1
|
He Y, Huang M, Wang Y, Cai X, Xiao F. Inhibition of CTSC contributes to psoriasis inflammation and keratinocyte hyperproliferation by NF-κB signaling pathway. Int Immunopharmacol 2025; 157:114808. [PMID: 40339488 DOI: 10.1016/j.intimp.2025.114808] [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: 12/28/2024] [Revised: 04/07/2025] [Accepted: 05/03/2025] [Indexed: 05/10/2025]
Abstract
It was found that mutations in the Cathepsin C (CTSC) gene are responsible for Papillon-Lefevre syndrome which has a characteristic clinical feature of palmoplantar hyperkeratosis and psoriasiform lesions. However, its function in psoriasis is unclear so far. This study aims to investigate the roles and mechanisms of CTSC in psoriasis. The expression of CTSC was investigated by the analysis of single cell RNA sequencing (scRNA-seq) data and skin lesions of psoriasis patients. The role of CTSC in psoriasis was analyzed in human immortalized keratinocytes (HaCaT) stimulated with different inflammatory factors and mice of imiquimod (IMQ)-induced psoriasiform dermatitis. We showed that the expression of CTSC was significantly increased in the analysis of scRNA-seq data, which was identified in skin lesions of psoriasis patients and IMQ-induced psoriasis-like mice, and primary human keratinocytes and HaCaT cells stimulated with a cocktail of cytokines. In the presence of inflammatory factors or IMQ, CTSC inhibitor and knockdown of CTSC using siRNA exhibited significantly increased keratinocytes proliferation and the levels of proinflammatory cytokines. In vitro and in vivo experiments further showed that inhibited CTSC in psoriasis could activate the pathway of nuclear factor-κB (NF-κB). This study firstly outlines that inhibition of CTSC can contribute to inflammation and keratinocyte hyperproliferation by NF-κB pathway in psoriasis. It may provide a new perspective on understanding of the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Yue He
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China; Institute of Dermatology, Anhui Medical University, Hefei 230032, Anhui, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei 230032, Anhui, China
| | - Maoxin Huang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China; Institute of Dermatology, Anhui Medical University, Hefei 230032, Anhui, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei 230032, Anhui, China
| | - Yu Wang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China; Institute of Dermatology, Anhui Medical University, Hefei 230032, Anhui, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei 230032, Anhui, China
| | - Xinying Cai
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China; Institute of Dermatology, Anhui Medical University, Hefei 230032, Anhui, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei 230032, Anhui, China; Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei 230032, Anhui, China
| | - Fengli Xiao
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China; Institute of Dermatology, Anhui Medical University, Hefei 230032, Anhui, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei 230032, Anhui, China; Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei 230032, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, Anhui, China; The Center for Scientific Research of Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
2
|
Kim J, Lee J, Lee J, Kim K, Li X, Zhou W, Cao J, Krueger JG. Psoriasis harbors multiple pathogenic type 17 T-cell subsets: Selective modulation by risankizumab. J Allergy Clin Immunol 2025; 155:1898-1912. [PMID: 39978685 PMCID: PMC12145251 DOI: 10.1016/j.jaci.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Recent single-cell studies indicated that IL-17-producing T cells (T17) have diverse subsets expressing IL-17A, IL-17F, or a combination in human psoriasis skin. However, it is unknown how T17 subsets are differently regulated by IL-23 versus IL-17A blockade. OBJECTIVE We sought to investigate how systemic monoclonal antibody injections blocking IL-23 versus IL-17A differently modify immune cell transcriptomes in human psoriasis skin. METHODS We analyzed a total of 93 human skin single-cell libraries, including 42 psoriasis pretreatment lesional skin, 25 psoriasis pretreatment nonlesional skin, 12 psoriasis posttreatment after IL-23 inhibition, 4 psoriasis posttreatment after IL-17A inhibition, and 10 control skin samples. CLINICALTRIALS gov NCT04630652. RESULTS Of the six T17 subsets identified, an IL17A+IFNG+ subset and an IL17F+IL10- subset expressed the IL-23 receptor along with other inflammatory cytokines, and IL-23 inhibition downregulated these potentially pathogenic T17 subsets. In contrast, T17 cells expressing both IL-17A and IL-17F did not express the IL-23 receptor, and the percentage of this potentially nonpathogenic T17 subset increased after IL-23 inhibition. In addition, the expression of the IL-17-negative regulation genes, such as TNFAIP3, increased in myeloid cells more after IL-23 inhibition than after IL-17A inhibition. CONCLUSIONS This study suggests multiple immune mechanisms of how IL-23 inhibition can modify the complex inflammatory environment present in psoriatic skin, highlighting the roles of specific T17 subsets in psoriasis development and background skin protection.
Collapse
Affiliation(s)
- Jaehwan Kim
- Department of Dermatology, University of California, Davis, Sacramento, Calif; Dermatology Section, Veterans Affairs Northern California Health Care System, Mather, Calif; Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY.
| | - Jongmi Lee
- Department of Dermatology, University of California, Davis, Sacramento, Calif; Dermatology Section, Veterans Affairs Northern California Health Care System, Mather, Calif
| | - Jongeun Lee
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY
| | - Katherine Kim
- Department of Dermatology, University of California, Davis, Sacramento, Calif; Dermatology Section, Veterans Affairs Northern California Health Care System, Mather, Calif
| | - Xuan Li
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY
| | - Wei Zhou
- Laboratory of Single-cell Genomics and Population Dynamics, The Rockefeller University, New York, NY
| | - Junyue Cao
- Laboratory of Single-cell Genomics and Population Dynamics, The Rockefeller University, New York, NY
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY.
| |
Collapse
|
3
|
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 PMCID: PMC12146056 DOI: 10.1016/j.smim.2025.101958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
|
4
|
Chen X, Zheng Y, Man X, Li W. Tissue-resident memory T cells and their function in skin diseases. Chin Med J (Engl) 2025; 138:1175-1183. [PMID: 40066785 PMCID: PMC12091617 DOI: 10.1097/cm9.0000000000003499] [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] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Indexed: 05/21/2025] Open
Abstract
ABSTRACT Tissue-resident memory T (TRM) cells are a recently defined subtype of non-recirculating memory T cells with longevity and protective functions in peripheral tissues. As an essential frontline defense against infections, TRM cells have been reported to robustly patrol the tissue microenvironment in malignancies. Accumulating evidence also implicates that TRM cells in the relapse of chronic inflammatory skin diseases such as psoriasis and vitiligo. In light of these developments, this review aims to synthesize these recent findings to enhance our understanding of TRM cell characteristics and actions. Therefore, after providing a brief overview of the general features of the TRM cells, including precursors, homing, retention, and maintenance, we discuss recent insights gained into their heterogeneous functions in skin diseases. Specifically, we explore their involvement in conditions such as psoriasis, vitiligo, fixed drug eruption - dermatological manifestations of drug reactions at the same spot, cutaneous T cell lymphoma, and melanoma. By integrating these diverse perspectives, this review develops a comprehensive model of TRM cell behavior in various skin-related pathologies. In conclusion, our review emphasizes that deciphering the characteristics and mechanisms of TRM cell actions holds potential not only for discovering methods to slow cancer growth but also for reducing the frequency of recurrent chronic inflammation in skin tissue.
Collapse
Affiliation(s)
- Xibei Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Yuxin Zheng
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Xiaoyong Man
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Wei Li
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, China
| |
Collapse
|
5
|
Duan W, Xu K, Gao Y, Huang S, Xia X, Liu X, Pan S, Jiao C, Cheng W, Guo Y, Zhao J, Shen JW. Bimetallic Plasmonic Nanozyme-Based Microneedle for Synergistic Ferroptosis Therapy of Melanoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e04203. [PMID: 40387609 DOI: 10.1002/advs.202504203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 05/03/2025] [Indexed: 05/20/2025]
Abstract
Melanoma is the most common malignant skin tumor, characterized by complexity, invasiveness, and heterogeneity. Conventional therapies often yield poor outcomes, posing significant clinical challenges. Here, a microneedle (MN) patch that integrates nanozyme and traditional Chinese medicine (TCM) for ferroptosis pathway-dependent combined therapy of melanoma is designed. To amplify therapeutic activity, a novel Au@MoS2 bimetallic plasmonic nanozyme (BPNzyme) is prepared through a simple aqueous synthesis strategy involving a two-step process. Owing to the synergy between heterostructures, this rationally designed BPNzyme exhibits significantly enhanced therapeutic characteristics, including near-infrared (NIR) photothermal effect, peroxidase-like activity, and glutathione peroxidase-like property, which can effectively reshape the tumor microenvironment and disrupt the redox homeostasis. Under the combined action of the TCM β-elemene (β-ELE) and NIR light, further enhancement of oxidative damage, lipid peroxidation, and glutathione peroxidase 4 expression downregulation are observed for skin tumor cells, validating the synergistic amplification of ferroptosis. Moreover, the transdermal delivery of BPNzyme and β-ELE using the soluble hyaluronic acid MN patch effectively achieves 99.8% tumor growth suppression without significant systemic toxicity in vivo. These findings highlight the potential of the rationally designed BPNzyme-based MN system as a promising innovative strategy for non-invasive, efficient, and safe combination therapy of melanoma.
Collapse
Affiliation(s)
- Wei Duan
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, P. R. China
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, Zhejiang University, Hangzhou, 310027, P. R. China
- State Key Laboratory of Molecular Engineering of Polymers (Fudan University), Shanghai, 200438, P. R. China
| | - Keying Xu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| | - Yue Gao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| | - Sheng Huang
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| | - Xueqian Xia
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| | - Xiang Liu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| | - Shuangxue Pan
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| | - Chunpeng Jiao
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao, 266580, P. R. China
| | - Weijian Cheng
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| | - Yong Guo
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| | - Jingwen Zhao
- School of Chemistry and Chemical Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Jia-Wei Shen
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| |
Collapse
|
6
|
Zhou L, Leung TH. Detection by Single-Cell RNA Sequencing of Virally Mediated Skin Diseases. JID INNOVATIONS 2025; 5:100348. [PMID: 40026481 PMCID: PMC11869500 DOI: 10.1016/j.xjidi.2025.100348] [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: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 03/05/2025] Open
Abstract
Viruses are well-documented agents of specific skin diseases. However, their role and precise mechanism of action in other skin diseases remain unknown. We describe a single-cell RNA-sequencing-based strategy to interrogate human skin biopsies for viral transcripts, permitting detection of viral infection within a sample, single-cell resolution of virally infected cells and identification of subsequent transcriptomic perturbations. We validate our pipeline with 100% sensitivity and specificity by (i) detecting Merkel cell polyomavirus in Merkel cell carcinoma samples, (ii) detecting specific human papillomavirus strains in known human papillomavirus-positive tumors, and (iii) detecting rubella virus transcripts in patients with known rubella-associated granulomas. We identify infection of known and previously unreported cell types and elucidate viral-mediated transcriptional perturbations. In rubella virus-infected cells, we discover macrophage-specific evolution of the rubella virus E1 capsid protein. Finally, we interrogate skin biopsies from many established nonvirally mediated inflammatory skin diseases and do not find consistent evidence of viral infection in any condition. Combining single-cell RNA-sequencing data with virome detection strategies represents a potentially powerful approach to investigate and elucidate virus-mediated gene regulation in health and disease.
Collapse
Affiliation(s)
- Linda Zhou
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Thomas H. Leung
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1226-1282. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [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: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
8
|
Tao R, Chen X, Wang Y, Li S, Zhou S, Aghayants S, Yan L, Zhang Q, Zhu Z. Mendelian Randomization Combined with Single-Cell Transcriptome Analysis Reveals the Role of the Key Gene PCLAF in the Pathogenesis of Atopic Dermatitis. Clin Cosmet Investig Dermatol 2025; 18:867-882. [PMID: 40225314 PMCID: PMC11992997 DOI: 10.2147/ccid.s506139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/27/2025] [Indexed: 04/15/2025]
Abstract
Background Atopic dermatitis (AD) is a chronic inflammatory skin condition characterized by itching and rashes, influenced by genetic, environmental, and immune factors. Despite significant research, the molecular mechanisms underlying AD are not fully understood. This study aims to integrate single-cell RNA sequencing (scRNA-seq) with Mendelian Randomization (MR) to uncover genetic and metabolic pathways contributing to AD. Materials and Methods Data from scRNA-seq and bulk RNA sequencing datasets were analyzed to identify differentially expressed genes. The edgeR package was used for differential expression analysis, and candidate genes were explored using MR, employing eQTL data to determine causal relationships with AD. The inverse variance weighted method facilitated MR analysis, while gene set enrichment analysis (GSEA) was used to identify pathways associated with AD. Single-cell analysis was performed with the Seurat package to explore cellular heterogeneity, and pseudotime and cellular communication analyses were conducted to understand cell differentiation and interactions in AD. Results The study identified key genes-PCLAF, MICB, CHAD, and CA4-linked to AD, with PCLAF notably acting as a risk factor. These genes are involved in cell cycle regulation, immune evasion, cell adhesion, and metabolic processes. The MR analysis highlighted lipid, amino acid, and energy metabolism as critical pathways in AD. Single-cell analysis revealed increased cellular communication in AD, especially in Langerhans cells, keratinocytes, and T cells, signifying dysregulated immune responses and inflammatory pathways. Pseudotime analysis indicated abnormal differentiation trajectories in these cell types. Conclusion Our study highlights the importance of PCLAF in the pathogenesis of AD, indicating it as a potential target for future therapeutic strategies aimed at alleviating the disease by addressing genetic and metabolic disruptions.
Collapse
Affiliation(s)
- Rui Tao
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People’s Republic of China
| | - Xuejie Chen
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People’s Republic of China
| | - Yingying Wang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People’s Republic of China
| | - Sicheng Li
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People’s Republic of China
| | - Shengzhi Zhou
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People’s Republic of China
| | - Sis Aghayants
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People’s Republic of China
| | - Lingling Yan
- Department of Plastic and Cosmetic Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei Province, 430061, People’s Republic of China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People’s Republic of China
| | - Zhanyong Zhu
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People’s Republic of China
| |
Collapse
|
9
|
Taylor MA, Yuan M, Wang S, North JP, Cheng JB, Cho RJ. A case of biologic-resistant hand dermatitis demonstrates dual T2/T17 transcriptomic abnormalities and responds to Janus kinase inhibition. Br J Dermatol 2025; 192:743-745. [PMID: 39499646 DOI: 10.1093/bjd/ljae430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/10/2024] [Accepted: 11/01/2024] [Indexed: 11/07/2024]
Abstract
Many patients do not respond to targeted pathway-specific biologic therapies but predicting such failure remains an outstanding challenge. Here, in a case of adult-onset hand dermatitis that did not respond to multiple targeted biologic therapies, we detected gene expression programmes activated in both atopic dermatitis and psoriasis, via high-resolution genetic profiling. Broader, kinase-based immunosuppression elicited a near-complete clinical response. Therefore, our data suggest that such cases harbouring greater molecular complexity might be identified prospectively, circumventing some failures of single-pathway blockade.
Collapse
Affiliation(s)
- Mark A Taylor
- Department of Dermatology, University of California, San Francisco, CA, USA
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
| | - Michelle Yuan
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
| | - Sijia Wang
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
- Department of Dermatology, Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jeffrey P North
- Dermatopathology Service, University of California, San Francisco, CA, USA
| | - Jeffrey B Cheng
- Department of Dermatology, University of California, San Francisco, CA, USA
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA, USA
| | - Raymond J Cho
- Department of Dermatology, University of California, San Francisco, CA, USA
| |
Collapse
|
10
|
Wang Y, Hong Y. Investigating the complex roles of immunocyte phenotypes in the pathogenesis of dermatitis: a causal inference Mendelian randomization analysis. Arch Dermatol Res 2025; 317:593. [PMID: 40100330 DOI: 10.1007/s00403-025-04072-7] [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: 01/13/2025] [Revised: 02/07/2025] [Accepted: 02/25/2025] [Indexed: 03/20/2025]
Abstract
The etiology of dermatitis involves complex interactions between immune cells, genetics, and environmental factors. While immunocyte phenotypes have been linked to various forms of dermatitis, their causal role remains unclear. We conducted a two-sample Mendelian randomization (MR) analysis to investigate the causal effects between 731 immunocyte phenotypes and four types of dermatitis: atopic dermatitis, contact dermatitis, infective dermatitis, and seborrhoeic dermatitis. Genetic variants were used as instrumental variables, and the inverse variance-weighted (IVW) method was employed to assess causality. Sensitivity analyses were performed to ensure robustness. The forward MR analysis identified significant associations between 22 immunocyte phenotypes and atopic dermatitis, 8 phenotypes with contact dermatitis, 5 with infective dermatitis, and 6 with seborrhoeic dermatitis. The reverse MR analysis suggested potential bidirectional interactions of atopic dermatitis and CD3 on CD28 + CD45RA- CD8br (OR = 0.924, P = 0.012). This study revealed causal relationships between specific immunocyte phenotypes and dermatitis subtypes, providing novel insights into the immunopathogenesis of dermatitis and potential therapeutic targets.
Collapse
Affiliation(s)
- Yi Wang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Yanggang Hong
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China.
| |
Collapse
|
11
|
Thau H, Gerjol BP, Hahn K, von Gudenberg RW, Knoedler L, Stallcup K, Emmert MY, Buhl T, Wyles SP, Tchkonia T, Tullius SG, Iske J. Senescence as a molecular target in skin aging and disease. Ageing Res Rev 2025; 105:102686. [PMID: 39929368 DOI: 10.1016/j.arr.2025.102686] [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: 11/20/2024] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
Skin aging represents a multifactorial process influenced by both intrinsic and extrinsic factors, collectively known as the skin exposome. Cellular senescence, characterized by stable cell cycle arrest and secretion of pro-inflammatory molecules, has been implicated as a key driver of physiological and pathological skin aging. Increasing evidence points towards the role of senescence in a variety of dermatological diseases, where the accumulation of senescent cells in the epidermis and dermis exacerbates disease progression. Emerging therapeutic strategies such as senolytics and senomorphics offer promising avenues to target senescent cells and mitigate their deleterious effects, providing potential treatments for both skin aging and senescence-associated skin diseases. This review explores the molecular mechanisms of cellular senescence and its role in promoting age-related skin changes and pathologies, while compiling the observed effects of senotherapeutics in the skin and discussing the translational relevance.
Collapse
Affiliation(s)
- Henriette Thau
- Van Cleve Cardiac Regenerative Medicine Program Mayo Clinic, Rochester, Minesota, USA; Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bastian P Gerjol
- Department of Internal Medicine, Klinik Hirslanden, Zurich, Switzerland
| | - Katharina Hahn
- Department of Dermatology, Venereology and Allergology, Göttingen University Medical Center, Göttingen, Germany
| | - Rosalie Wolff von Gudenberg
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Leonard Knoedler
- Department of Oral and Maxillofacial Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health Berlin, Germany
| | - Kenneth Stallcup
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Maximilian Y Emmert
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, Göttingen University Medical Center, Göttingen, Germany
| | | | - Tamar Tchkonia
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jasper Iske
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Huang J, Hu Y, Wang S, Liu Y, Sun X, Wang X, Yu H. Single-cell RNA sequencing in autoimmune diseases: New insights and challenges. Pharmacol Ther 2025; 267:108807. [PMID: 39894174 DOI: 10.1016/j.pharmthera.2025.108807] [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: 01/02/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
Autoimmune diseases involve a variety of cell types, yet the intricacies of their individual roles within molecular mechanisms and therapeutic strategies remain poorly understood. Single-cell RNA sequencing (scRNA-seq) offers detailed insights into transcriptional diversity at the single-cell level, significantly advancing research in autoimmune diseases. This article explores how scRNA-seq enhances the understanding of cellular heterogeneity and its potential applications in the etiology, diagnosis, treatment, and prognosis of autoimmune diseases. By revealing a comprehensive cellular landscape, scRNA-seq illuminates the functional regulation of different cell subtypes during disease progression. It aids in identifying diagnostic and prognostic markers, and analyzing cell communication networks to uncover potential therapeutic targets. Despite its valuable contributions, addressing the limitations of scRNA-seq is essential for making further advancements.
Collapse
Affiliation(s)
- Jialing Huang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Yuelin Hu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Shuqing Wang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Yuefang Liu
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Xin Sun
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Xin Wang
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Guizhou, China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Guizhou, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China.
| |
Collapse
|
13
|
Liu SY, Lv J, Li F, Zhang YX. Immunological profile of lactate metabolism-related genes in Psoriasis a comprehensive analysis based on bulk and single-cell RNA sequencing data. Sci Rep 2025; 15:6872. [PMID: 40011693 PMCID: PMC11865271 DOI: 10.1038/s41598-025-91237-z] [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] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/19/2025] [Indexed: 02/28/2025] Open
Abstract
Psoriasis (Pso) is a chronic erythema squamous disease often accompanied by metabolic abnormalities. Disturbances in lactate metabolism have been observed in the skin of patients with Pso, but the role of lactate metabolism in the pathogenesis of Pso remains unclear. To identify core genes associated with lactate metabolism in Pso, we downloaded and merged two Pso-related datasets (GSE13355, GSE109248) from the GEO database. Through comprehensive analysis, we analyzed the functions associated with these hub genes and the correlation between their expression levels and immune infiltration. Additionally, we explored lactate metabolism-related hub genes in different immune cells using single-cell sequencing data. We identified four lactate metabolism-related hub genes that are highly associated with Pso. Further analysis revealed that these four hub genes were significantly correlated with the levels of immune cell infiltration. To further elucidate the impact of lactate metabolism on immune cells, we analyzed single-cell sequencing data from healthy controls and Pso patients. The expression of lactate metabolism-related hub genes was primarily observed in moDCs and T cells. These results suggest that lactate metabolism-related genes and their functions are closely associated with changes in inflammatory cells in Pso patients. This study provides new insights into the role of lactate metabolism in the pathogenesis of Pso and highlights potential therapeutic targets for the disease.
Collapse
Affiliation(s)
- Si-Yu Liu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Juan Lv
- Department of Intensive Care Medicine, Jiangmen Wuyi Hospital of Traditional Chinese Medicine, Jiangmen, China
| | - Fangqing Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi-Xuan Zhang
- Plastic Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
14
|
Ran Y, Peng X, Xia Y, Liu H, Liu Y. Periostin in Bullous Pemphigoid: A Potential Biomarker of Disease Activity and Severity. Exp Dermatol 2025; 34:e70067. [PMID: 39989295 DOI: 10.1111/exd.70067] [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: 09/10/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
Elevated periostin levels are commonly observed in conditions characterised by increased IgE and eosinophilia, such as bullous pemphigoid (BP), which typically presents with eosinophil infiltration and elevated IgE levels. To explore the link between periostin levels and key clinical parameters in BP, serum periostin levels were assessed in 55 classic BP patients and 55 healthy controls using ELISA. Upon admission, the BP Disease Area Index (BPDAI) score, autoantibody levels, and peripheral blood immune cells of BP patients were evaluated. The investigation also employed the Olink proteomic platform to analyse circulating proinflammatory biomarkers. The results indicated significantly higher periostin levels in BP patients, showing a strong positive correlation with BPDAI scores, which was more pronounced compared to the correlation between BPDAI scores and BP 180 IgG or eosinophil counts. Correlation analysis revealed positive links between periostin levels in serum and attributes such as urticaria/erythema lesions, total IgE levels, serum BP180 IgG, BP180 IgE, BP230 IgE, and blood eosinophil counts. The findings from the Olink proteomic analysis provided additional evidence of the connection between periostin and type II inflammation in BP, which was further validated by the observed positive correlation between periostin and IL-13 using ELISA. Furthermore, it was observed that serum periostin levels decreased post-effective treatment. Overall, this study underscores a compelling association between periostin expression and the activity and severity of BP, as well as its reflection of type II inflammation.
Collapse
Affiliation(s)
- Yutong Ran
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xueting Peng
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yumin Xia
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hong Liu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yale Liu
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
15
|
Huang D, Liu X, Gao X, Choi CK, Giglio G, Farah L, Leung T, Wong KC, Kan LL, Chong JW, Meng Q, Liao J, Cheung PF, Wong C. Meteorin-like protein/METRNL/Interleukin-41 ameliorates atopic dermatitis-like inflammation. Allergy 2025; 80:474-488. [PMID: 38727640 PMCID: PMC11804313 DOI: 10.1111/all.16150] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/30/2024] [Accepted: 04/08/2024] [Indexed: 02/08/2025]
Abstract
BACKGROUND Meteorin-like protein (METRNL)/Interleukin-41 (IL-41) is a novel immune-secreted cytokine/myokine involved in several inflammatory diseases. However, how METRNL exerts its regulatory properties on skin inflammation remains elusive. This study aims to elucidate the functionality and regulatory mechanism of METRNL in atopic dermatitis (AD). METHODS METRNL levels were determined in skin and serum samples from patients with AD and subsequently verified in the vitamin D3 analogue MC903-induced AD-like mice model. The cellular target of METRNL activity was identified by multiplex immunostaining, single-cell RNA-seq and RNA-seq. RESULTS METRNL was significantly upregulated in lesions and serum of patients with dermatitis compared to healthy controls (p <.05). Following repeated MC903 exposure, AD model mice displayed elevated levels of METRNL in both ears and serum. Administration of recombinant murine METRNL protein (rmMETRNL) ameliorated allergic skin inflammation and hallmarks of AD in mice, whereas blocking of METRNL signaling led to the opposite. METRNL enhanced β-Catenin activation, limited the expression of Th2-related molecules that attract the accumulation of Arginase-1 (Arg1)hi macrophages, dendritic cells, and activated mast cells. CONCLUSIONS METRNL can bind to KIT receptor and subsequently alleviate the allergic inflammation of AD by inhibiting the expansion of immune cells, and downregulating inflammatory gene expression by regulating the level of active WNT pathway molecule β-Catenin.
Collapse
Affiliation(s)
- Danqi Huang
- Department of Chemical PathologyThe Chinese University of Hong KongHong KongChina
| | - Xiuting Liu
- Department of Dermatology, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Xun Gao
- Department of Chemical PathologyThe Chinese University of Hong KongHong KongChina
- Center of Clinical Laboratory Medicine, Zhongda HospitalSoutheast UniversityNanjingChina
| | - Chun Kit Choi
- Department of Chemical PathologyThe Chinese University of Hong KongHong KongChina
| | - Giovanni Giglio
- Bridge Institute of Experimental Tumor Therapy, West German Cancer CenterUniversity Hospital EssenEssenGermany
- Division of Solid Tumor Translational OncologyGerman Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZHeidelbergGermany
| | - Luay Farah
- Bridge Institute of Experimental Tumor Therapy, West German Cancer CenterUniversity Hospital EssenEssenGermany
- Division of Solid Tumor Translational OncologyGerman Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZHeidelbergGermany
| | - Ting‐Fan Leung
- Department of PaediatricsThe Chinese University of Hong KongHong KongChina
| | - Katie Ching‐Yau Wong
- Department of Chemical PathologyThe Chinese University of Hong KongHong KongChina
| | - Lea Ling‐Yu Kan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal PlantsThe Chinese University of Hong KongHong KongChina
| | | | - Qing‐Jun Meng
- Welcome Centre for Cell Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- Centre for Biological Timing, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Jinyue Liao
- Department of Chemical PathologyThe Chinese University of Hong KongHong KongChina
| | - Phyllis Fung‐Yi Cheung
- Bridge Institute of Experimental Tumor Therapy, West German Cancer CenterUniversity Hospital EssenEssenGermany
- Division of Solid Tumor Translational OncologyGerman Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZHeidelbergGermany
- Spatiotemporal tumor heterogeneity, German Cancer Consortium (DKTK)A Partnership Between German Cancer Research Center (DKFZ) and University HospitalEssenGermany
| | - Chun‐Kwok Wong
- Department of Chemical PathologyThe Chinese University of Hong KongHong KongChina
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal PlantsThe Chinese University of Hong KongHong KongChina
| |
Collapse
|
16
|
Zhu Y, Zhao L, Yan W, Ma H, Zhao W, Qu J, Zheng W, Zhang C, Du H, Yu M, Wan N, Ye H, Xie Y, Ke B, Xu Q, Sun H, Sun Y, Ouyang Z. Celastrol directly targets LRP1 to inhibit fibroblast-macrophage crosstalk and ameliorates psoriasis progression. Acta Pharm Sin B 2025; 15:876-891. [PMID: 40177548 PMCID: PMC11959968 DOI: 10.1016/j.apsb.2024.12.041] [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: 07/29/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
Psoriasis is an incurable chronic inflammatory disease that requires new interventions. Here, we found that fibroblasts exacerbate psoriasis progression by promoting macrophage recruitment via CCL2 secretion by single-cell multi-omics analysis. The natural small molecule celastrol was screened to interfere with the secretion of CCL2 by fibroblasts and improve the psoriasis-like symptoms in both murine and cynomolgus monkey models. Mechanistically, celastrol directly bound to the low-density lipoprotein receptor-related protein 1 (LRP1) β-chain and abolished its binding to the transcription factor c-Jun in the nucleus, which in turn inhibited CCL2 production by skin fibroblasts, blocked fibroblast-macrophage crosstalk, and ameliorated psoriasis progression. Notably, fibroblast-specific LRP1 knockout mice exhibited a significant reduction in psoriasis like inflammation. Taken together, from clinical samples and combined with various mouse models, we revealed the pathogenesis of psoriasis from the perspective of fibroblast-macrophage crosstalk, and provided a foundation for LRP1 as a novel potential target for psoriasis treatment.
Collapse
Affiliation(s)
- Yuyu Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Lixin Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Wei Yan
- Department of Dermatology and Venereology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyue Ma
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wanjun Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Wei Zheng
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Chenyang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Haojie Du
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Meng Yu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ning Wan
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Ye
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yicheng Xie
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Bowen Ke
- Department of Anesthesiology, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Haiyan Sun
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Zijun Ouyang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China
| |
Collapse
|
17
|
Alkon N, Chennareddy S, Cohenour ER, Ruggiero JR, Stingl G, Bangert C, Rindler K, Bauer WM, Weninger W, Griss J, Jonak C, Brunner PM. Single-cell sequencing delineates T-cell clonality and pathogenesis of the parapsoriasis disease group. J Allergy Clin Immunol 2025; 155:461-478. [PMID: 39278361 DOI: 10.1016/j.jaci.2024.09.004] [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: 03/27/2024] [Revised: 07/30/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Mycosis fungoides (MF), the most common cutaneous T-cell lymphoma, is often underdiagnosed in early stages because of similarities with benign dermatoses such as atopic dermatitis (AD). Furthermore, the delineation from what is called "parapsoriasis en plaque", a disease that can appear either in a small- or large-plaque form, is still controversial. OBJECTIVE We sought to characterize the parapsoriasis disease spectrum. METHODS We performed single-cell RNA sequencing of skin biopsies from patients within the parapsoriasis-to-early-stage MF spectrum, stratified for small and large plaques, and compared them to AD, psoriasis, and healthy control skin. RESULTS Six of 8 large-plaque lesions harbored either an expanded alpha/beta or gamma/delta T-cell clone with downregulation of CD7 expression, consistent with a diagnosis of early-stage MF. In contrast, 6 of 7 small-plaque lesions were polyclonal in nature, thereby lacking a lymphomatous phenotype, and also revealed a less inflammatory microenvironment than early-stage MF or AD. Of note, polyclonal small- and large-plaque lesions characteristically harbored a population of NPY+ innate lymphoid cells and displayed a stromal signature of complement upregulation and antimicrobial hyperresponsiveness in fibroblasts and sweat gland cells, respectively. These conditions were clearly distinct from AD or psoriasis, which uniquely harbored CD3+CRTH2+ IL-13 expressing "TH2A" cells, or strong type 17 inflammation, respectively. CONCLUSION These data position polyclonal small- and large-plaque parapsoriasis lesions as a separate disease entity that characteristically harbors a so far undescribed innate lymphoid cell population. We thus propose a new term, "polyclonal parapsoriasis en plaque", for this kind of lesion because they can be clearly differentiated from early- and advanced-stage MF, psoriasis, and AD on several cellular and molecular levels.
Collapse
Affiliation(s)
- Natalia Alkon
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sumanth Chennareddy
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emry R Cohenour
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John R Ruggiero
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Georg Stingl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christine Bangert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Katharina Rindler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang M Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| | - Patrick M Brunner
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
18
|
Deng CC, Xu XY, Zhang Y, Liu LC, Wang X, Chen JY, Yao LY, Zhu DH, Yang B. Single-cell RNA-seq reveals immune cell heterogeneity and increased Th17 cells in human fibrotic skin diseases. Front Immunol 2025; 15:1522076. [PMID: 39872534 PMCID: PMC11769821 DOI: 10.3389/fimmu.2024.1522076] [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: 11/03/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
Background Fibrotic skin disease represents a major global healthcare burden, characterized by fibroblast hyperproliferation and excessive accumulation of extracellular matrix components. The immune cells are postulated to exert a pivotal role in the development of fibrotic skin disease. Single-cell RNA sequencing has been used to explore the composition and functionality of immune cells present in fibrotic skin diseases. However, these studies detected the gene expression of all cells in fibrotic skin diseases and did not enrich immune cells. Thus, the precise immune cell atlas in fibrotic skin diseases remains unknown. In this study, we plan to investigate the intricate cellular landscape of immune cells in keloid, a paradigm of fibrotic skin diseases. Methods CD45+ immune cells were enriched by fluorescence-activated cell sorting. Single-cell RNA sequencing was used to analyze the cellular landscape of immune cells in keloid and normal scar tissues. Ki-67 staining, a scratch experiment, real-time PCR, and Western blotting were used to explore the effect of the Th17 cell supernatant on keloid fibroblasts. Results Our findings revealed the intricate cellular landscape of immune cells in fibrotic skin diseases. We found that the percentage of Th17 cells was significantly increased in keloids compared to normal scars. All the subclusters of macrophages and dendritic cells (DCs) showed similar proportions between keloid samples and normal scar samples. However, upregulated genes in keloid M1 macrophages, M2 macrophages, and cDC2 are associated with the MHC class II protein complex assembly and antigen assembly, indicating that macrophages and cDC2 are active in keloids. Functional studies suggested that the supernatant of Th17 cells could promote proliferation, collagen expression, and migration of keloid fibroblasts through interleukin 17A. Importantly, increased Th17 cells are also found in other fibrotic skin diseases, such as hypertrophic scars and scleroderma, suggesting this represents a broad mechanism for skin fibrosis. Conclusion In summary, we built a single-cell atlas of fibrotic skin diseases in this study. In addition, we explored the function of Th17 cell-fibroblast interaction in skin fibrosis. These findings will help to understand fibrotic skin disease pathogenesis in depth and identify potential targets for fibrotic skin disease treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ding-Heng Zhu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Gong Z, Yi H, Zhang J, Li W, Wang H, Guo P, Li C, Pan A, Cao Y, Lu Z, Jiang H. Role of Arg1 + ILC2s and ILCregs in gestational diabetes progression. Sci Rep 2025; 15:1580. [PMID: 39794391 PMCID: PMC11724119 DOI: 10.1038/s41598-025-85452-x] [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: 10/04/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Innate lymphoid cells (ILCs) are a newly discovered subset of immune cells that are responsible for regulation of the immune microenvironment. In particular, the ILC categories ILC2s and regulatory ILCs (ILCregs) are associated with immunosuppression and chronic inflammation. Chronic low-grade inflammation leads to insulin resistance, a major etiological factor in gestational diabetes mellitus (GDM). However, the influence of ILCs on GDM remains poorly understood. Therefore, this study aims to investigate the potential role of ILCs in the development and progression of GDM. This study included 19 patients diagnosed with GDM and 19 age- and body mass index-matched individuals in the control group. Flow cytometry was employed to assess the frequency and function of ILC subsets in peripheral blood (PB), cord blood (CB), and placental tissues. Additionally, ELISA was utilized to measure the levels of the cytokines TNF-α, IFN-γ, TGF-β, and IL-4/10/13/22 in the serum samples of patients. Compared to the control group with normal pregnancy, significantly elevated levels of ILC2s, Arg1+ILC2s, and ILCregs were detected in the PB, CB, and placental tissues of the GDM group. With regard to inflammation-related cytokines, the levels of IL-13/22 in PB serum were significantly elevated, while the TGF-β levels were significantly reduced in the GDM group compared to the control group (CG). Further, in the CB serum samples, IL-13 levels were elevated in the GDM group. Additionally, a negative correlation was observed between the number of ILC3s and the number of ILCregs present in umbilical cord blood, while the IL-13 level in peripheral blood was negatively correlated with the number of ILC3s. The present findings indicate that chronic low-grade inflammation mediated by Arg-1+ILC2s and ILCregs is closely associated with the pathogenesis of GDM.
Collapse
Affiliation(s)
- Zhangyun Gong
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Haixing Yi
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jie Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Wan Li
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Hao Wang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Peipei Guo
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Caihua Li
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Anan Pan
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhimin Lu
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Huanhuan Jiang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Wanshui Road No. 120, Hefei, 230000, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
20
|
Sai Krishna AVS, Sinha S, Satyanarayana Rao MR, Donakonda S. The impact of PTEN status on glioblastoma multiforme: A glial cell type-specific study identifies unique prognostic markers. Comput Biol Med 2025; 184:109395. [PMID: 39531927 DOI: 10.1016/j.compbiomed.2024.109395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/11/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma multiforme (GBM) is the most invasive form of brain tumor, accounting for 5 % of the cases per 100,000 people in various countries. The phosphatase and tensin homolog deleted from chromosome 10 (PTEN) is a well-known tumor suppressor, and its alteration leads to a deleterious effect on GBM progression. The molecular mechanism of tumorigenesis in glial cell types, driven by PTEN status, is yet to be elucidated. In this study, we analyzed publicly available single-cell transcriptome profiles of PTEN wild-type (WT) and NULL GBM patients. We compared them with normal brain data to uncover many unique gene sets influenced by PTEN status. The co-expression network analysis of differentially expressed genes (DEGs) between normal brain and PTEN (WT and NULL) identified highly interconnected genes. The weighted gene co-expression network analysis (WGCNA), based on the DESeq2 algorithm, identified glial cell-type-specific modules in PTEN status-dependent bulk RNA expression profiles. We overlapped network module gene sets from single-cell and bulk transcriptome profiles, and shared genes were considered for further analysis. The hallmark pathway enrichment analysis of the genes unique to PTEN-WT and NULL revealed various tumor growth-related pathways across the glial cell types. Further characterization of PTEN-WT and PTEN-NULL networks belonging to the single-cell and bulk RNA datasets revealed that PTEN status influences the network modules in astrocytes, microglia, and oligodendrocyte precursor cells. An integrated influence value algorithm identified hub genes for each glial cell type. The prognostic analysis identified clinically relevant hub genes specific to the cell type in PTEN-WT: GLIPR2 (astrocytes), CFH, IL32, MXRA5 (microglia), and PTEN-NULL: ID1 (astrocytes) and LAT2 (microglia). Our glial cell type-level transcriptome analysis unearthed unique molecular pathways and prognostic markers in PTEN status-dependent GBM patients.
Collapse
Affiliation(s)
- A V S Sai Krishna
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Swati Sinha
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, MS Ramaiah University of Applied Sciences, Bengaluru, India
| | | | - Sainitin Donakonda
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany.
| |
Collapse
|
21
|
Mattessich M, Reyna J, Aron E, Ay F, Kilmer M, Kleinstein SH, Konstorum A. nipalsMCIA: flexible multi-block dimensionality reduction in R via nonlinear iterative partial least squares. Bioinformatics 2024; 41:btaf015. [PMID: 39799512 PMCID: PMC11783316 DOI: 10.1093/bioinformatics/btaf015] [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: 06/21/2024] [Revised: 12/16/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025] Open
Abstract
SUMMARY With the increased reliance on multi-omics data for bulk and single-cell analyses, the availability of robust approaches to perform unsupervised learning for clustering, visualization, and feature selection is imperative. We introduce nipalsMCIA, an implementation of multiple co-inertia analysis (MCIA) for joint dimensionality reduction that solves the objective function using an extension to Nonlinear Iterative Partial Least Squares. We applied nipalsMCIA to both bulk and single-cell datasets and observed significant speed-up over other implementations for data with a large sample size and/or feature dimension. AVAILABILITY AND IMPLEMENTATION nipalsMCIA is available as a Bioconductor package at https://bioconductor.org/packages/release/bioc/html/nipalsMCIA.html, and includes detailed documentation and application vignettes.
Collapse
Affiliation(s)
- Max Mattessich
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL 60208, USA
| | - Joaquin Reyna
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Bioinformatics and Systems Biology PhD Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Edel Aron
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06510, USA
| | - Ferhat Ay
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Bioinformatics and Systems Biology PhD Program, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Misha Kilmer
- Department of Mathematics, Tufts University, Medford, MA 02155, USA
| | - Steven H Kleinstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06510, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Anna Konstorum
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
- Center for Computing Sciences, Institute for Defense Analyses, Bowie, MD 20715, USA
| |
Collapse
|
22
|
Bazzini C, Bertschi NL, Steck O, Luther F, Schärli S, Rolfes ED, Vallone A, Begré N, Nuoffer JM, Radonijc-Hoesli S, Simon D, Schlapbach C. Human T Helper 9 Cells Rely on Peroxisome Proliferator-Activated Receptor-γ-Mediated Cystine Uptake to Prevent Lipid Peroxidation and Bioenergetic Failure. J Invest Dermatol 2024:S0022-202X(24)03027-6. [PMID: 39725162 DOI: 10.1016/j.jid.2024.10.616] [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: 10/27/2023] [Revised: 10/10/2024] [Accepted: 10/31/2024] [Indexed: 12/28/2024]
Abstract
T helper 9 (Th9) cells are implicated in allergic skin inflammation and depend on the transcription factor peroxisome proliferator-activated receptor-γ (PPAR-γ) for full effector function. In this study, we uncovered a role for PPAR-γ in the amino acid metabolism of human Th9 cells. In in vitro-primed Th9 cells, PPAR-γ expression positively correlated with the expression of SLC7A8, which encodes LAT2, a transporter of large neutral amino acids, including cystine. Inhibition of PPAR-γ led to a compensatory upregulation of SLC7A11, a subunit of the cystine-glutamine antiporter xCT, indicating a specific need for cystine uptake in Th9 cells. Indeed, Th9 cells were sensitive to cystine deprivation, which triggered lipid peroxidation and bioenergetic failure, resulting in a ferroptosis-like form of cell death. This outcome was further enforced by additional PPAR-γ inhibition. Finally, combined SLC7A11 and PPAR-γ inhibition depleted Th9 cells in ex vivo samples of acute allergic skin inflammation. Overall, our data suggest that human Th9 cells are dependent on uptake of exogenous cystine, which opens up promising therapeutic strategies for their inhibition or depletion in allergic skin inflammation.
Collapse
Affiliation(s)
- Cecilia Bazzini
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nicole L Bertschi
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Oliver Steck
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Fabian Luther
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Stefanie Schärli
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Eva D Rolfes
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Angela Vallone
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nadja Begré
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jean-Marc Nuoffer
- Institute of Clinical Chemistry, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Susanne Radonijc-Hoesli
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Christoph Schlapbach
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
23
|
Seremet T, Di Domizio J, Girardin A, Yatim A, Jenelten R, Messina F, Saidoune F, Schlapbach C, Bogiatzi S, Minisini F, Garzorz-Stark N, Leuenberger M, Wüthrich H, Vernez M, Hohl D, Eyerich S, Eyerich K, Guenova E, Paul C, Gottardo R, Conrad C, Gilliet M. Immune modules to guide diagnosis and personalized treatment of inflammatory skin diseases. Nat Commun 2024; 15:10688. [PMID: 39695162 PMCID: PMC11655867 DOI: 10.1038/s41467-024-54559-6] [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: 04/23/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024] Open
Abstract
Previous advances have identified immune pathways associated with inflammatory skin diseases, leading to the development of targeted therapies. However, there is a lack of molecular approaches that delineate these pathways at the individual patient level for personalized diagnostic and therapeutic guidance. Here, we conduct a cross-comparison of expression profiles from multiple inflammatory skin diseases to identify gene modules defining relevant immune pathways. Seven modules are identified, representing key immune pathways: Th17, Th2, Th1, Type I IFNs, neutrophilic, macrophagic, and eosinophilic. These modules allow the development of a molecular map with high diagnostic efficacy for inflammatory skin diseases and clinico-pathologically undetermined cases. Aligning dominant modules with treatment targets offers a rational framework for treatment selection, improving response rates in both treatment-naïve patients and non-responders to targeted therapies. Overall, our approach offers precision medicine for inflammatory skin diseases, utilizing transcriptional modules to support diagnosis and guide personalized treatment selection.
Collapse
Affiliation(s)
- Teofila Seremet
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Jeremy Di Domizio
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Antoine Girardin
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Ahmad Yatim
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Raphael Jenelten
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Francesco Messina
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Fanny Saidoune
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Christoph Schlapbach
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sofia Bogiatzi
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Frederic Minisini
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Natalie Garzorz-Stark
- Department of Dermatology and Venereology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Matthieu Leuenberger
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Héloise Wüthrich
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Maxime Vernez
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Daniel Hohl
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Stefanie Eyerich
- Department of Dermatology and Venereology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Kilian Eyerich
- Department of Dermatology and Venereology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Emmanuella Guenova
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Carle Paul
- Department of Dermatology and Venereology, CHU Toulouse, Toulouse, France
| | - Raphael Gottardo
- Biomedical Data Science Center, CHUV, UNIL, and SIB, Lausanne, Switzerland
| | - Curdin Conrad
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland
| | - Michel Gilliet
- Department of Dermatology, Lausanne University Hospital CHUV and University of Lausanne, 1011, Lausanne, Switzerland.
| |
Collapse
|
24
|
Zhou B, Wu T, Li H, Yang J, Ma Z, Ling Y, Ma H, Huang C. Identification of CD19 as a shared biomarker via PPARγ/β-catenin/Wnt3a pathway linking psoriasis and major depressive disorder. J Affect Disord 2024; 367:75-87. [PMID: 39197550 DOI: 10.1016/j.jad.2024.08.159] [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: 04/22/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Psoriasis, a chronic inflammatory skin disorder, is frequently linked with metabolic, cardiovascular, and psychological comorbidities. Recent research has highlighted the correlation between psoriasis and major depressive disorder (MDD); however, the underlying mechanism remains unclear. METHODS Commonly differentially expressed genes (DEGs) in psoriasis and MDD were identified and visualized using data from the GEO database. Subsequently, functional enrichment analysis was conducted using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Genemania. The hub gene was selected through LASSO and Random Forest algorithms, validated in clinical tissues using Student's t-test and Receiver Operating Characteristic curve. To investigate the hub gene's function in disease phenotype, we established imiquimod (IMQ)-induced psoriasiform dermatitis and chronic unpredictable mild stress (CUMS) mouse models. Lentiviral shRNA interference was topically applied in mice, and downstream pathways were validated at the mRNA and protein levels. RESULTS A total of 395 overlapping DEGs were identified from GSE121212 and GSE54568 datasets, and twenty core genes were extracted. Functional enrichment analysis revealed that the core genes were significantly associated with the Wnt signaling pathway, neurodegeneration, and energy metabolism. CD19 was identified as the hub gene through algorithms, and external validation showed remarkable AUC values of 0.69 and 0.74, respectively. The level of CD19 increased significantly in IMQ-treated and CUMS-treated mice. Suppression of CD19 significantly alleviated the phenotypes of IMQ-induced psoriasiform dermatitis and CUMS-induced depressive-like behaviors by regulating the PPARγ/β-catenin/Wnt3a pathway. CONCLUSION CD19 may serve as a common biomarker or therapeutic target of psoriasis and MDD via PPARγ/β-catenin/Wnt3a pathway.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ting Wu
- Department of Dermatology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Haitao Li
- China Three Gorges University and Yichang Central People' Hospital, Yichang 443000, China
| | - Jiahao Yang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 4030030, China
| | - Zhujun Ma
- China Three Gorges University and Yichang Central People' Hospital, Yichang 443000, China
| | - Yunli Ling
- Beijing Huairou Hospital, Capital Medical University, Beijing 101400, China.
| | - Hanying Ma
- School of Life Sciences, Huanggang Normal University, Huanggang 438000, China.
| | - Changzheng Huang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
25
|
Anitua E, Tierno R, Martínez de Lagrán Z, Alkhraisat MH. Impact of inflammatory skin conditions on the biological profile of plasma rich in growth factor. Tissue Cell 2024; 91:102560. [PMID: 39299031 DOI: 10.1016/j.tice.2024.102560] [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: 07/01/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Plasma rich in growth factors (PRGF) can be used over patients suffering from dermatoses due to its anti-inflammatory effect. However, this population group might present soluble autoimmune components and there is limited information about the effect of chronic skin inflammation on PRGF bioactive properties. With the aim of characterizing PRGF composition, PRGF from healthy (H) donors and patients with atopic dermatitis (AD), psoriasis (PS), or lichen sclerosus (LS) was obtained. In order to reduce the inflammatory component, leukocyte exclusion and heat-inactivation (Immunosafe) were tested. Haematological-serological parameters, platelet functionality, clot microstructure, protein content and bioactivity were determined. Mean values and 95 % confidence intervals (mean[95 % CI]) were computed for key haematological parameters, such as platelet (410×103/mm3[371-449]) and leukocyte content (205×103/mm3[148-262]), platelet activation (resting: 4.3 %[3.1-5.5] and activated: 97.4 %[96.7-98.0]), the concentration of plasma proteins and morphogens, including immunoglobulins A (210.7 mg/dL[191.8-229.6]), G (933.1 mg/dL[887.2-978.9]), E (783.5 mg/dL[54.4-1512.6]), and M (115.0 mg/dL[97.1-133.0]), Complement Protein (31.6 mg/mL[26.6-36.6]), C-Reactive protein (3.1 mg/L[2.0-4.1]), TGF-β1 (35975.6 pg/mL[34221.3-37729.8]), fibronectin (146410.0 ng/mL[136518.3-156301.7]), PDGF-AB (13308.5 pg/mL[12401.0-14216.0]), CD40L (2389.3 pg/mL[1887.7-2890.8]), IL-4 (0.12 pg/mL[0.07-0.18]), IL-13 (35.4 pg/mL[21.0-49.7]), IL-1β (0.09 pg/mL[0.06-0.11]) and TNF-α (0.31 pg/mL[0.24-0.38]), and also for cell proliferation (332.9ngDNA/mL[317.4-348.3]), viability (135.6 %[132.0-139.2]) and migration (103.8cells/mm2[98.3-109.3]). Plasma from AD donors presented increased Immunoglobulin E (IgE) that was significantly reduced after Immunosafe along with the complement system and autoantibodies. Platelet functionality was altered for AD, but no microstructure differences were identified. Pathological groups presented reduced concentration of fibronectin (AD/LS) and Platelet-Derived Growth Factor (PDGF-AB) (P). Immunosafe treatment reduced Cluster of Differentiation 40 Protein (CD40L), interleukin 1β (IL-1β), and Tumor Necrosis Factor α (TNF-α) concentrations. Fibroblasts supplemented with PRGF obtained from pathological patients (PS/AD) showed reduced viability but Immunosafe increased cell proliferation and migration in SP (LS) and L-SP samples (PS/AD). In conclusion, PRGF derived from pathological patients present autoimmune components, but heat-inactivation or leukocyte exclusion could minimize local side effects.
Collapse
Affiliation(s)
- Eduardo Anitua
- University Institute for Regenerative Medicine and Oral Implantology (UIRMI), Vitoria, Spain; BTI Biotechnology Institute, Vitoria, Spain.
| | - Roberto Tierno
- University Institute for Regenerative Medicine and Oral Implantology (UIRMI), Vitoria, Spain; BTI Biotechnology Institute, Vitoria, Spain
| | | | - Mohammad H Alkhraisat
- University Institute for Regenerative Medicine and Oral Implantology (UIRMI), Vitoria, Spain; BTI Biotechnology Institute, Vitoria, Spain
| |
Collapse
|
26
|
Saito H, Tamari M, Motomura K, Ikutani M, Nakae S, Matsumoto K, Morita H. Omics in allergy and asthma. J Allergy Clin Immunol 2024; 154:1378-1390. [PMID: 39384073 DOI: 10.1016/j.jaci.2024.09.023] [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: 09/09/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024]
Abstract
This review explores the transformative impact of omics technologies on allergy and asthma research in recent years, focusing on advancements in high-throughput technologies related to genomics and transcriptomics. In particular, the rapid spread of single-cell RNA sequencing has markedly advanced our understanding of the molecular pathology of allergic diseases. Furthermore, high-throughput genome sequencing has accelerated the discovery of monogenic disorders that were previously overlooked as ordinary intractable allergic diseases. We also introduce microbiomics, proteomics, lipidomics, and metabolomics, which are quickly growing areas of research interest, although many of their current findings remain inconclusive as solid evidence. By integrating these omics data, we will gain deeper insights into disease mechanisms, leading to the development of precision medicine approaches that promise to enhance treatment outcomes.
Collapse
Affiliation(s)
- Hirohisa Saito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Masato Tamari
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kenichiro Motomura
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Masashi Ikutani
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Susumu Nakae
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan; Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Allergy Center, National Center for Child Health and Development, Tokyo, Japan.
| |
Collapse
|
27
|
Ma Y, Liu Y, Zhong Y, Li X, Xu Y, Chen L, Gong L, Huang H, Chen X, He Y, Qiang L. Oroxylin A attenuates psoriasiform skin inflammation by direct targeting p62 (sequestosome 1) via suppressing M1 macrophage polarization. Br J Pharmacol 2024; 181:5110-5132. [PMID: 39313956 DOI: 10.1111/bph.17349] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND AND PURPOSE Psoriasis results from the interplay of innate and adaptive immunity in the skin. Oroxylin A (OA) has shown anti-inflammatory effects in various disorders. This study explores oroxylin A potential in treating psoriasis, particularly its impact on type I macrophage (Mφ1) polarization. EXPERIMENTAL APPROACH Oroxylin A-mediated therapeutic effects were evaluated using imiquimod-induced or IL-23-injected psoriatic mice models, followed by proteomics assays to predict potential signalling and targeting proteins. Immunofluorescence and immunoblot assays verified that oroxylin A suppresses NF-κB signalling in Mφ1 macrophages. Co-immunoprecipitation and microscale thermophoresis (MST) assays further demonstrated that p62 (sequestosome 1) is the target protein for oroxylin A in macrophages. Oroxylin A-p62-mediated suppression of psoriasis was validated in an imiquimod-induced p62 conditional knockout (cKO) mice model. KEY RESULTS Oroxylin A demonstrated therapeutic efficacy in murine models induced by imiquimod or IL-23 by attenuating cutaneous inflammation and mitigating Mφ1 polarization via NF-κB signalling. Proteomics analysis suggested SQSTM1/p62 as a key target, confirmed to interact directly with oroxylin A. Oroxylin A disrupted the p62-PKCζ interaction by binding to PB1 domain of p62. Its anti-inflammatory effects were significantly reduced in macrophages from p62 cKO mice compared to the wild-type (WT) mice in psoriasis model, supporting oroxylin A role in suppressing Mφ1 polarization through its interaction with p62. CONCLUSION AND IMPLICATIONS Our findings demonstrated oroxylin A suppressed psoriasiform skin inflammation in mouse models by blocking the PKCζ-p62 interaction, subsequently inhibiting the activation of NF-κB p65 phosphorylation in macrophages.
Collapse
Affiliation(s)
- Yuxiang Ma
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Department of Pharmacology, Guilin Medical University, Guilin, China
| | - Yunyao Liu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - You Zhong
- Zhuhai United Laboratories Co., Ltd., Zhuhai, Guangdong, China
| | - Xiangzheng Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yujiao Xu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Leyi Chen
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Litong Gong
- Jiangsu Chia Tai-Tianqing Pharmaceutical Co., Ltd., Nanjing, China
| | - He Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xu Chen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Yuan He
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lei Qiang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| |
Collapse
|
28
|
Zhou X, Ning J, Cai R, Liu J, Yang H, Liu Q, Lv J, Bai Y. Multi-omic analysis revealed the immunological patterns and diagnostic value of exhausted T cell-derived PTTG1 in patients with psoriasis. Biochem Biophys Res Commun 2024; 734:150740. [PMID: 39342798 DOI: 10.1016/j.bbrc.2024.150740] [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: 07/15/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Psoriasis, characterized by chronic inflammation, is a persistent skin condition that is notoriously challenging to manage and prone to relapse. Despite significant advancements in its treatment, many adverse reactions still occur. Therefore, exploring the mechanisms behind the occurrence and development of psoriasis is extremely important. METHODS The weighted correlation network analysis (WGCNA) algorithm was used to identify phenotype-related genes in patients with psoriasis. We recruited clinical samples of patients with psoriasis, and used single-cell RNA sequencing (scRNA-seq) to visualize divergent genes and metabolisms of varied cells for the psoriasis. Various machine-learning methods were used to identify core genes, and molecular docking was used to analyze the stability of leptomycin B targeting pituitary tumor transforming 1 (PTTG1). Immunofluorescence (IHC) analysis, multiplex immunofluorescence (mIF) analysis, and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to validate the results. RESULTS Our results identified 1391 genes associated with the phenotype in patients with psoriasis and highlighted the significant alterations in T-cell functionality observed in the disease by WGCNA. There were nine distinct cellular clusters in psoriasis analyzed with the aid of scRNA-seq data. Each subtype of cell exhibited distinct genetic profiles, functional roles, signaling mechanisms, and metabolic characteristics. Machine-learning methods further demonstrated the potential diagnostic value of T cell-derived PTTG1 and its relationship with T-cell exhaustion in psoriasis. Lastly, the leptomycin B was scrutinized and verified had high stability targeting PTTG1. CONCLUSIONS This study elucidates the biological basis of psoriasis. At the same time, it was discovered that PTTG1 derived from exhausted T cells serves as a diagnostic biomarker for psoriasis. Leptomycin B could be a potential drug for targeted treatment of psoriasis on PTTG1.
Collapse
Affiliation(s)
- Xiangnan Zhou
- Department of Dermatology, China-Japan Friendship Hospital, National Center for Integrative Medicine, Beijing, 100029, China
| | - Jingyuan Ning
- State Key Laboratory of Medical Molecular Biology & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Rui Cai
- Beijing University of Chinese Medicine, China-Japan Friendship Clinical School of Medicine, Beijing, 100029, China
| | - Jiayi Liu
- Beijing University of Chinese Medicine, China-Japan Friendship Clinical School of Medicine, Beijing, 100029, China
| | - Haoyu Yang
- Department of Dermatology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Qingwu Liu
- Department of Dermatology, China-Japan Friendship Hospital, National Center for Integrative Medicine, Beijing, 100029, China
| | - Jingjing Lv
- Department of Dermatology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Yanping Bai
- Department of Dermatology, China-Japan Friendship Hospital, National Center for Integrative Medicine, Beijing, 100029, China.
| |
Collapse
|
29
|
McGrath JA, Hsu CK. Inflammatory dermatoses and an era of new diagnostic dermatopathology. Br J Dermatol 2024; 191:855-856. [PMID: 39066646 DOI: 10.1093/bjd/ljae306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024]
Affiliation(s)
- John A McGrath
- St John's Institute of Dermatology, King's College London, London, UK
| | - Chao-Kai Hsu
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
30
|
Nishide M, Shimagami H, Kumanogoh A. Single-cell analysis in rheumatic and allergic diseases: insights for clinical practice. Nat Rev Immunol 2024; 24:781-797. [PMID: 38914790 DOI: 10.1038/s41577-024-01043-3] [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: 05/08/2024] [Indexed: 06/26/2024]
Abstract
Since the advent of single-cell RNA sequencing (scRNA-seq) methodology, single-cell analysis has become a powerful tool for exploration of cellular networks and dysregulated immune responses in disease pathogenesis. Advanced bioinformatics tools have enabled the combined analysis of scRNA-seq data and information on various cell properties, such as cell surface molecular profiles, chromatin accessibility and spatial information, leading to a deeper understanding of pathology. This Review provides an overview of the achievements in single-cell analysis applied to clinical samples of rheumatic and allergic diseases, including rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis, allergic airway diseases and atopic dermatitis, with an expanded scope beyond peripheral blood cells to include local diseased tissues. Despite the valuable insights that single-cell analysis has provided into disease pathogenesis, challenges remain in translating single-cell findings into clinical practice and developing personalized treatment strategies. Beyond understanding the atlas of cellular diversity, we discuss the application of data obtained in each study to clinical practice, with a focus on identifying biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Masayuki Nishide
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan.
- Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Hiroshi Shimagami
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
- Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan.
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka, Japan.
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
31
|
Jairath V, Acosta Felquer ML, Cho RJ. IL-23 inhibition for chronic inflammatory disease. Lancet 2024; 404:1679-1692. [PMID: 39461795 DOI: 10.1016/s0140-6736(24)01750-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 10/29/2024]
Abstract
Biological monoclonal antibody drugs inhibit overactive cytokine signalling that drives chronic inflammatory disease in different organ systems. In the last 10 years, interleukin (IL)-23 inhibitors have attained an important position in the treatment of psoriatic skin and joint disease as well as inflammatory bowel diseases. Addressing an upstream pathological mechanism shared between these disorders, this drug class has high efficacy rates and a durable response that extends dosing intervals up to 3 months. Pooled clinical trial data show objective disease improvement for more than 70% of patients with psoriasis and up to 50% of patients with inflammatory bowel disease. The first antibody inhibitor for IL-23A targeted a p40 subunit shared with IL-12. Subsequently, even greater improvement was established for inhibitors of the p19 protein unique to IL-23A. IL-23 p19 inhibitors elicit clinical response in both bio-naive and bio-exposed patients and show superiority to tumour necrosis factor α inhibitors in plaque psoriasis. Reported differences in efficacy between p19 inhibitors suggest that individual drug action might be modulated by antibody affinity. Although long-term safety data are accumulating, rates of serious adverse events and infections for interleukin (IL)-23 inhibitors are similar to the rates for placebo across approved indications.
Collapse
Affiliation(s)
- Vipul Jairath
- Departments of Medicine, Division of Gastroenterology, Western University, Ontario, ON, Canada
| | - Maria Laura Acosta Felquer
- Rheumatology Unit, Internal Medicine Service, Hospital Italiano de Buenos Aires and Instituto Universitario Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Raymond Jaihyun Cho
- Department of Dermatology, University of California, San Francisco, CA, USA.
| |
Collapse
|
32
|
Silverberg JI, Rosmarin D, Chovatiya R, Bieber T, Schleicher S, Beck L, Gooderham M, Chaudhry S, Fanton C, Yu D, Levy J, Liu Y, Miyazaki T, Tagliaferri M, Schmitz C, Nirula A, Kotzin B, Zalevsky J. The regulatory T cell-selective interleukin-2 receptor agonist rezpegaldesleukin in the treatment of inflammatory skin diseases: two randomized, double-blind, placebo-controlled phase 1b trials. Nat Commun 2024; 15:9230. [PMID: 39455575 PMCID: PMC11511931 DOI: 10.1038/s41467-024-53384-1] [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: 05/16/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Regulatory T cell (Treg) impairment is implicated in the pathogenesis of chronic inflammatory diseases, but relatively little is known about the therapeutic potential of Treg restoration. Here we present clinical evidence for the Treg-selective interleukin-2 receptor agonist rezpegaldesleukin (REZPEG) in two randomized, double-blind, placebo-controlled Phase 1b trials in patients with moderate-to-severe atopic dermatitis (AD) (NCT04081350) or chronic plaque psoriasis (PsO) (NCT04119557). Key inclusion criteria for AD included an Eczema Area and Severity Index (EASI) score ≥ 16 and a validated Investigator Global Assessment for Atopic Dermatitis (vIGA-AD) ≥ 3, and for PsO included a Psoriasis Area and Severity Index (PASI) score of ≥ 12 and a static Physician's Global Assessment (sPGA) score of ≥ 3. REZPEG is safe and well-tolerated and demonstrates consistent pharmacokinetics in participants receiving subcutaneous doses of 10 to 12 µg/kg or 24 µg/kg once every 2 weeks for 12 weeks, meeting the primary and secondary objectives, respectively. AD patients receiving the higher dose demonstrate an 83% improvement in EASI score after 12 weeks of treatment. EASI improvement of ≥ 75% (EASI-75) and vIGA-AD responses are maintained for 36 weeks after treatment discontinuation in 71% and 80% of week 12 responders, respectively. These exploratory clinical improvements are accompanied by sustained increases in CD25bright Tregs. REZPEG thus represents a homeostatic approach to cutaneous disease therapy and holds clinical potential in providing long-term, treatment-free disease control.
Collapse
Affiliation(s)
- Jonathan I Silverberg
- Department of Dermatology, George Washington University School of Medicine, Washington, DC, USA
| | - David Rosmarin
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Raj Chovatiya
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
- Center for Medical Dermatology + Immunology Research, Chicago, IL, USA
| | - Thomas Bieber
- Department of Dermatology, University Hospital, Zürich, Switzerland
- Medicine Campus, Davos, Switzerland
| | | | - Lisa Beck
- University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | | - Danni Yu
- Nektar Therapeutics, San Francisco, CA, USA
| | | | - Yi Liu
- Nektar Therapeutics, San Francisco, CA, USA
| | | | | | | | - Ajay Nirula
- Recludix Pharma, San Diego, CA, USA, formerly affiliated with Eli Lilly and Company, Indianapolis, IN, USA
| | | | | |
Collapse
|
33
|
Passeron T, Reinhardt M, Ehst B, Weiss J, Sluzevich J, Sticherling M, Reygagne P, Wohlrab J, Hertl M, Fazel N, Muscianisi E, Fan H, Hampele I, Compagno N. Secukinumab in adult patients with lichen planus: efficacy and safety results from the randomized placebo-controlled proof-of-concept PRELUDE study. Br J Dermatol 2024; 191:680-690. [PMID: 38735684 DOI: 10.1093/bjd/ljae181] [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: 12/04/2023] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND Patients with lichen planus (LP) refractory to available therapies often experience a high disease burden, representing a population with a clear unmet need for new treatments. OBJECTIVES To evaluate the efficacy and safety of secukinumab 300 mg over 32 weeks in adult patients with biopsy-proven cutaneous LP (CLP), mucosal LP (MLP) or lichen planopilaris (LPP) that is inadequately controlled by topical corticosteroids. METHODS PRELUDE was a randomized double-blind placebo-controlled phase II proof-of-concept study that enrolled patients with CLP, MLP or LPP. Eligible patients were randomized to either secukinumab 300 mg every 4 weeks for 32 weeks (SECQ4W) or placebo for 16 weeks followed by secukinumab 300 mg every 2 weeks (SECQ2W) for 16 weeks. The primary endpoint was achievement of the newly designed Investigator's Global Assessment (IGA) score ≤ 2 at week 16. RESULTS Overall, 111 patients were randomized (n = 37 each) to CLP, MLP and LPP cohorts. As the proof-of-concept criteria were not met for any of the three cohorts, the primary objective was not met. A numerically higher proportion of patients achieved IGA ≤ 2 response at week 16 with SECQ4W vs. placebo in the MLP {37.5% [95% credibility interval (Crl) 20.3-57.2] vs. 23.1% (95% Crl 6.5-49.2)} and LPP cohorts [37.5% (95% Crl 20.2-57.3) vs. 30.8% (95% Crl 10.8-57.6)]. In the LPP cohort, a sustained response for IGA ≤ 2 from week 16 to week 32 was achieved with SECQ4W (week 16, 37.5%; week 32, 45.8%), and a substantial improvement was observed in IGA ≤ 2 response in patients from this cohort who switched from placebo (week 16, 30.8%) to SECQ2W after week 16 (week 32, 63.6%). The safety profile was consistent with the known profile of secukinumab and showed no new or unexpected signals. CONCLUSIONS PRELUDE is the first randomized controlled basket trial evaluating interleukin (IL)-17A inhibition with secukinumab across three subtypes of LP. Secukinumab was well tolerated and safe, showing different response rates across the three subtypes, with numerical IGA improvements in MLP and LPP, and no response in CLP. The study raises the question of a differential role of IL-17A across LP subtypes. The novel IGA score showed significant correlation with both patient- and physician-reported outcome measurements.
Collapse
Affiliation(s)
- Thierry Passeron
- University Côte d'Azur, CHU Nice, Department of Dermatology, Nice, France
- University Côte d'Azur, INSERM U1065, C3M, Nice, France
| | | | | | | | - Jason Sluzevich
- Department of Dermatology, Mayo Clinic, Jacksonville, FL, USA
| | - Michael Sticherling
- Hautklinik, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU) Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | | | - Johannes Wohlrab
- Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Michael Hertl
- Newport Dermatology & Laser Associates, Newport Beach, CA, USA
| | - Nasim Fazel
- Department of Dermatology, University of California at Davis School of Medicine, Sacramento, CA, USA
| | | | - Heng Fan
- Novartis Pharma Shanghai, Shanghai, China
| | | | | |
Collapse
|
34
|
Zhang Y, Xu H, Tang Y, Li Y, Zheng F. The levels of amino acid metabolites in serum induce the pathogenesis of atopic dermatitis by mediating the inflammatory protein S100A12. Sci Rep 2024; 14:23435. [PMID: 39379513 PMCID: PMC11461510 DOI: 10.1038/s41598-024-74522-1] [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: 03/16/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease affecting tens of millions of people globally. The causal relationship between metabolites and AD pathology has not yet been formally indicated, and the mediating mechanism by which metabolites affect AD has not yet been explored. This study aimed to determine the genetic relationship between metabolites and AD and to determine the pathways through which amino acid metabolites affect AD. Meta-analysis integrates the results of multiple GWAS analyses using METAL software. Using bidirectional two-sample Mendelian randomization (MR), we analyzed the causal relationships between metabolites and AD. The principal MR test of causal effects was conducted using inverse-variance weighted regression, and we used reverse MR analysis to exclude reverse causality. We also performed the MR-PRESSO test to detect and correct for possible pleiotropic effects, and used the Cochran Q test to assess heterogeneity. Two-step MR was utilized to analyze the mediating factors between amino acid metabolites and the onset of AD. The correlation between mediating factors (inflammatory protein S100A12) and immune cell infiltration was analyzed using the edgeR and GSVA software packages. Using single-cell sequencing data from skin tissues of patients with AD, we studied the regulatory role of the S100A12 gene in immune cells. Multiple drug databases and macromolecular docking were used to search for S100A12-targeting drugs. Bidirectional two-sample MR analyses indicated that twenty-two metabolites and one inflammatory protein (S100A12) were significantly associated with AD pathogenesis. S100A12 is a mediator of amino acid metabolites (N6-methyllysine; N2-acetyl,N6,N6-dimethyllysine and N6,N6-dimethyllysine) that are genetically associated with AD. S100A12 was positively correlated with the infiltration of multiple immune cell types in lesional AD skin. The amino acid metabolites N6-methyllysine; N2-acetyl,N6,N6-dimethyllysine and N6,N6-dimethyllysine influence AD pathogenesis by mediating S100A12 expression.
Collapse
Affiliation(s)
- Yaqi Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Heng Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuhang Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| | - Fengjie Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
35
|
Khalaf AT, Abdalla AN, Ren K, Liu X. Cold atmospheric plasma (CAP): a revolutionary approach in dermatology and skincare. Eur J Med Res 2024; 29:487. [PMID: 39367460 PMCID: PMC11453049 DOI: 10.1186/s40001-024-02088-9] [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: 07/11/2024] [Accepted: 09/28/2024] [Indexed: 10/06/2024] Open
Abstract
Cold atmospheric plasma (CAP) technology has emerged as a revolutionary therapeutic technology in dermatology, recognized for its safety, effectiveness, and minimal side effects. CAP demonstrates substantial antimicrobial properties against bacteria, viruses, and fungi, promotes tissue proliferation and wound healing, and inhibits the growth and migration of tumor cells. This paper explores the versatile applications of CAP in dermatology, skin health, and skincare. It provides an in-depth analysis of plasma technology, medical plasma applications, and CAP. The review covers the classification of CAP, its direct and indirect applications, and the penetration and mechanisms of action of its active components in the skin. Briefly introduce CAP's suppressive effects on microbial infections, detailing its impact on infectious skin diseases and its specific effects on bacteria, fungi, viruses, and parasites. It also highlights CAP's role in promoting tissue proliferation and wound healing and its effectiveness in treating inflammatory skin diseases such as psoriasis, atopic dermatitis, and vitiligo. Additionally, the review examines CAP's potential in suppressing tumor cell proliferation and migration and its applications in cosmetic and skincare treatments. The therapeutic potential of CAP in treating immune-mediated skin diseases is also discussed. CAP presents significant promise as a dermatological treatment, offering a safe and effective approach for various skin conditions. Its ability to operate at room temperature and its broad spectrum of applications make it a valuable tool in dermatology. Finally, introduce further research is required to fully elucidate its mechanisms, optimize its use, and expand its clinical applications.
Collapse
Grants
- grant number JCYJ20220530114204010 This work was supported by the Department of Dermatology, Southern University of Science and Technology Hospital, Shenzhen, China
- grant number JCYJ20220530114204010 This work was supported by the Department of Dermatology, Southern University of Science and Technology Hospital, Shenzhen, China
- grant number JCYJ20220530114204010 This work was supported by the Department of Dermatology, Southern University of Science and Technology Hospital, Shenzhen, China
- grant number JCYJ20220530114204010 This work was supported by the Department of Dermatology, Southern University of Science and Technology Hospital, Shenzhen, China
Collapse
Affiliation(s)
- Ahmad Taha Khalaf
- Medical College, Anhui University of Science and Technology (AUST), Huainan, 232001, China
| | - Ahmed N Abdalla
- Faculty of Electronic Information Engineering, Huaiyin Institute of Technology, Huai'an, 223003, China
| | - Kaixuan Ren
- Department of Dermatology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710006, China
| | - Xiaoming Liu
- Department of Dermatology, Southern University of Science and Technology Hospital, Shenzhen, 518055, China.
| |
Collapse
|
36
|
Lee SH, Sacks DL. Resilience of dermis resident macrophages to inflammatory challenges. Exp Mol Med 2024; 56:2105-2112. [PMID: 39349826 PMCID: PMC11542019 DOI: 10.1038/s12276-024-01313-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: 05/17/2024] [Revised: 06/24/2024] [Accepted: 07/04/2024] [Indexed: 10/03/2024] Open
Abstract
The skin serves as a complex barrier organ populated by tissue-resident macrophages (TRMs), which play critical roles in defense, homeostasis, and tissue repair. This review examines the functions of dermis resident TRMs in different inflammatory settings, their embryonic origins, and their long-term self-renewal capabilities. We highlight the M2-like phenotype of dermal TRMs and their specialized functions in perivascular and perineuronal niches. Their interactions with type 2 immune cells, autocrine cytokines such as IL-10, and their phagocytic clearance of apoptotic cells have been explored as mechanisms for M2-like dermal TRM self-maintenance and function. In conclusion, we address the need to bridge murine models with human studies, with the possibility of targeting TRMs to promote skin immunity or restrain cutaneous pathology.
Collapse
Affiliation(s)
- Sang Hun Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David L Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
37
|
Sol S, Boncimino F, Todorova K, Mandinova A. Unraveling the Functional Heterogeneity of Human Skin at Single-Cell Resolution. Hematol Oncol Clin North Am 2024; 38:921-938. [PMID: 38839486 DOI: 10.1016/j.hoc.2024.05.001] [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: 06/07/2024]
Abstract
The skin consists of several cell populations, including epithelial, immune, and stromal cells. Recently, there has been a significant increase in single-cell RNA-sequencing studies, contributing to the development of a consensus Human Skin Cell Atlas. The aim is to understand skin biology better and identify potential therapeutic targets. The present review utilized previously published single-cell RNA-sequencing datasets to explore human skin's cellular and functional heterogeneity. Additionally, it summarizes the functional significance of newly identified cell subpopulations in processes such as wound healing and aging.
Collapse
Affiliation(s)
- Stefano Sol
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Fabiana Boncimino
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Kristina Todorova
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Anna Mandinova
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA; Broad Institute of Harvard and MIT, 7 Cambridge Center, MA 02142, USA; Harvard Stem Cell Institute, 7 Divinity Avenue Cambridge, MA 02138, USA.
| |
Collapse
|
38
|
Schepps S, Xu J, Yang H, Mandel J, Mehta J, Tolotta J, Baker N, Tekmen V, Nikbakht N, Fortina P, Fuentes I, LaFleur B, Cho RJ, South AP. Skin in the game: a review of single-cell and spatial transcriptomics in dermatological research. Clin Chem Lab Med 2024; 62:1880-1891. [PMID: 38656304 DOI: 10.1515/cclm-2023-1245] [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: 11/03/2023] [Accepted: 02/29/2024] [Indexed: 04/26/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) are two emerging research technologies that uniquely characterize gene expression microenvironments on a cellular or subcellular level. The skin, a clinically accessible tissue composed of diverse, essential cell populations, serves as an ideal target for these high-resolution investigative approaches. Using these tools, researchers are assembling a compendium of data and discoveries in healthy skin as well as a range of dermatologic pathophysiologies, including atopic dermatitis, psoriasis, and cutaneous malignancies. The ongoing advancement of single-cell approaches, coupled with anticipated decreases in cost with increased adoption, will reshape dermatologic research, profoundly influencing disease characterization, prognosis, and ultimately clinical practice.
Collapse
Affiliation(s)
- Samuel Schepps
- Department of Dermatology and Cutaneous Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
| | - Jonathan Xu
- Department of Dermatology and Cutaneous Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
| | - Henry Yang
- Department of Dermatology and Cutaneous Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
| | - Jenna Mandel
- Department of Dermatology and Cutaneous Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
| | - Jaanvi Mehta
- Department of Dermatology and Cutaneous Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
| | - Julianna Tolotta
- Department of Dermatology and Cutaneous Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
| | - Nicole Baker
- Department of Dermatology and Cutaneous Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
| | - Volkan Tekmen
- Department of Dermatology and Cutaneous Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
| | - Neda Nikbakht
- Department of Dermatology and Cutaneous Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
- Department of Pharmacology, Physiology and Cancer Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
| | - Paolo Fortina
- Department of Pharmacology, Physiology and Cancer Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
- International Federation of Clinical Chemistry Working Group on Single Cell and Spatial Transcriptomics, Milan, Italy
| | - Ignacia Fuentes
- International Federation of Clinical Chemistry Working Group on Single Cell and Spatial Transcriptomics, Milan, Italy
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Directora de Investigación Fundación DEBRA Chile, Santiago, Chile
| | - Bonnie LaFleur
- International Federation of Clinical Chemistry Working Group on Single Cell and Spatial Transcriptomics, Milan, Italy
- R. Ken Coit College of Pharmacy, University of Arizona, University of Arizona Cancer Center, Tucson, AZ, USA
| | - Raymond J Cho
- International Federation of Clinical Chemistry Working Group on Single Cell and Spatial Transcriptomics, Milan, Italy
- Department of Dermatology, University of San Francisco, San Francisco, CA, USA
| | - Andrew P South
- Department of Pharmacology, Physiology and Cancer Biology, 6559 Thomas Jefferson University , Philadelphia, PA, USA
- International Federation of Clinical Chemistry Working Group on Single Cell and Spatial Transcriptomics, Milan, Italy
| |
Collapse
|
39
|
Sati S, Huang J, Kersh AE, Jones P, Ahart O, Murphy C, Prouty SM, Hedberg ML, Jain V, Gregory SG, Leung DH, Seykora JT, Rosenbach M, Leung TH. Recruitment of CXCR4+ type 1 innate lymphoid cells distinguishes sarcoidosis from other skin granulomatous diseases. J Clin Invest 2024; 134:e178711. [PMID: 39225100 PMCID: PMC11364400 DOI: 10.1172/jci178711] [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: 12/20/2023] [Accepted: 06/25/2024] [Indexed: 09/04/2024] Open
Abstract
Sarcoidosis is a multiorgan granulomatous disease that lacks diagnostic biomarkers and targeted treatments. Using blood and skin from patients with sarcoid and non-sarcoid skin granulomas, we discovered that skin granulomas from different diseases exhibit unique immune cell recruitment and molecular signatures. Sarcoid skin granulomas were specifically enriched for type 1 innate lymphoid cells (ILC1s) and B cells and exhibited molecular programs associated with formation of mature tertiary lymphoid structures (TLSs), including increased CXCL12/CXCR4 signaling. Lung sarcoidosis granulomas also displayed similar immune cell recruitment. Thus, granuloma formation was not a generic molecular response. In addition to tissue-specific effects, patients with sarcoidosis exhibited an 8-fold increase in circulating ILC1s, which correlated with treatment status. Multiple immune cell types induced CXCL12/CXCR4 signaling in sarcoidosis, including Th1 T cells, macrophages, and ILCs. Mechanistically, CXCR4 inhibition reduced sarcoidosis-activated immune cell migration, and targeting CXCR4 or total ILCs attenuated granuloma formation in a noninfectious mouse model. Taken together, our results show that ILC1s are a tissue and circulating biomarker that distinguishes sarcoidosis from other skin granulomatous diseases. Repurposing existing CXCR4 inhibitors may offer a new targeted treatment for this devastating disease.
Collapse
Affiliation(s)
- Satish Sati
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jianhe Huang
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Anna E. Kersh
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Parker Jones
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Olivia Ahart
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Christina Murphy
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Stephen M. Prouty
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Matthew L. Hedberg
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | - Simon G. Gregory
- Duke Molecular Physiology Institute, Durham, North Carolina, USA
| | | | - John T. Seykora
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Misha Rosenbach
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Thomas H. Leung
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
40
|
Zhu X, Wu W. The impact of lipidome on five inflammatory skin diseases: a Mendelian randomization study. Arch Dermatol Res 2024; 316:565. [PMID: 39177801 DOI: 10.1007/s00403-024-03294-5] [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: 07/01/2024] [Revised: 07/16/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
OBJECTIVE Two-sample Mendelian randomization (TSMR) was employed to examine the association between lipidome and five inflammatory skin diseases. METHOD To evaluate the association between various molecular subtypes of lipidome and the risk of five inflammatory skin diseases, we analyzed a comprehensive GWAS dataset comprising 179 lipidome. The Two-Sample Mendelian Randomization (TSMR) method was employed to investigate causal relationships. Heterogeneity and pleiotropy were assessed using Cochran's Q test, MR-Egger intercept test, and MR-PRESSO global test. Additionally, a sensitivity analysis was conducted to evaluate the influence of individual single nucleotide polymorphisms on Mendelian Randomization study. RESULTS Using 179 serum lipidome as exposures and five common inflammatory skin diseases as outcomes, we investigated their associations in this large-scale study. Our findings reveal significant impacts of glycerophospholipids, glycerolipids, and sphingomyelins on inflammatory skin diseases. Glycerophospholipids were protective against pemphigus but predominantly posed risks for other inflammatory skin diseases. Specifically, phosphatidylcholine (16:0_0:0) exhibited the most significant risk association with lichen planus (OR = 1.25, 95% CI 1.11-1.40, P < 0.001). Conversely, glycerolipids showed no effect on lichen planus but were protective against pemphigus while potentially posing risks for other conditions. Triacylglycerol (46:2) showed the most substantial risk association with vitiligo (OR = 1.99, 95% CI 1.35-2.93, P < 0.001). Furthermore, sphingomyelins had no effect on atopic dermatitis but posed potential risks for other inflammatory skin diseases. Sphingomyelin (d40:1) notably emerged as a significant risk factor for pemphigus (OR = 1.91, 95% CI 1.37-2.66, P < 0.001). CONCLUSIONS This study has elucidated the potential harmful effects of glycerophospholipids, glycerolipids, and sphingomyelins on inflammatory skin diseases, while also providing valuable insights for future research into the pathophysiology, prevention and treatment of these conditions.
Collapse
Affiliation(s)
- Xu Zhu
- Department of Dermatology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Wenzhong Wu
- Department of Dermatology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China.
| |
Collapse
|
41
|
Ren G, Zhang Y, Liu J, Cheng W, Wu D, Han M, Zeng Y, Zhao X, Hu L, Zeng M, Gurram RK, Hu X, Zhou B, Hou Z, Zhu J, Jin W, Zhong C. Decreased GATA3 levels cause changed mouse cutaneous innate lymphoid cell fate, facilitating hair follicle recycling. Dev Cell 2024; 59:1809-1823.e6. [PMID: 38723629 PMCID: PMC11265981 DOI: 10.1016/j.devcel.2024.04.015] [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: 08/15/2023] [Revised: 02/08/2024] [Accepted: 04/16/2024] [Indexed: 07/25/2024]
Abstract
In mice, skin-resident type 2 innate lymphoid cells (ILC2s) exhibit some ILC3-like characteristics. However, the underlying mechanism remains elusive. Here, we observed lower expression of the ILC2 master regulator GATA3 specifically in cutaneous ILC2s (cILC2s) compared with canonical ILC2s, in line with its functionally divergent role in transcriptional control in cILC2s. Decreased levels of GATA3 enabled the expansion of RORγt fate-mapped (RORγtfm+) cILC2s after postnatal days, displaying certain similarities to ILC3s. Single-cell trajectory analysis showed a sequential promotion of the RORγtfm+ cILC2 divergency by RORγt and GATA3. Notably, during hair follicle recycling, these RORγtfm+ cILC2s accumulated around the hair follicle dermal papilla (DP) region to facilitate the process. Mechanistically, we found that GATA3-mediated integrin α3β1 upregulation on RORγtfm+ cILC2s was required for their positioning around the DP. Overall, our study demonstrates a distinct regulatory role of GATA3 in cILC2s, particularly in promoting the divergence of RORγtfm+ cILC2s to facilitate hair follicle recycling.
Collapse
Affiliation(s)
- Guanqun Ren
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Yime Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Jiamin Liu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Wenwen Cheng
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Di Wu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China; Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Mengwei Han
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Yanyu Zeng
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Xingyu Zhao
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Luni Hu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Min Zeng
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China; Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Rama Krishna Gurram
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD 20892, USA
| | - Xiaole Hu
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Bo Zhou
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zhiyuan Hou
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, Peking University Health Science Center, Beijing 100191, China
| | - Jinfang Zhu
- Molecular and Cellular Immunoregulation Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD 20892, USA
| | - Wenfei Jin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.
| | - Chao Zhong
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Researches on Major Immunology-Related Diseases, Peking University, Beijing 100191, China.
| |
Collapse
|
42
|
Roso-Mares A, Andújar I, Díaz Corpas T, Sun BK. Non-coding RNAs as skin disease biomarkers, molecular signatures, and therapeutic targets. Hum Genet 2024; 143:801-812. [PMID: 37580609 DOI: 10.1007/s00439-023-02588-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/23/2023] [Indexed: 08/16/2023]
Abstract
Non-coding RNAs (ncRNAs) are emerging as biomarkers, molecular signatures, and therapeutic tools and targets for diseases. In this review, we focus specifically on skin diseases to highlight how two classes of ncRNAs-microRNAs and long noncoding RNAs-are being used to diagnose medical conditions of unclear etiology, improve our ability to guide treatment response, and predict disease prognosis. Furthermore, we explore how ncRNAs are being used as both as drug targets and associated therapies have unique benefits, risks, and challenges to development, but offer a distinctive promise for improving patient care and outcomes.
Collapse
Affiliation(s)
- Andrea Roso-Mares
- Department of Dermatology, University of California San Diego, San Diego, CA, USA
- Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Isabel Andújar
- Department of Pharmacology, University of Valencia, Valencia, Spain
| | - Tania Díaz Corpas
- Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
- Department of Dermatology, Hospital Dr Peset, Valencia, Spain
| | - Bryan K Sun
- Department of Dermatology, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
43
|
Patel JR, Joel MZ, Lee KK, Kambala A, Cornman H, Oladipo O, Taylor M, Imo BU, Ma EZ, Manjunath J, Kollhoff AL, Deng J, Parthasarathy V, Cravero K, Marani M, Szeto M, Zhao R, Sankararaman S, Li R, Henry S, Pritchard T, Rebecca V, Kwatra MM, Ho WJ, Dong X, Kang S, Kwatra SG. Single-Cell RNA Sequencing Reveals Dysregulated POSTN+WNT5A+ Fibroblast Subclusters in Prurigo Nodularis. J Invest Dermatol 2024; 144:1568-1578.e5. [PMID: 38246584 DOI: 10.1016/j.jid.2023.12.021] [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: 10/09/2023] [Revised: 12/17/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Prurigo nodularis (PN) is an intensely pruritic, inflammatory skin disease with a poorly understood pathogenesis. We performed single-cell transcriptomic profiling of 28,695 lesional and nonlesional PN cells. Lesional PN has increased dysregulated fibroblasts (FBs) and myofibroblasts. FBs in lesional PN were shifted toward a cancer-associated FB-like phenotype, with POSTN+WNT5A+ cancer-associated FBs increased in PN and similarly so in squamous cell carcinoma. A multicenter cohort study revealed an increased risk of squamous cell carcinoma and cancer-associated FB-associated malignancies (breast and colorectal) in patients with PN. Systemic fibroproliferative diseases (renal sclerosis and idiopathic pulmonary fibrosis) were upregulated in patients with PN. Ligand-receptor analyses demonstrated an FB neuronal axis with FB-derived WNT5A and periostin interactions with neuronal receptors melanoma cell adhesion molecule and ITGAV. These findings identify a pathogenic and targetable POSTN+WNT5A+ FB subpopulation that may predispose cancer-associated FB-associated malignancies in patients with PN.
Collapse
Affiliation(s)
- Jay R Patel
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marina Z Joel
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kevin K Lee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anusha Kambala
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hannah Cornman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Olusola Oladipo
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Matthew Taylor
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brenda Umenita Imo
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Emily Z Ma
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jaya Manjunath
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alexander L Kollhoff
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - June Deng
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Varsha Parthasarathy
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Karen Cravero
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Melika Marani
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mindy Szeto
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ryan Zhao
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sreenidhi Sankararaman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ruixiang Li
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shanae Henry
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thomas Pritchard
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vito Rebecca
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Madan M Kwatra
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, USA; Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Won Jin Ho
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sewon Kang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shawn G Kwatra
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
44
|
Anitua E, Tierno R, Azkargorta M, Elortza F, Alkhraisat MH. Effect of Health Status and Heat-Induced Inactivation on the Proteomic Profile of Plasma Rich in Growth Factors Obtained from Donors with Chronic Inflammatory Skin Conditions. Biomolecules 2024; 14:763. [PMID: 39062477 PMCID: PMC11275043 DOI: 10.3390/biom14070763] [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: 05/20/2024] [Revised: 06/16/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Atopic dermatitis, psoriasis and lichen sclerosus are among the most challenging conditions treated by dermatologists worldwide, with potentially significant physical, social and psychological impacts. Emerging evidence suggests that autologous-platelet-rich plasma could be used to manage skin inflammation. However, the presence of soluble autoimmune components could hinder their therapeutic potential. The aim of this study was to analyze the proteomic profile of plasma rich in growth factors (PRGFs) obtained from donors with inflammatory skin conditions to evaluate the impact of skin health status on the composition and bioactivity of PRGF-based treatments. Venous blood from healthy volunteers and patients with psoriasis, lichen sclerosus and atopic dermatitis was processed to produce PRGF supernatant. Half of the samples were subjected to an additional thermal treatment (56 °C) to inactivate inflammatory and immune molecules. Proteomic analysis was performed to assess the protein profile of PRGFs from healthy and non-healthy patients and the effect of Immunosafe treatment. Differential abundance patterns of several proteins related to key biological processes have been identified, including complement activation, blood coagulation, and glycolysis- and gluconeogenesis-related genes. These results also demonstrate that the thermal treatment (Immunosafe) contributes to the inactivation of the complement system and, as a consequence, reduction in the immunogenic potential of PRGF products.
Collapse
Affiliation(s)
- Eduardo Anitua
- University Institute for Regenerative Medicine and Oral Implantology (UIRMI), 01007 Vitoria, Spain; (R.T.); (M.H.A.)
- BTI-Biotechnology Institute, 01005 Vitoria, Spain
| | - Roberto Tierno
- University Institute for Regenerative Medicine and Oral Implantology (UIRMI), 01007 Vitoria, Spain; (R.T.); (M.H.A.)
- BTI-Biotechnology Institute, 01005 Vitoria, Spain
| | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, 48160 Derio, Spain; (M.A.); (F.E.)
| | - Félix Elortza
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, 48160 Derio, Spain; (M.A.); (F.E.)
| | - Mohammad H. Alkhraisat
- University Institute for Regenerative Medicine and Oral Implantology (UIRMI), 01007 Vitoria, Spain; (R.T.); (M.H.A.)
- BTI-Biotechnology Institute, 01005 Vitoria, Spain
| |
Collapse
|
45
|
Jin SP, Lee K, Bang YJ, Jeon YH, Jung S, Choi SJ, Lee JS, Kim J, Guttman-Yassky E, Park CG, Kim HJ, Hong S, Lee DH. Mapping the immune cell landscape of severe atopic dermatitis by single-cell RNA-seq. Allergy 2024; 79:1584-1597. [PMID: 38817208 DOI: 10.1111/all.16121] [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: 03/14/2023] [Revised: 03/10/2024] [Accepted: 03/18/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Efforts to profile atopic dermatitis (AD) tissues have intensified, yet comprehensive analysis of systemic immune landscapes in severe AD remains crucial. METHODS Employing single-cell RNA sequencing, we analyzed over 300,000 peripheral blood mononuclear cells from 12 severe AD patients (Eczema area and severity index (EASI) > 21) and six healthy controls. RESULTS Results revealed significant immune cell shifts in AD patients, including increased Th2 cell abundance, reduced NK cell clusters with compromised cytotoxicity, and correlated Type 2 innate lymphoid cell proportions with disease severity. Moreover, unique monocyte clusters reflecting activated innate immunity emerged in very severe AD (EASI > 30). While overall dendritic cells (DCs) counts decreased, a distinct Th2-priming subset termed "Th2_DC" correlated strongly with disease severity, validated across skin tissue data, and flow cytometry with additional independent severe AD samples. Beyond the recognized role of Th2 adaptive immunity, our findings highlight significant innate immune cell alterations in severe AD, implicating their roles in disease pathogenesis and therapeutic potentials. CONCLUSION Apart from the widely recognized role of Th2 adaptive immunity in AD pathogenesis, alterations in innate immune cells and impaired cytotoxic cells have also been observed in severe AD. The impact of these alterations on disease pathogenesis and the effectiveness of potential therapeutic targets requires further investigation.
Collapse
Affiliation(s)
- Seon-Pil Jin
- Department of Dermatology, Seoul National University Hospital, Seoul, Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea
- Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Seoul, Korea
| | - Kyungchun Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, Korea
| | - Yoon Ji Bang
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Yun-Hui Jeon
- Department of Dermatology, Seoul National University Hospital, Seoul, Korea
- Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Seoul, Korea
| | - Sunyoung Jung
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - So-Jung Choi
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
| | - Ji Su Lee
- Department of Dermatology, Seoul National University Hospital, Seoul, Korea
- Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Seoul, Korea
| | - Junhan Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, Korea
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Chung-Gyu Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Seoul National University Hospital, Seoul, Korea
| | - Hyun Je Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Seoul National University Hospital, Seoul, Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Korea
| | - Seunghee Hong
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul, Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University Hospital, Seoul, Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea
- Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
46
|
Guo Y, Mao W, Bai N, Jin L, Tang S, Lin X, Ni J, Liu X, Fu H, Shou Q. Integrated network pharmacological analysis revealed that Smilax glabra Roxb. alleviates IMQ-induced psoriatic skin inflammation through regulating T cell immune response. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117836. [PMID: 38301985 DOI: 10.1016/j.jep.2024.117836] [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: 12/04/2023] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 02/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Psoriasis is an autoimmune disease characterized by dysfunctional T cells and dysregulated immune responses. Smilax glabra Roxb. (SGR) is a formulation used in Traditional Chinese Medicine for the treatment of inflammatory skin disorders, including psoriasis. This study explores the scientific basis for its use by examining the effects of SGR on T cell differentiation and insulin receptor signaling, relevant pathways implicated in the pathophysiology of psoriasis. AIM OF THE STUDY This study investigates the therapeutic potential of SGR (a Chinese medicine) in psoriasis and its impact on T cell differentiation. MATERIALS AND METHODS An integrated network pharmacology and bioinformatics approach was employed to elucidate the mechanisms of SGR in regulating T cell differentiation. A psoriasis mouse model was utilized to evaluate the effects of SGR on T cell subsets. Immunohistochemistry and gene expression analyses were conducted to investigate the modulation of insulin receptor signaling pathways by SGR. RESULTS SGR treatment effectively reset the expression of various T cell subsets in the psoriasis mouse model, suggesting its ability to regulate T cell differentiation and immune function. Furthermore, SGR treatment inhibited insulin receptor signaling and downstream pathways, including PI3K/AKT and ERK, in psoriatic skin lesions. This indicates that SGR may exert its therapeutic effects through modulation of the insulin receptor signaling pathway. CONCLUSIONS This study provides novel insights into the therapeutic potential of SGR in psoriasis. By modulating T cell differentiation and targeting the insulin receptor signaling pathway, SGR holds promise as a potential treatment option for psoriasis.
Collapse
Affiliation(s)
- Yingxue Guo
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Weiye Mao
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China; Zhezhong Laboratory, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Ningning Bai
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Lu Jin
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Shuiyan Tang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xiaochen Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Jianyu Ni
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xia Liu
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Huiying Fu
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Qiyang Shou
- Second Clinical Medical College, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China; Zhezhong Laboratory, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
47
|
Liu G, Wang Z, Li S. Heterogeneity and plasticity of tissue-resident memory T cells in skin diseases and homeostasis: a review. Front Immunol 2024; 15:1378359. [PMID: 38779662 PMCID: PMC11109409 DOI: 10.3389/fimmu.2024.1378359] [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: 01/29/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Skin tissue-resident memory T (Trm) cells are produced by antigenic stimulation and remain in the skin for a long time without entering the peripheral circulation. In the healthy state Trm cells can play a patrolling and surveillance role, but in the disease state Trm cells differentiate into various phenotypes associated with different diseases, exhibit different localizations, and consequently have local protective or pathogenic roles, such as disease recurrence in vitiligo and maintenance of immune homeostasis in melanoma. The most common surface marker of Trm cells is CD69/CD103. However, the plasticity of tissue-resident memory T cells after colonization remains somewhat uncertain. This ambiguity is largely due to the variation in the functionality and ultimate destination of Trm cells produced from memory cells differentiated from diverse precursors. Notably, the presence of Trm cells is not stationary across numerous non-lymphoid tissues, most notably in the skin. These cells may reenter the blood and distant tissue sites during the recall response, revealing the recycling and migration potential of the Trm cell progeny. This review focuses on the origin and function of skin Trm cells, and provides new insights into the role of skin Trm cells in the treatment of autoimmune skin diseases, infectious skin diseases, and tumors.
Collapse
Affiliation(s)
- Guomu Liu
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun, China
| | - Ziyue Wang
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Shanshan Li
- Department of Dermatology and Venereology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
48
|
Kratchmarov R, Djeddi S, Dunlap G, He W, Jia X, Burk CM, Ryan T, McGill A, Allegretti JR, Kataru RP, Mehrara BJ, Taylor EM, Agarwal S, Bhattacharyya N, Bergmark RW, Maxfield AZ, Lee S, Roditi R, Dwyer DF, Boyce JA, Buchheit KM, Laidlaw TM, Shreffler WG, Rao DA, Gutierrez-Arcelus M, Brennan PJ. TCF1-LEF1 co-expression identifies a multipotent progenitor cell (T H2-MPP) across human allergic diseases. Nat Immunol 2024; 25:902-915. [PMID: 38589618 PMCID: PMC11849131 DOI: 10.1038/s41590-024-01803-2] [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] [Received: 01/21/2024] [Accepted: 03/06/2024] [Indexed: 04/10/2024]
Abstract
Repetitive exposure to antigen in chronic infection and cancer drives T cell exhaustion, limiting adaptive immunity. In contrast, aberrant, sustained T cell responses can persist over decades in human allergic disease. To understand these divergent outcomes, we employed bioinformatic, immunophenotyping and functional approaches with human diseased tissues, identifying an abundant population of type 2 helper T (TH2) cells with co-expression of TCF7 and LEF1, and features of chronic activation. These cells, which we termed TH2-multipotent progenitors (TH2-MPP) could self-renew and differentiate into cytokine-producing effector cells, regulatory T (Treg) cells and follicular helper T (TFH) cells. Single-cell T-cell-receptor lineage tracing confirmed lineage relationships between TH2-MPP, TH2 effectors, Treg cells and TFH cells. TH2-MPP persisted despite in vivo IL-4 receptor blockade, while thymic stromal lymphopoietin (TSLP) drove selective expansion of progenitor cells and rendered them insensitive to glucocorticoid-induced apoptosis in vitro. Together, our data identify TH2-MPP as an aberrant T cell population with the potential to sustain type 2 inflammation and support the paradigm that chronic T cell responses can be coordinated over time by progenitor cells.
Collapse
Affiliation(s)
- Radomir Kratchmarov
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah Djeddi
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Garrett Dunlap
- Division of Rheumatology, Inflammation, Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wenqin He
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaojiong Jia
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Caitlin M Burk
- Center for Immunology and Inflammatory Diseases and Food Allergy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tessa Ryan
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alanna McGill
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica R Allegretti
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raghu P Kataru
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Babak J Mehrara
- Plastic and Reconstructive Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Erin M Taylor
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Shailesh Agarwal
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Neil Bhattacharyya
- Massachusetts Eye & Ear Institute, Harvard Medical School, Boston, MA, USA
| | - Regan W Bergmark
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Surgery and Public Health, Brigham and Women's Hospital, Boston, MA, USA
| | - Alice Z Maxfield
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stella Lee
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rachel Roditi
- Division of Otolaryngology Head and Neck Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel F Dwyer
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua A Boyce
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathleen M Buchheit
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tanya M Laidlaw
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wayne G Shreffler
- Center for Immunology and Inflammatory Diseases and Food Allergy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Patrick J Brennan
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
Lin Q, Cai B, Ke R, Chen L, Ni X, Liu H, Lin X, Wang B, Shan X. Integrative bioinformatics and experimental validation of hub genetic markers in acne vulgaris: Toward personalized diagnostic and therapeutic strategies. J Cosmet Dermatol 2024; 23:1777-1799. [PMID: 38268224 DOI: 10.1111/jocd.16152] [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: 10/20/2023] [Accepted: 12/10/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Acne vulgaris is a widespread chronic inflammatory dermatological condition. The precise molecular and genetic mechanisms of its pathogenesis remain incompletely understood. This research synthesizes existing databases, targeting a comprehensive exploration of core genetic markers. METHODS Gene expression datasets (GSE6475, GSE108110, and GSE53795) were retrieved from the GEO. Differentially expressed genes (DEGs) were identified using the limma package. Enrichment analyses were conducted using GSVA for pathway assessment and clusterProfiler for GO and KEGG analyses. PPI networks and immune cell infiltration were analyzed using the STRING database and ssGSEA, respectively. We investigated the correlation between hub gene biomarkers and immune cell infiltration using Spearman's rank analysis. ROC curve analysis validated the hub genes' diagnostic accuracy. miRNet, TarBase v8.0, and ChEA3 identified miRNA/transcription factor-gene interactions, while DrugBank delineated drug-gene interactions. Experiments utilized HaCaT cells stimulated with Propionibacterium acnes, treated with retinoic acid and methotrexate, and evaluated using RT-qPCR, ELISA, western blot, lentiviral transduction, CCK-8, wound-healing, and transwell assays. RESULTS There were 104 genes with consistent differences across the three datasets of paired acne and normal skin. Functional analyses emphasized the significant enrichment of these DEGs in immune-related pathways. PPI network analysis pinpointed hub genes PTPRC, CXCL8, ITGB2, and MMP9 as central players in acne pathogenesis. Elevated levels of specific immune cell infiltration in acne lesions corroborated the inflammatory nature of the disease. ROC curve analysis identified the acne diagnostic potential of four hub genes. Key miRNAs, particularly hsa-mir-124-3p, and central transcription factors like TFEC were noted as significant regulators. In vitro validation using HaCaT cells confirmed the upregulation of hub genes following Propionibacterium acnes exposure, while CXCL8 knockdown reduced pro-inflammatory cytokines, cell proliferation, and migration. DrugBank insights led to the exploration of retinoic acid and methotrexate, both of which mitigated gene expression upsurge and inflammatory mediator secretion. CONCLUSION This comprehensive study elucidated pivotal genes associated with acne pathogenesis, notably PTPRC, CXCL8, ITGB2, and MMP9. The findings underscore potential biomarkers, therapeutic targets, and the therapeutic potential of agents like retinoic acid and methotrexate. The congruence between bioinformatics and experimental validations suggests promising avenues for personalized acne treatments.
Collapse
Affiliation(s)
- Qian Lin
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Beichen Cai
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Ruonan Ke
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian, China
| | - Lu Chen
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xuejun Ni
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Hekun Liu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Xinjian Lin
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, Fujian, China
| | - Biao Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiuying Shan
- Department of Plastic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
50
|
Taylor MA, El Kurdi A, Hailer A, Wang S, Yuan M, Mukhopadhyay S, Bhutani T, North JP, Cho RJ, Cheng JB. Optimizing Single T-Cell Transcriptomic Discrimination of Atopic Dermatitis Versus Psoriasis Vulgaris. J Invest Dermatol 2024; 144:898-901.e3. [PMID: 37879399 DOI: 10.1016/j.jid.2023.09.283] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 10/27/2023]
Affiliation(s)
- Mark A Taylor
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Abdullah El Kurdi
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Ashley Hailer
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA; Dermatology, Veterans Affairs Medical Center, San Francisco, California, USA
| | - Sijia Wang
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA; Dermatology, Veterans Affairs Medical Center, San Francisco, California, USA; Department of Dermatology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Michelle Yuan
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | | | - Tina Bhutani
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey P North
- Dermatopathology Service, University of California San Francisco, San Francisco, California, USA
| | - Raymond J Cho
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA.
| | - Jeffrey B Cheng
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA; Dermatology, Veterans Affairs Medical Center, San Francisco, California, USA.
| |
Collapse
|