmental stress factors: tobacco and air pollution. FRONTIERS IN TOXICOLOGY 2023; 5:1256399. [PMID: 37886123 PMCID: PMC10598764 DOI: 10.3389/ftox.2023.1256399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/25/2023] [Indexed: 10/28/2023] Open
Abstract
Aging is one of the most dynamic biological processes in the human body and is known to carry significant impacts on individuals' self-esteem. Skin pigmentation is a highly heritable trait made possible by complex, strictly controlled cellular and molecular mechanisms. Genetic, environmental and endocrine factors contribute to the modulation of melanin's amount, type and distribution in the skin layers. One of the hallmarks of extrinsic skin aging induced by environmental stress factors is the alteration of the constitutive pigmentation pattern clinically defined as senile lentigines and/or melasma or other pigmentary dyschromias. The complexity of pollutants and tobacco smoke as environmental stress factors warrants a thorough understanding of the mechanisms by which they impact skin pigmentation through repeated and long-term exposure. Pre-clinical and clinical studies demonstrated that pollutants are known to induce reactive oxygen species (ROS) or inflammatory events that lead directly or indirectly to skin hyperpigmentation. Another mechanistic direction is provided by Aryl hydrocarbon Receptors (AhR) which were shown to mediate processes leading to skin hyperpigmentation in response to pollutants by regulation of melanogenic enzymes and transcription factors involved in melanin biosynthesis pathway. In this context, we will discuss a diverse range of New Approach Methodologies (NAMs) capable to provide mechanistic insights of the cellular and molecular pathways involved in the action of environmental stress factors on skin pigmentation and to support the design of raw ingredients and formulations intended to counter their impact and of any subsequently needed clinical studies.
Collapse
|