1
|
Schell SL, Sennett ML, Feehan RP, Wallace TE, Meiszberg EC, Longenecker AL, Helm MF, Kirby JS, Nelson AM. Pilot study of topical ruxolitinib demonstrates efficacy and blunting of heterogeneous inflammatory processes in mild hidradenitis suppurativa. Br J Dermatol 2025; 192:845-856. [PMID: 39708369 DOI: 10.1093/bjd/ljae495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/15/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Therapeutic options for mild hidradenitis suppurativa (HS) represent a significant gap in the current treatment landscape, with no U.S. Food and Drug Administration-approved therapies for early-stage HS. Topical Janus kinase (JAK) inhibitors (JAKi) are a compelling option due to the known upregulation of inflammatory JAK signalling in HS lesions and the recent success of systemic JAKi for the treatment of moderate-to-severe HS. OBJECTIVES To assess the clinical efficacy of ruxolitinib in a pilot cohort and to investigate the underlying biologic mechanisms associated with clinical response. METHODS This was a pilot single-site open-label prospective 24-week clinical trial of topical ruxolitinib (NCT04414514). Men and women with mild HS (Hurley stage I or II), with active inflammatory nodules, were recruited. All participants were observed for 8 weeks to monitor lesion counts (observational phase); active therapy (treatment phase) was then administered for 16 weeks. Topical ruxolitinib 1.5% cream was applied twice daily, covering the entirety of each HS-affected body site. Clinician- and patient-reported outcome measures were recorded throughout the study. Lesional skin punch biopsies were taken at the start and end of treatment for downstream mechanistic RNA sequencing and histological analyses. RESULTS Ten participants were enrolled in the study; four dropped out before the treatment phase of the trial. Six individuals with Hurley stage I (no tunnels) HS completed the study, five of whom successfully achieved Hidradenitis Suppurativa Clinical Response (HiSCR50) through 16 weeks of therapy. In this interim analysis, differential gene expression and gene set enrichment analyses revealed reduced activation of JAK-dependent interferon, interleukin (IL)-6, IL-2 and epidermal growth factor receptor signalling, and antimicrobial and keratinocyte responses. In contrast, signatures of wound healing and lipid metabolism were increased following JAKi treatment, indicating a return to homeostasis. Histological analyses revealed that clinically responsive patients had significantly reduced epidermal and dermal inflammation. Affected inflammatory infiltrate included neutrophils, T cells and plasma cells, with the predominantly affected cell types specific to the patient. CONCLUSIONS Collectively, the broad activity of topical ruxolitinib on inflammatory signalling processes resulted in promising efficacy, even with heterogeneity in baseline inflammation, in this pilot cohort. Importantly, topical treatment not only resolved epidermal inflammation, but also cleared deeper inflammatory infiltrate. The observed efficacy provides rationale to further investigate topical JAKi and other novel topical treatments in HS.
Collapse
Affiliation(s)
- Stephanie L Schell
- The Pennsylvania State University College of Medicine, Department of Dermatology, Hershey, PA, USA
| | - Mackenzie L Sennett
- The Pennsylvania State University College of Medicine, Department of Dermatology, Hershey, PA, USA
| | - Robert P Feehan
- The Pennsylvania State University College of Medicine, Department of Dermatology, Hershey, PA, USA
| | - Tierney E Wallace
- The Pennsylvania State University College of Medicine, Department of Dermatology, Hershey, PA, USA
| | - Elizabeth C Meiszberg
- The Pennsylvania State University College of Medicine, Department of Dermatology, Hershey, PA, USA
| | - Amy L Longenecker
- The Pennsylvania State University College of Medicine, Department of Dermatology, Hershey, PA, USA
| | - Matthew F Helm
- The Pennsylvania State University College of Medicine, Department of Dermatology, Hershey, PA, USA
| | - Joslyn S Kirby
- The Pennsylvania State University College of Medicine, Department of Dermatology, Hershey, PA, USA
| | - Amanda M Nelson
- The Pennsylvania State University College of Medicine, Department of Dermatology, Hershey, PA, USA
| |
Collapse
|
2
|
Kazmi A, Gill R, Restrepo P, Ji AL. The spatial and single-cell landscape of skin: Charting the multiscale regulation of skin immune function. Semin Immunol 2025; 78:101958. [PMID: 40267702 DOI: 10.1016/j.smim.2025.101958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
Immune regulation is a key function of the skin, a barrier tissue that exhibits spatial compartmentalization of innate and adaptive immune cells. Recent advances in single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) have facilitated systems-based investigations into the molecular and cellular features of skin immunity at single-cell resolution, identifying cell types that maintain homeostasis in a coordinated manner, and those that exhibit dysfunctional cell-cell interactions in disease. Here, we review how technological innovation is uncovering the multiple scales of heterogeneity in the immune landscape of the skin. The microanatomic scale encompasses the skin's diverse cellular components and multicellular spatial organization, which govern the functional cell interactions and behaviors necessary to protect the host. On the macroanatomic scale, understanding heterogeneity in cutaneous tissue architecture across anatomical sites promises to unearth additional functional immune variation and resulting disease consequences. We focus on how single-cell and spatial dissection of the immune system in experimental models and in humans has led to a deeper understanding of how each cell type in the skin contributes to overall immune function in a context-dependent manner. Finally, we highlight translational opportunities for adopting these technologies, and insights gleaned from them, into the clinic.
Collapse
Affiliation(s)
- Abiha Kazmi
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raman Gill
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paula Restrepo
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew L Ji
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
3
|
Mishra B, Gou Y, Tan Z, Wang Y, Hu G, Athar M, Mukhtar MS. Integrative systems biology framework discovers common gene regulatory signatures in mechanistically distinct inflammatory skin diseases. NPJ Syst Biol Appl 2025; 11:21. [PMID: 40016271 PMCID: PMC11868562 DOI: 10.1038/s41540-025-00498-x] [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: 11/14/2023] [Accepted: 02/04/2025] [Indexed: 03/01/2025] Open
Abstract
More than 20% of the population across the world is affected by non-communicable inflammatory skin diseases including psoriasis, atopic dermatitis, hidradenitis suppurativa, rosacea, etc. Many of these chronic diseases are painful and debilitating with limited effective therapeutic interventions. This study aims to identify common regulatory pathways and master regulators that regulate the molecular pathogenesis of inflammatory skin diseases. We designed an integrative systems biology framework to identify the significant regulators across several diseases. Network analytics unraveled 55 high-value proteins as significant regulators in molecular pathogenesis which can serve as putative drug targets for more effective treatments. We identified IKZF1 as a shared master regulator in hidradenitis suppurativa, atopic dermatitis, and rosacea with known disease-derived molecules for developing efficacious combinatorial treatments for these diseases. The proposed framework is very modular and indicates a significant path of molecular mechanism-based drug development from complex transcriptomics data and other multi-omics data.
Collapse
Affiliation(s)
- Bharat Mishra
- Department of Biology, The University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Yifei Gou
- Department of Genetics & Biochemistry, Clemson University, Clemson, SC, USA
| | - Zhengzhi Tan
- Department of Genetics & Biochemistry, Clemson University, Clemson, SC, USA
| | - Yiqing Wang
- Department of Genetics & Biochemistry, Clemson University, Clemson, SC, USA
| | - Getian Hu
- Department of Genetics & Biochemistry, Clemson University, Clemson, SC, USA
| | - Mohammad Athar
- Department of Dermatology, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - M Shahid Mukhtar
- Department of Genetics & Biochemistry, Clemson University, Clemson, SC, USA.
- Department of Biology, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
4
|
Moltrasio C, Moura R, Conti A, Fania L, Jaschke W, Caposiena Caro RD, Chersi K, Margiotta FM, Di Cesare A, Rosi E, Regensberger F, Boeckle B, Frischhut N, Cappellani S, Del Vecchio C, Nardacchione EM, Zalaudek I, von Stebut E, Berti I, Boniotto M, d'Adamo AP, Schmuth M, Dini V, Prignano F, Abeni D, Chiricozzi A, Marzano AV, Crovella S, Tricarico PM. Polygenic Score: A Tool for Evaluating the Genetic Background of Sporadic Hidradenitis Suppurativa. J Invest Dermatol 2024:S0022-202X(24)03042-2. [PMID: 39736307 DOI: 10.1016/j.jid.2024.11.019] [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: 06/25/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025]
Abstract
Sporadic hidradenitis suppurativa (spHS) is a multifactorial disease in which genetic predisposition is intertwined with environmental factors. Owing to the still-to-date limited knowledge of spHS genetics, we calculated polygenic scores (PGSs) to study the genetic underpinnings that contribute to spHS within European demographic. A total of 256 patients with spHS and 1686 healthy controls were analyzed across 6 European clinical centers. PGSs were calculated using a clumping and thresholding technique on 70% of the total sample, with the remaining 30% used for testing. The PANTHER tool was used to identify overrepresented genes. We generated a PGS characterized by 923 SNPs with a statistically significant association with spHS (P = 2 × 10-2). The statistically significant age-, sex-, and ancestry-adjusted association of our developed PGSs in spHS allows us to attribute a genetic contribution to the susceptibility of spHS (pseudo-R2 = 0.0053). Variants enriched for developing PGSs show a statistically significant preference for mapping to genes that encode primarily for cell adhesion proteins. Although this study developed a polygenic model associated with spHS, the low number of patients enrolled is a limitation. However, we believe that with larger experimental datasets, our model has the potential to serve as a valuable tool for predicting spHS states in future studies.
Collapse
Affiliation(s)
- Chiara Moltrasio
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ronald Moura
- Department of Advanced Diagnostics, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Andrea Conti
- SS Endocrinologia, auxologia e diabetologia, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Luca Fania
- Dermatology Unit, IDI-IRCSS, Rome, Italy
| | - Wolfram Jaschke
- Department of Dermatology, Venereology and Allergy, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Karin Chersi
- Dermatology Clinic, Hospital Clinics Giuliano Isontino (ASUGI), University of Trieste, Trieste, Italy
| | | | - Antonella Di Cesare
- Section of Dermatology, Department of Health Science, University of Florence, Florence, Italy
| | - Elia Rosi
- Section of Dermatology, Department of Health Science, University of Florence, Florence, Italy
| | - Florian Regensberger
- Department of Dermatology, Venereology and Allergy, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Boeckle
- Department of Dermatology, Venereology and Allergy, Medical University of Innsbruck, Innsbruck, Austria
| | - Nina Frischhut
- Department of Dermatology, Venereology and Allergy, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefania Cappellani
- Department of Advanced Diagnostics, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Cecilia Del Vecchio
- Department of Medical Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | | | - Iris Zalaudek
- Dermatology Clinic, Hospital Clinics Giuliano Isontino (ASUGI), University of Trieste, Trieste, Italy; Department of Medical Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | | | - Irene Berti
- Pediatric Department, Institute of Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Michele Boniotto
- INSERM, IMRB, Translational Neuropsychiatry, University Paris Est Créteil, Créteil, France
| | - Adamo Pio d'Adamo
- Department of Advanced Diagnostics, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy; Department of Medical Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Matthias Schmuth
- Department of Dermatology, Venereology and Allergy, Medical University of Innsbruck, Innsbruck, Austria
| | - Valentina Dini
- Department of Dermatology, University of Pisa, Pisa, Italy
| | - Francesca Prignano
- Section of Dermatology, Department of Health Science, University of Florence, Florence, Italy
| | - Damiano Abeni
- Clinical Epidemiology Unit, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Rome, Italy
| | - Andrea Chiricozzi
- Dermatologia, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica Del Sacro Cuore, Rome, Italy; Dermatologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Angelo Valerio Marzano
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Sergio Crovella
- Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar
| | - Paola Maura Tricarico
- Pediatric Department, Institute of Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy.
| |
Collapse
|
5
|
Pratap Kashyap M, Mishra B, Sinha R, Jin L, Gou Y, Kumar N, Goliwas KF, Haque S, Deshane J, Berglund E, Berglund D, Elewski BE, Elmets CA, Athar M, Mukhtar MS, Raman C. CD2 expressing innate lymphoid and T cells are critical effectors of immunopathogenesis in hidradenitis suppurativa. Proc Natl Acad Sci U S A 2024; 121:e2409274121. [PMID: 39560648 PMCID: PMC11621750 DOI: 10.1073/pnas.2409274121] [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: 05/14/2024] [Accepted: 09/26/2024] [Indexed: 11/20/2024] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic, debilitating inflammatory skin disease with a poorly understood immunopathogenesis. Here, we report that HS lesional skin is characterized by the expansion of innate lymphocytes and T cells expressing CD2, an essential activation receptor and adhesion molecule. Lymphocytes expressing elevated CD2 predominated with unique spatial distribution throughout the epidermis and hypodermis in the HS lesion. CD2+ cells were mainly innate lymphocytes expressing the NK cell marker, CD56, and CD4+ T cells. Importantly, these CD2+ cells interacted with CD58 (LFA3) expressing epidermal keratinocytes and fibroblasts in the hypodermis. Granzyme Abright NKT cells (CD2+CD3+CD56bright) clustered with α-SMA expressing fibroblasts juxtaposed to epithelialized tunnels and fibrotic regions of the hypodermis. Whereas NK cells (CD2+CD56dim) were perforin+, granzymes A+ and B+, and enriched adjacent to hyperplastic follicular epidermis and tunnels of HS showing presence of apoptotic cells. The cytokines IL-12, IL-15, and IL-18, which enhance NK cell maturation and function were significantly elevated in HS. Ex vivo HS skin explant cultures treated with CD2:CD58 interaction-blocking anti-CD2 monoclonal antibody attenuated secretion of inflammatory cytokines/chemokines and suppressed inflammatory gene signature. Additionally, CD2:CD58 blockade altered miRNAs involved in NK/NKT differentiation and/or function. In summary, we show that a cellular network of heterogenous NKT and NK cell populations drives inflammation and is critical in the pathobiology of HS, including tunnel formation and fibrosis. Finally, CD2 blockade is a viable immunotherapeutic approach for the effective management of HS.
Collapse
Affiliation(s)
- Mahendra Pratap Kashyap
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Bharat Mishra
- Department of Biology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Rajesh Sinha
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Lin Jin
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - YiFei Gou
- Department of Biology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Nilesh Kumar
- Department of Biology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Kayla F. Goliwas
- Department of Medicine (Division of Pulmonary, Allergy and Critical Care Medicine) at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Safiya Haque
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Jessy Deshane
- Department of Medicine (Division of Pulmonary, Allergy and Critical Care Medicine) at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Erik Berglund
- Department of Research and Development, ITB-MED AB, Stockholm113 68, Sweden
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, and Division of Transplantation Surgery, Karolinska Institute, Stockholm171 76, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm171 77, Sweden
| | - David Berglund
- Department of Research and Development, ITB-MED AB, Stockholm113 68, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala751 85, Sweden
| | - Boni E. Elewski
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Craig A. Elmets
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - Mohammad Athar
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| | - M. Shahid Mukhtar
- Department of Biology at the University of Alabama at Birmingham, Birmingham, AL35294
- Department of Genetics & Biochemistry at Clemson University, Clemson, SC29634
| | - Chander Raman
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, University of Alabama at Birmingham, Birmingham, AL35294
- Department of Dermatology at the University of Alabama at Birmingham, Birmingham, AL35294
| |
Collapse
|
6
|
Zhang Y, Jia W, Wang X, Mao Q, Luo L, Kong L, Guo Y, Mo R, Bu W, Li C. Inflammatory loop involving Staphylococcus aureus, IL-36γ, and cathepsin S drives immunity disorders in familial acne inversa keratinocytes. Heliyon 2024; 10:e31509. [PMID: 38947455 PMCID: PMC11214400 DOI: 10.1016/j.heliyon.2024.e31509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 07/02/2024] Open
Abstract
Acne inversa (AI) is an inflammatory skin disease associated with nicastrin (NCSTN) mutations. Despite the dysregulated bacterial-host immune interactions being an essential event in AI, the interaction between bacteria and keratinocytes in AI pathophysiology remains unclear. In this study, the NCSTN gene was suppressed using short hairpin RNA in HaCaT cells. Using RNA sequencing, real-time polymerase chain reaction, and western blotting, the expression of IL-36 cytokines was analyzed. The impact of Staphylococcus aureus on AI keratinocyte inflammation and underlying regulatory molecules was investigated by exposing the HaCaT cells to S. aureus. By stimulating NCSTN knockdown HaCaT cells with IFN-γ, the expression and regulatory mechanism of Cathepsin S (Cat S), an IL-36γ cleavage and activating protease, were investigated. After NCSTN knockdown, the IL-36α expression increased, and the IL-36Ra expression was downregulated. NCSTN/MEK/ERK impairment-induced Krüppel-like factor 4 (KLF4) up-regulation in concert with S. aureus-induced nuclear factor kappa B elevation acts synergistically to promote IL-36γ production with the subsequent IL-8 activation in HaCaT cells. NCSTN/MEK/ERK impairment was also observed in familial AI lesions. IFN-γ-induced Cat S in keratinocytes was enhanced after NCSTN knockdown. The expression of IFN-II pathway molecules was significantly upregulated in both NCSTN knockdown HaCaT cells and familial AI lesions. The Cat S expression was significantly elevated in the patient's AI lesions. Our findings suggested a synergistic relationship between S. aureus and NCSTN/MAPK/KLF4 axis in IL-36γ-induced familial AI keratinocytes.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, 210042, China
- Department of Dermatology and Venereology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Weixue Jia
- Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Xue Wang
- Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Qiuxia Mao
- Department of Dermatology, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu, 214400, China
| | - Lingling Luo
- Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Lingzhuo Kong
- Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Youming Guo
- Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Ran Mo
- Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Wenbo Bu
- Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| | - Chengrang Li
- Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, 210042, China
| |
Collapse
|
7
|
Nebo ID, Frew JW, Gudjonsson JE, Petukhova L. Tissue comparability and bias in hidradenitis suppurativa transcriptomic studies. Proc Natl Acad Sci U S A 2024; 121:e2404503121. [PMID: 38805291 PMCID: PMC11161763 DOI: 10.1073/pnas.2404503121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Affiliation(s)
- Ikenna David Nebo
- Columbia Vagelos College of Physicians and Surgeons, Columbia University, New York, NY10032
| | - John W. Frew
- Laboratory of Translational Cutaneous Medicine, Ingham Institute for Applied Medical Research, Sydney, NSW2170, Australia
- School of Clinical Medicine, University of New South Wales, Sydney, NSW2052, Australia
- Department of Dermatology, Liverpool Hospital, Sydney, NSW2170, Australia
- Holdsworth House Medical Practice, Sydney, NSW2010, Australia
| | | | - Lynn Petukhova
- The Ronald O. Perelman Department of Dermatology, New York University (NYU) Grossman School of Medicine, New York University (NYU) Langone Health, New York, NY10016
- Department of Population Health, New York University (NYU) Grossman School of Medicine, New York University (NYU) Langone Health, New York, NY10016
| |
Collapse
|
8
|
Jin L, Kashyap MP, Haque ST, Li C, Raman C, Athar M. Reply to Nebo et al.: Pathogenesis of advanced hidradenitis suppurativa: Are there differences based on anatomical location, gender, and race? Proc Natl Acad Sci U S A 2024; 121:e2405298121. [PMID: 38805283 PMCID: PMC11161802 DOI: 10.1073/pnas.2405298121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Affiliation(s)
- Lin Jin
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL35294-0019
| | - Mahendra P. Kashyap
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL35294-0019
| | - Safiya T. Haque
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL35294-0019
- Heersink School of Medicine, Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL35294
| | - Changzhao Li
- Division of Dermatopathology, Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL35294
| | - Chander Raman
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL35294-0019
| | - Mohammad Athar
- Center for Epigenomics and Translational Research in Inflammatory Skin Diseases, Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL35294-0019
| |
Collapse
|
9
|
Chhabra G, Ahmad N. Molecular chaos under the skin: Epigenetic reprogramming in hidradenitis suppurativa. Proc Natl Acad Sci U S A 2024; 121:e2319797121. [PMID: 38147564 PMCID: PMC10769847 DOI: 10.1073/pnas.2319797121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023] Open
Affiliation(s)
- Gagan Chhabra
- Department of Dermatology, University of Wisconsin, Madison, WI53705
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, WI53705
- William S. Middleton Veterans Affairs Medical Center, Madison, WI53705
| |
Collapse
|
10
|
Nardacchione EM, Tricarico PM, Moura R, d’Adamo AP, Thasneem A, Suleman M, Marzano AV, Crovella S, Moltrasio C. Unraveling the Epigenetic Tapestry: Decoding the Impact of Epigenetic Modifications in Hidradenitis Suppurativa Pathogenesis. Genes (Basel) 2023; 15:38. [PMID: 38254928 PMCID: PMC10815754 DOI: 10.3390/genes15010038] [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: 11/29/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic autoinflammatory skin disorder, which typically occurs during puberty or early adulthood. The pathogenesis of HS is complex and multifactorial; a close interaction between hormonal, genetic, epigenetics factors, host-specific aspects, and environmental influences contributes to the susceptibility, onset, severity, and clinical course of this disease, although the exact molecular mechanisms are still being explored. Epigenetics is currently emerging as an interesting field of investigation that could potentially shed light on the molecular intricacies underlying HS, but there is much still to uncover on the subject. The aim of this work is to provide an overview of the epigenetic landscape involved in HS. Specifically, in this in-depth review we provide a comprehensive overview of DNA methylation/hydroxymethylation, histone modifications, and non-coding RNAs (such as microRNA-miRNA-132, miRNA-200c, miRNA-30a-3p, miRNA-100-5b, miRNA-155-5p, miRNA-338-5p) dysregulation in HS patients. An interesting element of epigenetic regulation in HS is that the persistent inflammatory milieu observed in HS lesional skin could be exacerbated by an altered methylation profile and histone acetylation pattern associated with key inflammatory genes. Deepening our knowledge on the subject could enable the development of targeted epigenetic therapies to potentially restore normal gene expression patterns, and subsequentially ameliorate, or even reverse, the progression of the disease. By deciphering the epigenetic code governing HS, we strive to usher in a new era of personalized and effective interventions for this enigmatic dermatological condition.
Collapse
Affiliation(s)
- Elena Maria Nardacchione
- Department of Advanced Diagnostics, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (E.M.N.); (P.M.T.); (R.M.); (A.P.d.)
| | - Paola Maura Tricarico
- Department of Advanced Diagnostics, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (E.M.N.); (P.M.T.); (R.M.); (A.P.d.)
| | - Ronald Moura
- Department of Advanced Diagnostics, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (E.M.N.); (P.M.T.); (R.M.); (A.P.d.)
| | - Adamo Pio d’Adamo
- Department of Advanced Diagnostics, Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (E.M.N.); (P.M.T.); (R.M.); (A.P.d.)
- Department of Medical Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Ayshath Thasneem
- Laboratory of Animal Research Center (LARC), Qatar University, Doha 2713, Qatar; (A.T.); (M.S.); (S.C.)
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha 2713, Qatar
| | - Muhammad Suleman
- Laboratory of Animal Research Center (LARC), Qatar University, Doha 2713, Qatar; (A.T.); (M.S.); (S.C.)
| | - Angelo Valerio Marzano
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Sergio Crovella
- Laboratory of Animal Research Center (LARC), Qatar University, Doha 2713, Qatar; (A.T.); (M.S.); (S.C.)
| | - Chiara Moltrasio
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| |
Collapse
|