51
|
He J, Zhao M, Ma X, Li D, Kong J, Yang F. The role and application of three IFN-related reactions in psoriasis. Biomed Pharmacother 2023; 167:115603. [PMID: 37776636 DOI: 10.1016/j.biopha.2023.115603] [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: 07/10/2023] [Revised: 09/16/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
The pathophysiology of psoriasis is a highly complicated one. Due to the disease's specificity, it not only affects the patient's skin negatively but also manifests systemic pathological changes. These clinical symptoms seriously harm the patient's physical and mental health. IFN, a common immunomodulatory factor, has been increasingly demonstrated to have a significant role in the development of psoriatic skin disease. Psoriasis is connected with a variety of immunological responses. New targets for the therapy of autoimmune skin diseases may emerge from further research on the mechanics of the associated IFN upstream and downstream pathways. Different forms of IFNs do not behave in the same manner in psoriasis, and understanding how different types of IFNs are involved in psoriasis may provide a better notion for future research. This review focuses on the involvement of three types of IFNs in psoriasis and related therapeutic investigations, briefly describing the three IFNs' production and signaling, as well as the dual effects of IFNs on the skin. It is intended that it would serve as a model for future research.
Collapse
Affiliation(s)
- Jiaming He
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Minghui Zhao
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaoyu Ma
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Dilong Li
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingyan Kong
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Fan Yang
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
52
|
Affolter VK. Cytotoxic dermatitis: Review of the interface dermatitis pattern in veterinary skin diseases. Vet Pathol 2023; 60:770-782. [PMID: 37650259 DOI: 10.1177/03009858231195080] [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: 09/01/2023]
Abstract
Interface dermatitis or lichenoid interface dermatitis refers to a cutaneous inflammatory pattern in which keratinocyte cell death is the essential feature. These terms have evolved from the originally described lichenoid tissue reaction. These lesions are the basis for an important group of skin diseases in animals and people where cytotoxic T-cell-mediated epidermal damage is a major pathomechanism. Yet, for largely historical reasons these commonly used morphological diagnostic terms do not reflect the essential nature of the lesion. An emphasis on subsidiary lesions, such as the presence of a lichenoid band, and definitions based on anatomical features, such as location at the dermo-epidermal location, may cause confusion and even misdiagnosis. This review covers historical aspects of the terminology, including the origin of terms such as "lichenoid." The types of cell death involved and the histopathologic lesions are described. Etiopathogenesis is discussed in terms of aberrations of immune/inflammatory mechanisms focusing on cutaneous lupus erythematosus, erythema multiforme, and Stevens-Johnson syndrome/toxic epidermal necrolysis. Mechanisms have most extensively been studied in humans and laboratory animals and the discussion is centered on these species. As interface dermatitis is firmly entrenched in dermatological parlance, rather than using "cytotoxic" as its substitute, the terminologies "interface cytotoxic dermatitis" and "panepidermal cytotoxic dermatitis" are recommended, based on location and extent of epithelium affected.
Collapse
|
53
|
Gao Z, Zhai X, Yan G, Tian Y, Huang X, Wu Q, Yuan L, Su L. Bioinformatics analyses of gene expression profile to identify pathogenic mechanisms for COVID-19 infection and cutaneous lupus erythematosus. Front Immunol 2023; 14:1268912. [PMID: 38022551 PMCID: PMC10644101 DOI: 10.3389/fimmu.2023.1268912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Objective The global mortality rates have surged due to the ongoing coronavirus disease 2019 (COVID-19), leading to a worldwide catastrophe. Increasing incidents of patients suffering from cutaneous lupus erythematosus (CLE) exacerbations after either contracting COVID-19 or getting immunized against it, have been observed in recent research. However, the precise intricacies that prompt this unexpected complication are yet to be fully elucidated. This investigation seeks to probe into the molecular events inciting this adverse outcome. Method Gene expression patterns from the Gene Expression Omnibus (GEO) database, specifically GSE171110 and GSE109248, were extracted. We then discovered common differentially expressed genes (DEGs) in both COVID-19 and CLE. This led to the creation of functional annotations, formation of a protein-protein interaction (PPI) network, and identification of key genes. Furthermore, regulatory networks relating to these shared DEGs and significant genes were constructed. Result We identified 214 overlapping DEGs in both COVID-19 and CLE datasets. The following functional enrichment analysis of these DEGs highlighted a significant enrichment in pathways related to virus response and infectious disease in both conditions. Next, a PPI network was constructed using bioinformatics tools, resulting in the identification of 5 hub genes. Finally, essential regulatory networks including transcription factor-gene and miRNA-gene interactions were determined. Conclusion Our findings demonstrate shared pathogenesis between COVID-19 and CLE, offering potential insights for future mechanistic investigations. And the identification of common pathways and key genes in these conditions may provide novel avenues for research.
Collapse
Affiliation(s)
- Zhenyu Gao
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Xinchao Zhai
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Guoqing Yan
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Yao Tian
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Xia Huang
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Qingchao Wu
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Lin Yuan
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Linchong Su
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Minda Hospital of Hubei Minzu University, Enshi, China
- Department of Rheumatology and Immunology, Minda Hospital of Hubei Minzu University, Enshi, China
| |
Collapse
|
54
|
Abstract
Systemic lupus erythematosus (SLE) is a severe multisystem autoimmune disease that can cause injury in almost every body system. While considered a classic example of autoimmunity, it is still relatively poorly understood. Treatment with immunosuppressive agents is challenging, as many agents are relatively non-specific, and the underlying disease is characterized by unpredictable flares and remissions. This State of The Art Review provides a comprehensive current summary of systemic lupus erythematosus based on recent literature. In basic and translational science, this summary includes the current state of genetics, epigenetics, differences by ancestry, and updates about the molecular and immunological pathogenesis of systemic lupus erythematosus. In clinical science, the summary includes updates in diagnosis and classification, clinical features and subphenotypes, and current guidelines and strategies for treatment. The paper also provides a comprehensive review of the large number of recent clinical trials in systemic lupus erythematosus. Current knowns and unknowns are presented, and potential directions for the future are suggested. Improved knowledge of immunological pathogenesis and the molecular differences that exist between patients should help to personalize treatment, minimize side effects, and achieve better outcomes in this difficult disease.
Collapse
Affiliation(s)
- Eric F Morand
- School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
- Department of Rheumatology, Monash Health, Melbourne, VIC, Australia
| | | | | | | |
Collapse
|
55
|
Zaver SA, Sarkar MK, Egolf S, Zou J, Tiwaa A, Capell BC, Gudjonsson JE, Simpson CL. Targeting SERCA2 in organotypic epidermis reveals MEK inhibition as a therapeutic strategy for Darier disease. JCI Insight 2023; 8:e170739. [PMID: 37561594 PMCID: PMC10561730 DOI: 10.1172/jci.insight.170739] [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: 03/21/2023] [Accepted: 08/08/2023] [Indexed: 08/12/2023] Open
Abstract
Mutation of the ATP2A2 gene encoding sarco-endoplasmic reticulum calcium ATPase 2 (SERCA2) was linked to Darier disease more than 2 decades ago; however, there remain no targeted therapies for this disorder causing recurrent skin blistering and infections. Since Atp2a2-knockout mice do not phenocopy its pathology, we established a human tissue model of Darier disease to elucidate its pathogenesis and identify potential therapies. Leveraging CRISPR/Cas9, we generated human keratinocytes lacking SERCA2, which replicated features of Darier disease, including weakened intercellular adhesion and defective differentiation in organotypic epidermis. To identify pathogenic drivers downstream of SERCA2 depletion, we performed RNA sequencing and proteomics analysis. SERCA2-deficient keratinocytes lacked desmosomal and cytoskeletal proteins required for epidermal integrity and exhibited excess MAPK signaling, which modulates keratinocyte adhesion and differentiation. Immunostaining patient biopsies substantiated these findings, with lesions showing keratin deficiency, cadherin mislocalization, and ERK hyperphosphorylation. Dampening ERK activity with MEK inhibitors rescued adhesive protein expression and restored keratinocyte sheet integrity despite SERCA2 depletion or chemical inhibition. In sum, coupling multiomic analysis with human organotypic epidermis as a preclinical model, we found that SERCA2 haploinsufficiency disrupts critical adhesive components in keratinocytes via ERK signaling and identified MEK inhibition as a treatment strategy for Darier disease.
Collapse
Affiliation(s)
- Shivam A. Zaver
- Division of Dermatology, Department of Medicine, and
- Medical Scientist Training Program, University of Washington, Seattle, Washington, USA
| | - Mrinal K. Sarkar
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Shaun Egolf
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jonathan Zou
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Afua Tiwaa
- Division of Dermatology, Department of Medicine, and
| | - Brian C. Capell
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Cory L. Simpson
- Division of Dermatology, Department of Medicine, and
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
56
|
Londe AC, Fernandez-Ruiz R, Julio PR, Appenzeller S, Niewold TB. Type I Interferons in Autoimmunity: Implications in Clinical Phenotypes and Treatment Response. J Rheumatol 2023; 50:1103-1113. [PMID: 37399470 DOI: 10.3899/jrheum.2022-0827] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 07/05/2023]
Abstract
Type I interferon (IFN-I) is thought to play a role in many systemic autoimmune diseases. IFN-I pathway activation is associated with pathogenic features, including the presence of autoantibodies and clinical phenotypes such as more severe disease with increased disease activity and damage. We will review the role and potential drivers of IFN-I dysregulation in 5 prototypic autoimmune diseases: systemic lupus erythematosus, dermatomyositis, rheumatoid arthritis, primary Sjögren syndrome, and systemic sclerosis. We will also discuss current therapeutic strategies that directly or indirectly target the IFN-I system.
Collapse
Affiliation(s)
- Ana Carolina Londe
- A.C. Londe, MSc, Autoimmunity Lab, and Graduate Program in Physiopathology, School of Medical Science, State University of Campinas, Campinas, São Paulo, Brazil
| | - Ruth Fernandez-Ruiz
- R. Fernandez-Ruiz, MD, Department of Medicine, Hospital for Special Surgery, New York, New York, USA
| | - Paulo Rogério Julio
- P. Rogério Julio, MSc, Autoimmunity Lab, and Graduate Program of Child and Adolescent Health, School of Medical Science, State University of Campinas, Campinas, São Paulo, Brazil
| | - Simone Appenzeller
- S. Appenzeller, MD, PhD, Autoimmunity Lab, and Rheumatology Unit, Department of Medicine, School of Medical Science, State University of Campinas, Campinas, São Paulo, Brazil
| | - Timothy B Niewold
- T.B. Niewold, MD, Department of Medicine, Hospital for Special Surgery, New York, New York, USA.
| |
Collapse
|
57
|
Lu D, Zhu X, Hong T, Yao X, Xie Z, Chen L, Wang Y, Zhang K, Ren Y, Cao Y, Wang X. Serum Metabolomics Analysis of Skin-Involved Systemic Lupus Erythematosus: Association of Anti-SSA Antibodies with Photosensitivity. J Inflamm Res 2023; 16:3811-3822. [PMID: 37667802 PMCID: PMC10475307 DOI: 10.2147/jir.s426337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/23/2023] [Indexed: 09/06/2023] Open
Abstract
Purpose Systemic lupus erythematosus is a heterogeneous autoimmune disease in which skin involvement is a common manifestation. It is currently thought that the photosensitivity of SLE skin involvement is associated with anti-SSA antibodies. This study aimed to expand the current state of knowledge surrounding the molecular pathophysiology of SLE skin photosensitivity through Serum metabolomics analysis. Patients and Methods The serum metabolites of 23 cases of skin-involved SLE (SI) group, 14 cases of no SI (NSI) group, and 30 cases of healthy controls (HC) were analyzed by using UPLC-MS/MS technology, and subgroup analysis was performed according to the expression of anti-SSA antibodies in SI. MetaboAnalyst 5.0 was used for enrichment analysis and ROC curve construction, identifying serum metabolic markers of skin-involved SLE associated with anti-SSA antibodies. Results We identified several metabolites and metabolic pathways associated with SLE photosensitivity. Two metabolites, SM (d18:1/24:0) and gamma-CEHC can distinguish between anti-SSA antibody-positive and negative SI, with AUC of 0.829 and 0.806. These two photosensitization-related substances may be potential markers of skin involvement in SLE associated with anti-SSA antibody. Conclusion This study provides new insights into the pathogenesis of SI patients, and provides a new molecular biological basis for the association between anti-SSA antibodies and skin photoallergic manifestations of SLE.
Collapse
Affiliation(s)
- Dingqi Lu
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Xinchao Zhu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Tao Hong
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Xinyi Yao
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Zhiming Xie
- The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Liying Chen
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Yihan Wang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Kaiyuan Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Yating Ren
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Yi Cao
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| | - Xinchang Wang
- The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310053, People’s Republic of China
| |
Collapse
|
58
|
Tanaka Y, Kusuda M, Yamaguchi Y. Interferons and systemic lupus erythematosus: Pathogenesis, clinical features, and treatments in interferon-driven disease. Mod Rheumatol 2023; 33:857-867. [PMID: 36440704 DOI: 10.1093/mr/roac140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/24/2022] [Accepted: 11/09/2022] [Indexed: 08/27/2023]
Abstract
Type I interferons (IFNs) have recently received a lot of attention with the elucidation of the pathogenesis of systemic lupus erythematosus (SLE). Type I IFNs are associated with many SLE symptoms and play a role in the pathogenesis of autoimmune diseases that may occur concurrently with SLE, such as Sjögren's syndrome, antiphospholipid syndrome, myositis, scleroderma, and interferonopathy. Type I IFNs could be the link between these diseases. However, direct measurement of type I IFN levels and the IFN gene signature is currently unavailable in clinical practice. This review discusses type I IFN signalling in SLE, investigates the role of type I IFN in the clinical manifestations and symptoms associated with SLE and other IFN-related diseases, and discusses the clinical tests that can be used to diagnose SLE and measure disease activity. In addition, the role of type I IFN-blocking therapies as potential treatments for SLE is discussed.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | | | | |
Collapse
|
59
|
Hile GA, Coit P, Xu B, Victory AM, Gharaee-Kermani M, Estadt SN, Maz MP, Martens JWS, Wasikowski R, Dobry C, Tsoi LC, Iglesias-Bartolome R, Berthier CC, Billi AC, Gudjonsson JE, Sawalha AH, Kahlenberg JM. Regulation of Photosensitivity by the Hippo Pathway in Lupus Skin. Arthritis Rheumatol 2023; 75:1216-1228. [PMID: 36704840 PMCID: PMC10313771 DOI: 10.1002/art.42460] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/05/2022] [Accepted: 01/24/2023] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Photosensitivity is one of the most common manifestations of systemic lupus erythematosus (SLE), yet its pathogenesis is not well understood. The normal-appearing epidermis of patients with SLE exhibits increased ultraviolet B (UVB)-driven cell death that persists in cell culture. Here, we investigated the role of epigenetic modification and Hippo signaling in enhanced UVB-induced apoptosis seen in SLE keratinocytes. METHODS We analyzed DNA methylation in cultured keratinocytes from SLE patients compared to keratinocytes from healthy controls (n = 6/group). Protein expression was validated in cultured keratinocytes using immunoblotting and immunofluorescence. An immortalized keratinocyte line overexpressing WWC1 was generated via lentiviral vector. WWC1-driven changes were inhibited using a large tumor suppressor kinase 1/2 (LATS1/2) inhibitor (TRULI) and small interfering RNA (siRNA). The interaction between the Yes-associated protein (YAP) and the transcriptional enhancer associate domain (TEAD) was inhibited by overexpression of an N/TERT cell line expressing a tetracycline-inducible green fluorescent protein-tagged protein that inhibits YAP-TEAD binding (TEADi). Apoptosis was assessed using cleaved caspase 3/7 and TUNEL staining. RESULTS Hippo signaling was the top differentially methylated pathway in SLE versus control keratinocytes. SLE keratinocytes (n = 6) showed significant hypomethylation (Δβ = -0.153) and thus overexpression of the Hippo regulator WWC1 (P = 0.002). WWC1 overexpression increased LATS1/2 kinase activation, leading to YAP cytoplasmic retention and altered proapoptotic transcription in SLE keratinocytes. Accordingly, UVB-mediated apoptosis in keratinocytes could be enhanced by WWC1 overexpression or YAP-TEAD inhibition, mimicking SLE keratinocytes. Importantly, inhibition of LATS1/2 with either the chemical inhibitor TRULI or siRNA effectively eliminated enhanced UVB-apoptosis in SLE keratinocytes. CONCLUSION Our work unravels a novel driver of photosensitivity in SLE: overactive Hippo signaling in SLE keratinocytes restricts YAP transcriptional activity, leading to shifts that promote UVB apoptosis.
Collapse
Affiliation(s)
- Grace A. Hile
- Department of Dermatology, University of Michigan; Ann Arbor, USA
| | - Patrick Coit
- Division of Rheumatology, University of Michigan; Ann Arbor, USA
- Graduate Program in Immunology, University of Michigan; Ann Arbor, USA
- Departments of Pediatrics, Medicine, and Immunology, and Lupus Center of Excellence, University of Pittsburgh; Pittsburgh, USA
| | - Bin Xu
- Division of Rheumatology, University of Michigan; Ann Arbor, USA
| | | | - Mehrnaz Gharaee-Kermani
- Department of Dermatology, University of Michigan; Ann Arbor, USA
- Division of Rheumatology, University of Michigan; Ann Arbor, USA
| | - Shannon N. Estadt
- Graduate Program in Immunology, University of Michigan; Ann Arbor, USA
| | - Mitra P. Maz
- Graduate Program in Immunology, University of Michigan; Ann Arbor, USA
| | | | - Rachael Wasikowski
- Department of Dermatology, University of Michigan; Ann Arbor, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan; Ann Arbor, USA
| | - Craig Dobry
- Department of Dermatology, University of Michigan; Ann Arbor, USA
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan; Ann Arbor, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan; Ann Arbor, USA
- Department of Biostatistics, University of Michigan; Ann Arbor, MI 48109, USA
| | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health; Bethesda, USA
| | - Celine C. Berthier
- Department of Computational Medicine & Bioinformatics, University of Michigan; Ann Arbor, USA
- Department of Biostatistics, University of Michigan; Ann Arbor, MI 48109, USA
| | - Allison C. Billi
- Department of Dermatology, University of Michigan; Ann Arbor, USA
| | | | - Amr H. Sawalha
- Departments of Pediatrics, Medicine, and Immunology, and Lupus Center of Excellence, University of Pittsburgh; Pittsburgh, USA
| | - J. Michelle Kahlenberg
- Department of Dermatology, University of Michigan; Ann Arbor, USA
- Division of Rheumatology, University of Michigan; Ann Arbor, USA
| |
Collapse
|
60
|
Lu X, Jin H. A Review of CRISPR-Based Advances in Dermatological Diseases. Mol Diagn Ther 2023; 27:445-456. [PMID: 37041404 DOI: 10.1007/s40291-023-00642-5] [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: 02/07/2023] [Indexed: 04/13/2023]
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR) has revolutionized biomedical research by offering novel approaches to genetic and epigenetic manipulation. In dermatology, it has significantly promoted our understanding of complex diseases, and shown great potential in therapeutic applications. In this review, we introduce the adoption of CRISPR technology as a tool to study different types of skin disorders, including monogenic genodermatoses, inflammatory disorders, and cutaneous infections. We highlight the promising preclinical results of CRISPR-mediated treatment and important mechanic discoveries in investigative studies. Future opportunities and remaining challenges are also discussed. We predict that CRISPR will be more extensively used for dermatological research and even be accessible to patients in the future.
Collapse
Affiliation(s)
- Xinyi Lu
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, 100730, China
| | - Hongzhong Jin
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, 100730, China.
| |
Collapse
|
61
|
Niebel D, de Vos L, Fetter T, Brägelmann C, Wenzel J. Cutaneous Lupus Erythematosus: An Update on Pathogenesis and Future Therapeutic Directions. Am J Clin Dermatol 2023; 24:521-540. [PMID: 37140884 PMCID: PMC10157137 DOI: 10.1007/s40257-023-00774-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 05/05/2023]
Abstract
Lupus erythematosus comprises a spectrum of autoimmune diseases that may affect various organs (systemic lupus erythematosus [SLE]) or the skin only (cutaneous lupus erythematosus [CLE]). Typical combinations of clinical, histological and serological findings define clinical subtypes of CLE, yet there is high interindividual variation. Skin lesions arise in the course of triggers such as ultraviolet (UV) light exposure, smoking or drugs; keratinocytes, cytotoxic T cells and plasmacytoid dendritic cells (pDCs) establish a self-perpetuating interplay between the innate and adaptive immune system that is pivotal for the pathogenesis of CLE. Therefore, treatment relies on avoidance of triggers and UV protection, topical therapies (glucocorticosteroids, calcineurin inhibitors) and rather unspecific immunosuppressive or immunomodulatory drugs. Yet, the advent of licensed targeted therapies for SLE might also open new perspectives in the management of CLE. The heterogeneity of CLE might be attributable to individual variables and we speculate that the prevailing inflammatory signature defined by either T cells, B cells, pDCs, a strong lesional type I interferon (IFN) response, or combinations of the above might be suitable to predict therapeutic response to targeted treatment. Therefore, pretherapeutic histological assessment of the inflammatory infiltrate could stratify patients with refractory CLE for T-cell-directed therapies (e.g. dapirolizumab pegol), B-cell-directed therapies (e.g. belimumab), pDC-directed therapies (e.g. litifilimab) or IFN-directed therapies (e.g. anifrolumab). Moreover, Janus kinase (JAK) and spleen tyrosine kinase (SYK) inhibitors might broaden the therapeutic armamentarium in the near future. A close interdisciplinary exchange with rheumatologists and nephrologists is mandatory for optimal treatment of lupus patients to define the best therapeutic strategy.
Collapse
Affiliation(s)
- Dennis Niebel
- Department of Dermatology, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Luka de Vos
- Department of Dermatology, University Hospital Bonn, 53127, Bonn, Germany
| | - Tanja Fetter
- Department of Dermatology, University Hospital Bonn, 53127, Bonn, Germany
| | | | - Jörg Wenzel
- Department of Dermatology, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
62
|
Moneta GM, Bracaglia C, Caiello I, Farroni C, Pires Marafon D, Carlomagno R, Hiraki L, Vivarelli M, Gianviti A, Carbogno S, Ferlin W, de Min C, Silverman E, Carsetti R, De Benedetti F, Marasco E. Persistently active interferon-γ pathway and expansion of T-bet + B cells in a subset of patients with childhood-onset systemic lupus erythematosus. Eur J Immunol 2023; 53:e2250319. [PMID: 37204055 DOI: 10.1002/eji.202250319] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/14/2023] [Accepted: 05/04/2023] [Indexed: 05/20/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease causing significant morbidity and mortality, despite important improvements in its management in the last decades. The objective of this work is to investigate the role of IFN-γ in the pathogenesis of childhood-onset systemic lupus erythematosus (cSLE), evaluating the crosstalk between IFN-α and IFN-γ and the expression of T-bet, a transcription factor induced by IFN-γ, in B cells of patients with cSLE. Expression levels of both IFN-α and IFN-γ-induced genes were upregulated in patients with cSLE. We found increased serum levels of CXCL9 and CXCL10 in patients with cSLE. Type I IFN score decreased with initiation of immunosuppressive treatment; conversely, type II IFN score and levels of CXCL9 were not significantly affected by immunosuppressive treatment. Type II IFN score and CXCL9 were significantly higher in patients with lupus nephritis. We observed the expansion of a population of naïve B cells expressing T-bet in a cluster of patients with cSLE. IFN-γ, but not IFN-α, induced the expression of T-bet in B cells. Our data suggest that IFN-γ is hyperactive in cSLE, especially in patients with lupus nephritis, and it is not modulated by therapy. Our data reinforce the potential of IFN-γ as a therapeutic target in SLE.
Collapse
Affiliation(s)
- Gian Marco Moneta
- Division of Rheumatology, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Claudia Bracaglia
- Division of Rheumatology, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Ivan Caiello
- Division of Rheumatology, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Chiara Farroni
- B Cell Physiopathology Unit, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | | | - Linda Hiraki
- Division of Rheumatology, SickKids Hospital, Toronto, Canada
| | - Marina Vivarelli
- Division of Nephrology and Dialysis, Department of Pediatric Subspecialties, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Alessandra Gianviti
- Division of Nephrology and Dialysis, Department of Pediatric Subspecialties, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Simone Carbogno
- Division of Rheumatology, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| | - Walter Ferlin
- Light Chain Bioscience - Novimmune SA, Plan-Les-Ouates Geneva, Switzerland
| | | | - Earl Silverman
- Division of Rheumatology, SickKids Hospital, Toronto, Canada
| | - Rita Carsetti
- B Cell Physiopathology Unit, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | - Emiliano Marasco
- Division of Rheumatology, Ospedale Pediatrico Bambino Gesù IRCCS, Rome, Italy
| |
Collapse
|
63
|
Kowalski EH, Stolarczyk A, Richardson CT. Successful treatment of severe chronic cutaneous lupus with anifrolumab: A series of 6 cases. JAAD Case Rep 2023; 37:21-29. [PMID: 37324181 PMCID: PMC10265470 DOI: 10.1016/j.jdcr.2023.04.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023] Open
Affiliation(s)
- Eric H. Kowalski
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York
| | - Ania Stolarczyk
- University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Christopher T. Richardson
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
64
|
Nikolopoulos D, Parodis I. Janus kinase inhibitors in systemic lupus erythematosus: implications for tyrosine kinase 2 inhibition. Front Med (Lausanne) 2023; 10:1217147. [PMID: 37457579 PMCID: PMC10344364 DOI: 10.3389/fmed.2023.1217147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Aberrant activation of the Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathway is common in systemic lupus erythematosus (SLE), conferring immune-mediated properties in target tissues. Multiple cytokines activate different combinations of JAKs and STATs to alter the cell fate of target tissue and induce end-organ damage. Thus, the simultaneous blockade of several different cytokines by small molecules acting downstream intracellular signalling has gained traction. JAK inhibitors have been approved for the treatment of several rheumatic diseases, yet hitherto not for SLE. Nevertheless, JAK inhibitors including tofacitinib, baricitinib, and deucravacitinib have shown merit as treatments for SLE. Tofacitinib, a JAK1/3 inhibitor, reduced cholesterol levels, improved vascular function, and decreased the type I interferon signature in SLE patients. Baricitinib, a JAK1/2 inhibitor, demonstrated significant improvements in lupus rashes and arthritis in a phase 2 and a phase 3 randomised controlled trial, but the results were not replicated in another phase 3 trial. Deucravacitinib, a selective tyrosine kinase 2 (TYK2) inhibitor, yielded greater response rates than placebo in a phase 2 trial of SLE and will be investigated in larger phase 3 trials. TYK2 is activated in response to cytokines actively involved in lupus pathogenesis; this review highlights the potential of targeting TYK2 as a promising therapy for SLE.
Collapse
Affiliation(s)
- Dionysis Nikolopoulos
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Medical Unit of Gastroenterology, Dermatology, and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Medical Unit of Gastroenterology, Dermatology, and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
65
|
Fagone P, Piombino E, Mangano K, De Pasquale R, Nicoletti F, Caltabiano R. Evaluation of the Involvement of Heme Oxygenase-1 Expression in Discoid Lupus Erythematosus Lesions. Antioxidants (Basel) 2023; 12:1352. [PMID: 37507892 PMCID: PMC10376595 DOI: 10.3390/antiox12071352] [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: 05/04/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Discoid lupus erythematosus (DLE) is a chronic autoimmune disease that primarily affects the skin, causing red, scaly patches that may be disfiguring and can cause permanent scarring. This study aimed to investigate the potential clinical and therapeutic applications of heme oxygenase-1 (HMOX1) in the context of DLE. Immunohistochemical staining and bioinformatics analysis were performed on skin biopsy samples from DLE patients to examine the levels of HMOX1 and to correlate with markers of inflammation. Our study revealed a negative correlation between HMOX1 levels and the inflammatory status of DLE lesions, as well as an inverse correlation between HMOX1 levels and the infiltration of M1 macrophages and activated mastocytes. These findings suggest that HMOX1 plays a crucial role in the regulation of inflammation in DLE and could be a potential therapeutic target and biomarker for DLE.
Collapse
Affiliation(s)
- Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Eliana Piombino
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, 95123 Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Rocco De Pasquale
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Rosario Caltabiano
- Department of Medical, Surgical and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, 95123 Catania, Italy
| |
Collapse
|
66
|
Ma F, Plazyo O, Billi AC, Tsoi LC, Xing X, Wasikowski R, Gharaee-Kermani M, Hile G, Jiang Y, Harms PW, Xing E, Kirma J, Xi J, Hsu JE, Sarkar MK, Chung Y, Di Domizio J, Gilliet M, Ward NL, Maverakis E, Klechevsky E, Voorhees JJ, Elder JT, Lee JH, Kahlenberg JM, Pellegrini M, Modlin RL, Gudjonsson JE. Single cell and spatial sequencing define processes by which keratinocytes and fibroblasts amplify inflammatory responses in psoriasis. Nat Commun 2023; 14:3455. [PMID: 37308489 PMCID: PMC10261041 DOI: 10.1038/s41467-023-39020-4] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/26/2023] [Indexed: 06/14/2023] Open
Abstract
The immunopathogenesis of psoriasis, a common chronic inflammatory disease of the skin, is incompletely understood. Here we demonstrate, using a combination of single cell and spatial RNA sequencing, IL-36 dependent amplification of IL-17A and TNF inflammatory responses in the absence of neutrophil proteases, which primarily occur within the supraspinous layer of the psoriatic epidermis. We further show that a subset of SFRP2+ fibroblasts in psoriasis contribute to amplification of the immune network through transition to a pro-inflammatory state. The SFRP2+ fibroblast communication network involves production of CCL13, CCL19 and CXCL12, connected by ligand-receptor interactions to other spatially proximate cell types: CCR2+ myeloid cells, CCR7+ LAMP3+ dendritic cells, and CXCR4 expressed on both CD8+ Tc17 cells and keratinocytes, respectively. The SFRP2+ fibroblasts also express cathepsin S, further amplifying inflammatory responses by activating IL-36G in keratinocytes. These data provide an in-depth view of psoriasis pathogenesis, which expands our understanding of the critical cellular participants to include inflammatory fibroblasts and their cellular interactions.
Collapse
Affiliation(s)
- Feiyang Ma
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Olesya Plazyo
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Allison C Billi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xianying Xing
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Rachael Wasikowski
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Grace Hile
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yanyun Jiang
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Paul W Harms
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Enze Xing
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joseph Kirma
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jingyue Xi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Jer-En Hsu
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Mrinal K Sarkar
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yutein Chung
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jeremy Di Domizio
- Department of Dermatology, University Hospital of Lausanne, 1011, Lausanne, Switzerland
| | - Michel Gilliet
- Department of Dermatology, University Hospital of Lausanne, 1011, Lausanne, Switzerland
| | - Nicole L Ward
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Emanual Maverakis
- Department of Dermatology, University of California Davis, Sacramento, CA, USA
| | - Eynav Klechevsky
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - John J Voorhees
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - James T Elder
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
- Ann Arbor Veterans Affairs Medical Center, Ann Arbor, MI, 48105, USA
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - J Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Johann E Gudjonsson
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
67
|
Sarkar MK, Uppala R, Zeng C, Billi AC, Tsoi LC, Kidder A, Xing X, Perez White BE, Shao S, Plazyo O, Sirobhushanam S, Xing E, Jiang Y, Gallagher KA, Voorhees JJ, Kahlenberg JM, Gudjonsson JE. Keratinocytes sense and eliminate CRISPR DNA through STING/IFN-κ activation and APOBEC3G induction. J Clin Invest 2023; 133:e159393. [PMID: 36928117 PMCID: PMC10145927 DOI: 10.1172/jci159393] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
CRISPR/Cas9 has been proposed as a treatment for genetically inherited skin disorders. Here we report that CRISPR transfection activates STING-dependent antiviral responses in keratinocytes, resulting in heightened endogenous interferon (IFN) responses through induction of IFN-κ, leading to decreased plasmid stability secondary to induction of the cytidine deaminase gene APOBEC3G. Notably, CRISPR-generated KO keratinocytes had permanent suppression of IFN-κ and IFN-stimulated gene (ISG) expression, secondary to hypermethylation of the IFNK promoter region by the DNA methyltransferase DNMT3B. JAK inhibition via baricitinib prior to CRISPR transfection increased transfection efficiency, prevented IFNK promoter hypermethylation, and restored normal IFN-κ activity and ISG responses. This work shows that CRISPR-mediated gene correction alters antiviral responses in keratinocytes, has implications for future gene therapies for inherited skin diseases using CRISPR technology, and suggests pharmacologic JAK inhibition as a tool for facilitating and attenuating inadvertent selection effects in CRISPR/Cas9 therapeutic approaches.
Collapse
Affiliation(s)
| | - Ranjitha Uppala
- Department of Dermatology, and
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | - Shuai Shao
- Department of Dermatology, and
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shannxi, China
| | | | - Sirisha Sirobhushanam
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Yanyun Jiang
- Department of Dermatology, and
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Katherine A. Gallagher
- Section of Vascular Surgery, Department of Surgery
- Department of Microbiology and Immunology, and
| | | | - J. Michelle Kahlenberg
- Department of Dermatology, and
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, Michigan, USA
- Taubman Medical Research Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Johann E. Gudjonsson
- Department of Dermatology, and
- Taubman Medical Research Institute, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
68
|
Neale RE, Lucas RM, Byrne SN, Hollestein L, Rhodes LE, Yazar S, Young AR, Berwick M, Ireland RA, Olsen CM. The effects of exposure to solar radiation on human health. Photochem Photobiol Sci 2023; 22:1011-1047. [PMID: 36856971 PMCID: PMC9976694 DOI: 10.1007/s43630-023-00375-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 03/02/2023]
Abstract
This assessment by the Environmental Effects Assessment Panel (EEAP) of the Montreal Protocol under the United Nations Environment Programme (UNEP) evaluates the effects of ultraviolet (UV) radiation on human health within the context of the Montreal Protocol and its Amendments. We assess work published since our last comprehensive assessment in 2018. Over the last four years gains have been made in knowledge of the links between sun exposure and health outcomes, mechanisms, and estimates of disease burden, including economic impacts. Of particular note, there is new information about the way in which exposure to UV radiation modulates the immune system, causing both harms and benefits for health. The burden of skin cancer remains high, with many lives lost to melanoma and many more people treated for keratinocyte cancer, but it has been estimated that the Montreal Protocol will prevent 11 million cases of melanoma and 432 million cases of keratinocyte cancer that would otherwise have occurred in the United States in people born between 1890 and 2100. While the incidence of skin cancer continues to rise, rates have stabilised in younger populations in some countries. Mortality has also plateaued, partly due to the use of systemic therapies for advanced disease. However, these therapies are very expensive, contributing to the extremely high economic burden of skin cancer, and emphasising the importance and comparative cost-effectiveness of prevention. Photodermatoses, inflammatory skin conditions induced by exposure to UV radiation, can have a marked detrimental impact on the quality of life of sufferers. More information is emerging about their potential link with commonly used drugs, particularly anti-hypertensives. The eyes are also harmed by over-exposure to UV radiation. The incidence of cataract and pterygium is continuing to rise, and there is now evidence of a link between intraocular melanoma and sun exposure. It has been estimated that the Montreal Protocol will prevent 63 million cases of cataract that would otherwise have occurred in the United States in people born between 1890 and 2100. Despite the clearly established harms, exposure to UV radiation also has benefits for human health. While the best recognised benefit is production of vitamin D, beneficial effects mediated by factors other than vitamin D are emerging. For both sun exposure and vitamin D, there is increasingly convincing evidence of a positive role in diseases related to immune function, including both autoimmune diseases and infection. With its influence on the intensity of UV radiation and global warming, the Montreal Protocol has, and will have, both direct and indirect effects on human health, potentially changing the balance of the risks and benefits of spending time outdoors.
Collapse
Affiliation(s)
- R E Neale
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- School of Public Health, University of Queensland, Brisbane, QLD, Australia.
| | - R M Lucas
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT, Australia
| | - S N Byrne
- School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - L Hollestein
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - L E Rhodes
- Dermatology Research Centre, School of Biological Sciences, University of Manchester, Salford Royal Hospital, Northern Care Alliance NHS Trust, Manchester, UK
| | - S Yazar
- Garvan Medical Research Institute, Sydney, NSW, Australia
| | | | - M Berwick
- University of New Mexico Comprehensive Cancer Center, Albuquerque, USA
| | - R A Ireland
- School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - C M Olsen
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Frazer Institute, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
69
|
Athanassiou L, Kostoglou-Athanassiou I, Koutsilieris M, Shoenfeld Y. Vitamin D and Autoimmune Rheumatic Diseases. Biomolecules 2023; 13:709. [PMID: 37189455 PMCID: PMC10135889 DOI: 10.3390/biom13040709] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/12/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Vitamin D is a steroid hormone with potent immune-modulating properties. It has been shown to stimulate innate immunity and induce immune tolerance. Extensive research efforts have shown that vitamin D deficiency may be related to the development of autoimmune diseases. Vitamin D deficiency has been observed in patients with rheumatoid arthritis (RA) and has been shown to be inversely related to disease activity. Moreover, vitamin D deficiency may be implicated in the pathogenesis of the disease. Vitamin D deficiency has also been observed in patients with systemic lupus erythematosus (SLE). It has been found to be inversely related to disease activity and renal involvement. In addition, vitamin D receptor polymorphisms have been studied in SLE. Vitamin D levels have been studied in patients with Sjogren's syndrome, and vitamin D deficiency may be related to neuropathy and the development of lymphoma in the context of Sjogren's syndrome. Vitamin D deficiency has been observed in ankylosing spondylitis, psoriatic arthritis (PsA), and idiopathic inflammatory myopathies. Vitamin D deficiency has also been observed in systemic sclerosis. Vitamin D deficiency may be implicated in the pathogenesis of autoimmunity, and it may be administered to prevent autoimmune disease and reduce pain in the context of autoimmune rheumatic disorders.
Collapse
Affiliation(s)
- Lambros Athanassiou
- Department of Rheumatology, Asclepeion Hospital, Voula, GR16673 Athens, Greece
- Department of Physiology, Medical School, University of Athens, GR11527 Athens, Greece
| | | | - Michael Koutsilieris
- Department of Physiology, Medical School, University of Athens, GR11527 Athens, Greece
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
70
|
Lilja S, Li X, Smelik M, Lee EJ, Loscalzo J, Marthanda PB, Hu L, Magnusson M, Sysoev O, Zhang H, Zhao Y, Sjöwall C, Gawel D, Wang H, Benson M. Multi-organ single-cell analysis reveals an on/off switch system with potential for personalized treatment of immunological diseases. Cell Rep Med 2023; 4:100956. [PMID: 36858042 PMCID: PMC10040389 DOI: 10.1016/j.xcrm.2023.100956] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/30/2022] [Accepted: 02/03/2023] [Indexed: 03/03/2023]
Abstract
Prioritization of disease mechanisms, biomarkers, and drug targets in immune-mediated inflammatory diseases (IMIDs) is complicated by altered interactions between thousands of genes. Our multi-organ single-cell RNA sequencing of a mouse IMID model, namely collagen-induced arthritis, shows highly complex and heterogeneous expression changes in all analyzed organs, even though only joints showed signs of inflammation. We organized those into a multi-organ multicellular disease model, which shows predicted molecular interactions within and between organs. That model supports that inflammation is switched on or off by altered balance between pro- and anti-inflammatory upstream regulators (URs) and downstream pathways. Meta-analyses of human IMIDs show a similar, but graded, on/off switch system. This system has the potential to prioritize, diagnose, and treat optimal combinations of URs on the levels of IMIDs, subgroups, and individual patients. That potential is supported by UR analyses in more than 600 sera from patients with systemic lupus erythematosus.
Collapse
Affiliation(s)
- Sandra Lilja
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Mavatar, Inc, Vasagatan, 11120 Stockholm, Sweden
| | - Xinxiu Li
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Martin Smelik
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Eun Jung Lee
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Ganwong 26460, Korea
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pratheek Bellur Marthanda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Lang Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Mattias Magnusson
- The National Board of Health and Welfare, Socialstyrelsen, 11259 Stockholm, Sweden
| | - Oleg Sysoev
- Department of Computer and Information Science, Linköping University, 58183 Linköping, Sweden
| | - Huan Zhang
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden
| | - Yelin Zhao
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Christopher Sjöwall
- Biomedical and Clinical Sciences, Division of Inflammation and Infection/Rheumatology, Linköping University, 58183 Linköping, Sweden
| | - Danuta Gawel
- Mavatar, Inc, Vasagatan, 11120 Stockholm, Sweden
| | - Hui Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Mikael Benson
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden.
| |
Collapse
|
71
|
Zaver SA, Sarkar MK, Egolf S, Zou J, Tiwaa A, Capell BC, Gudjonsson JE, Simpson CL. Targeting SERCA2 in organotypic epidermis reveals MEK inhibition as a therapeutic strategy for Darier disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531620. [PMID: 36945477 PMCID: PMC10028894 DOI: 10.1101/2023.03.07.531620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Mutation of the ATP2A2 gene encoding sarco-endoplasmic reticulum calcium ATPase 2 (SERCA2) was linked to Darier disease more than two decades ago; however, there remain no targeted therapies for this disorder causing recurrent skin blistering and infections. Since Atp2a2 knockout mice do not phenocopy its pathology, we established a human tissue model of Darier disease to elucidate its pathogenesis and identify potential therapies. Leveraging CRISPR/Cas9, we generated human keratinocytes lacking SERCA2, which replicated features of Darier disease, including weakened intercellular adhesion and defective differentiation in organotypic epidermis. To identify pathogenic drivers downstream of SERCA2 depletion, we performed RNA sequencing and proteomic analysis. SERCA2-deficient keratinocytes lacked desmosomal and cytoskeletal proteins required for epidermal integrity and exhibited excess MAP kinase signaling, which modulates keratinocyte adhesion and differentiation. Immunostaining patient biopsies substantiated these findings with lesions showing keratin deficiency, cadherin mis-localization, and ERK hyper-phosphorylation. Dampening ERK activity with MEK inhibitors rescued adhesive protein expression and restored keratinocyte sheet integrity despite SERCA2 depletion or chemical inhibition. In sum, coupling multi-omic analysis with human organotypic epidermis as a pre-clinical model, we found that SERCA2 haploinsufficiency disrupts critical adhesive components in keratinocytes via ERK signaling and identified MEK inhibition as a treatment strategy for Darier disease.
Collapse
|
72
|
Corbin D, Christian L, Rapp CM, Liu L, Rohan CA, Travers JB. New concepts on abnormal UV reactions in systemic lupus erythematosus and a screening tool for assessment of photosensitivity. Skin Res Technol 2023; 29:e13247. [PMID: 36973991 PMCID: PMC10059080 DOI: 10.1111/srt.13247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 11/11/2022] [Indexed: 03/09/2023]
Affiliation(s)
- Danielle Corbin
- Department of Pharmacology & ToxicologyBoonshoft School of Medicine at Wright State UniversityDaytonOhioUSA
| | - Lea Christian
- Department of Pharmacology & ToxicologyBoonshoft School of Medicine at Wright State UniversityDaytonOhioUSA
| | - Christine M. Rapp
- Department of Pharmacology & ToxicologyBoonshoft School of Medicine at Wright State UniversityDaytonOhioUSA
| | - Langni Liu
- Department of Pharmacology & ToxicologyBoonshoft School of Medicine at Wright State UniversityDaytonOhioUSA
| | - Craig A. Rohan
- Department of Pharmacology & ToxicologyBoonshoft School of Medicine at Wright State UniversityDaytonOhioUSA
- Department of DermatologyBoonshoft School of Medicine at Wright State UniversityDaytonOhioUSA
- Department of Medicine (Dermatology)Dayton Veterans Administration Medical CenterDaytonOhioUSA
| | - Jeffrey B. Travers
- Department of Pharmacology & ToxicologyBoonshoft School of Medicine at Wright State UniversityDaytonOhioUSA
- Department of DermatologyBoonshoft School of Medicine at Wright State UniversityDaytonOhioUSA
- Department of Medicine (Dermatology)Dayton Veterans Administration Medical CenterDaytonOhioUSA
| |
Collapse
|
73
|
Bai W, Yang F, Xu H, Wei W, Li H, Zhang L, Zhao Y, Shi X, Zhang Y, Zeng X, Leng X. A multi-center, open-label, randomized study to explore efficacy and safety of baricitinib in active primary Sjogren's syndrome patients. Trials 2023; 24:112. [PMID: 36793118 PMCID: PMC9930286 DOI: 10.1186/s13063-023-07087-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 01/10/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Primary Sjogren's syndrome (pSS) is a systemic autoimmune disease involving multiple organ systems. The Janus kinase/signal transduction and activator of transcription (JAK/STAT) signaling pathway is a key pathway involving the pathogenesis of pSS. Baricitinib, a selective JAK1 and JAK2 inhibitor, has been approved for treatment of active rheumatoid arthritis and reported in treatment of some other autoimmune diseases including systemic lupus erythematosus. We have found that baricitinib might be effective and safe in pSS in a pilot study. However, there is no published clinical evidence of baricitinib in pSS. Hence, we conducted this randomized study to further explore the efficacy and safety of baricitinib in pSS. METHODS This is a multi-center, prospective, open-label, randomized study to compare the efficacy of baricitinib + hydroxychloroquine (HCQ) with HCQ alone in pSS patients. We plan to involve 87 active pSS patients with European League Against Rheumatism pSS disease activity index (ESSDAI) ≥ 5 from eight different tertiary centers in China. Patients will be randomized (2:1) to receive baricitinib 4 mg per day + HCQ 400 mg per day or HCQ 400 mg per day alone. We will switch HCQ to baricitinib + HCQ if the patient in the latter group has no ESSDAI response at week 12. The final evaluation will be at week 24. The primary endpoint is the percentage of ESSDAI response, or minimal clinically important improvement (MCII), which was defined as an improvement of ESSDAI at least three points at week 12. The secondary endpoints include EULAR pSS patient-reported index (ESSPRI) response, change of Physician's Global Assessment (PGA) score, serological activity parameters, salivary gland function test, and focus score on labial salivary gland biopsy. DISCUSSION This is the first randomized controlled study to evaluate the clinical efficacy and safety of baricitinib in pSS. We hope that the result of this study can provide more reliable evidence of the efficacy and safety of baricitinib in pSS. TRIAL REGISTRATION ClinicalTrials.gov NCT05016297. Registered on 19 Aug 2021.
Collapse
Affiliation(s)
- Wei Bai
- grid.506261.60000 0001 0706 7839Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China ,grid.424020.00000 0004 0369 1054National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, 100730 Beijing, China ,grid.413106.10000 0000 9889 6335State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, 100730 China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730 China
| | - Fan Yang
- grid.506261.60000 0001 0706 7839Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 China ,grid.424020.00000 0004 0369 1054National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, 100730 Beijing, China ,grid.413106.10000 0000 9889 6335State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, 100730 China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730 China
| | - Huji Xu
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, 200003 China
| | - Wei Wei
- grid.412645.00000 0004 1757 9434Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongbin Li
- grid.413375.70000 0004 1757 7666Department of Rheumatology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia China
| | - Liyun Zhang
- grid.470966.aDepartment of Rheumatology, Third Hospital of Shanxi Medical University, Bethune Hospital Shanxi Academy of Medical Sciences, Taiyuan, Shanxi China
| | - Yi Zhao
- grid.413259.80000 0004 0632 3337Department of Rheumatology and Allergy, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaofei Shi
- grid.453074.10000 0000 9797 0900Department of Rheumatology and Immunology, The First Affiliated Hospital and College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan China
| | - Yan Zhang
- grid.460007.50000 0004 1791 6584Department of Rheumatology and Immunology, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China. .,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, 100730, Beijing, China. .,State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, 100730, China. .,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Xiaomei Leng
- Department of Rheumatology and Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China. .,National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, 100730, Beijing, China. .,State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Beijing, 100730, China. .,Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| |
Collapse
|
74
|
Solé C, Domingo S, Penzo E, Moliné T, Porres L, Aparicio G, Ferrer B, Cortés-Hernández J. Downregulation of miR-885-5p Promotes NF-κB Pathway Activation and Immune Recruitment in Cutaneous Lupus Erythematosus. J Invest Dermatol 2023; 143:209-219.e13. [PMID: 36049539 DOI: 10.1016/j.jid.2022.08.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/21/2022] [Accepted: 08/01/2022] [Indexed: 01/25/2023]
Abstract
Cutaneous lupus erythematosus (CLE) has a specific microRNA expression profile. MiR-885-5p has been found to be downregulated in the epidermis of CLE lesions; however, its biological role in the disease has not been studied. In this study, we show that miR-885-5p is markedly reduced in CLE keratinocytes (KCs) with IFN-α and UVB being strong miR-885-5p regulators in vitro. Microarray expression profiling of anti‒miR-885-5p‒transfected KCs identified PSMB5 as a direct target. Specific inhibition of miR-885-5p increased epidermal proliferation by modulating keratin 16 gene K16, BIRC5, TP63, and CDK4 proliferative genes and promoted NF-κB signaling pathway in human primary KCs by increasing IκBα degradation. Silencing PSMB5 rescued the effect of miR-885-5p inhibition, indicating that miR-885-5p regulates proliferation and NF-κB activation by targeting PSMB5 in KCs. In addition, inhibition of miR-885-5p increased the ability of KCs to attract leukocytes in a PSMB5-independent manner. We identified TRAF1 as another direct target, and its silencing reduced leukocyte migration. Collectively, our findings suggest that UVB and IFN-ɑ downregulate miR-885-5p in CLE KCs, leading to epidermal inflammation by NF-κB activity enhancement and proliferation through PSMB5 and immune recruitment through TRAF1. Our data indicate that miR-885-5p is a potential therapeutic target in CLE.
Collapse
Affiliation(s)
- Cristina Solé
- Rheumatology Research Group - Lupus Unit, Vall d'Hebrón University Hospital, Vall d'Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.
| | - Sandra Domingo
- Rheumatology Research Group - Lupus Unit, Vall d'Hebrón University Hospital, Vall d'Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Eleonora Penzo
- Rheumatology Research Group - Lupus Unit, Vall d'Hebrón University Hospital, Vall d'Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Teresa Moliné
- Department of Pathology, Vall d'Hebrón University Hospital, Barcelona, Spain
| | - Laura Porres
- Rheumatology Research Group - Lupus Unit, Vall d'Hebrón University Hospital, Vall d'Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Gloria Aparicio
- Department of Dermatology, Vall d'Hebrón University Hospital, Barcelona, Spain
| | - Berta Ferrer
- Department of Pathology, Vall d'Hebrón University Hospital, Barcelona, Spain
| | - Josefina Cortés-Hernández
- Rheumatology Research Group - Lupus Unit, Vall d'Hebrón University Hospital, Vall d'Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
75
|
Zhou JY, Sarkar MK, Okamura K, Harris JE, Gudjonsson JE, Fitzgerald KA. Activation of the NLRP1 inflammasome in human keratinocytes by the dsDNA mimetic poly(dA:dT). Proc Natl Acad Sci U S A 2023; 120:e2213777120. [PMID: 36693106 PMCID: PMC9945980 DOI: 10.1073/pnas.2213777120] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The accrual of cytosolic DNA leads to transcription of type I IFNs, proteolytic maturation of the IL-1 family of cytokines, and pyroptotic cell death. Caspase-1 cleaves pro-IL1β to generate mature bioactive cytokine and gasdermin D which facilitates IL-1 release and pyroptotic cell death. Absent in melanoma-2 (AIM2) is a sensor of dsDNA leading to caspase-1 activation, although in human monocytes, cGAS-STING acting upstream of NLRP3 mediates the dsDNA-activated inflammasome response. In healthy human keratinocytes, AIM2 is not expressed yet caspase-1 is activated by the synthetic dsDNA mimetic poly(dA:dT). Here, we show that this response is not mediated by either AIM2 or the cGAS-STING-NLRP3 pathway and is instead dependent on NLRP1. Poly(dA:dT) is unique in its ability to activate NLRP1, as conventional linear dsDNAs fail to elicit NLRP1 activation. DsRNA was recently shown to activate NLRP1 and prior work has shown that poly(dA:dT) is transcribed into an RNA intermediate that stimulates the RNA sensor RIG-I. However, poly(dA:dT)-dependent RNA intermediates are insufficient to activate NLRP1. Instead, poly(dA:dT) results in oxidative nucleic acid damage and cellular stress, events which activate MAP3 kinases including ZAKα that converge on p38 to activate NLRP1. Collectively, this work defines a new activator of NLRP1, broadening our understanding of sensors that recognize poly(dA:dT) and advances the understanding of the immunostimulatory potential of this potent adjuvant.
Collapse
Affiliation(s)
- Jeffrey Y. Zhou
- aDivision of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA01605
| | - Mrinal K. Sarkar
- bDepartment of Dermatology, University of Michigan, Ann Arbor, MI48109
| | - Ken Okamura
- cDepartment of Dermatology, University of Massachusetts Chan Medical School, Worcester, MA01605
| | - John E. Harris
- cDepartment of Dermatology, University of Massachusetts Chan Medical School, Worcester, MA01605
| | | | - Katherine A. Fitzgerald
- aDivision of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA01605
- 1To whom correspondence may be addressed.
| |
Collapse
|
76
|
Miyagawa F. Current Knowledge of the Molecular Pathogenesis of Cutaneous Lupus Erythematosus. J Clin Med 2023; 12:987. [PMID: 36769633 PMCID: PMC9918007 DOI: 10.3390/jcm12030987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/01/2023] Open
Abstract
Cutaneous lupus erythematosus (CLE) is an autoimmune disease, which can be limited to the skin or associated with systemic lupus erythematosus (SLE). Gene expression analysis has revealed that both the innate and adaptive immune pathways are activated in CLE. Ultraviolet (UV) light, the predominant environmental factor associated with CLE, induces apoptosis in keratinocytes, and the endogenous nucleic acids released from the apoptotic cells are recognized via pattern recognition receptors, including Toll-like receptors. This leads to the production of type I interferon, a major contributor to the pathogenesis of CLE, by plasmacytoid dendritic cells. UV irradiation can also induce the externalization of autoantigens, such as SS-A/Ro, exposing them to circulating autoantibodies. T-helper 1 cells have been reported to play important roles in the adaptive immune response to CLE. Other environmental factors associated with CLE include drugs and cigarette smoke. Genetic factors also confer a predisposition to the development of CLE, and many susceptibility genes have been identified. Monogenetic forms of CLE also exist. This article aims to review current knowledge about the pathogenesis of CLE. A better understanding of the environmental, genetic, and immunoregulatory factors that drive CLE may provide important insights for the treatment of CLE.
Collapse
Affiliation(s)
- Fumi Miyagawa
- Department of Dermatology, Nara Medical University School of Medicine, 840 Shijo, Kashihara, Nara 634-8522, Japan
| |
Collapse
|
77
|
Kim ST, Muñoz-Grajales C, Dunn SE, Schneider R, Johnson SR, Touma Z, Ahmad Z, Bonilla D, Atenafu EG, Hiraki LT, Bookman A, Wither J. Interferon and interferon-induced cytokines as markers of impending clinical progression in ANA + individuals without a systemic autoimmune rheumatic disease diagnosis. Arthritis Res Ther 2023; 25:21. [PMID: 36765391 PMCID: PMC9912609 DOI: 10.1186/s13075-023-02997-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 01/26/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Elevated levels of interferons (IFNs) are a characteristic feature of systemic autoimmune rheumatic diseases (SARDs) and may be useful in predicting impending symptomatic progression in anti-nuclear antibody-positive (ANA+) individuals lacking a SARD diagnosis. Typically, these are measured by their effect on gene expression in the blood, which has limited their utility in clinical settings. Here, we assessed whether the measurement of serum IFN-α or selected IFN-induced cytokines accurately mirrors IFN-induced gene expression in ANA+ individuals and investigated their utility as biomarkers of clinical progression. METHODS A total of 280 subjects were studied, including 50 ANA- healthy controls, 160 ANA+ individuals without a SARD diagnosis (96 asymptomatic, 64 with undifferentiated connective tissue disease), and 70 SARD patients. IFN-induced gene expression was measured by nanoString and cytokine levels by ELISA or Simoa. ANA+ individuals lacking a SARD diagnosis who had the new onset of SARD criteria over the subsequent 2 years were defined as progressors. RESULTS Measurement of IFN-α levels by high-sensitivity ELISA or Simoa correlated much better with IFN-induced gene expression than measurement of CXCL-10 or Galectin-9 levels. Despite this, high CXCL-10 and Galectin-9 levels were better predictors of subsequent progression in ANA+ individuals than measures of IFN-α or IFN-induced gene expression with the optimal combination of predictive cytokines (CXCL-10 and IFN-α as measured by ELISA), resulting in a specificity and positive predictive value of 100%. CONCLUSION Easily performed ELISA assays for CXCL-10 and IFN-α can be used to predict ANA+ individuals at high risk of imminent symptomatic progression.
Collapse
Affiliation(s)
- Sonya T. Kim
- grid.231844.80000 0004 0474 0428Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 0S8 Canada
| | - Carolina Muñoz-Grajales
- grid.231844.80000 0004 0474 0428Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 0S8 Canada ,grid.17063.330000 0001 2157 2938Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Shannon E. Dunn
- grid.17063.330000 0001 2157 2938Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON Canada ,grid.415502.7Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada
| | - Raphael Schneider
- grid.415502.7Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada ,grid.415502.7Division of Neurology, St. Michael’s Hospital Unity Health, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Sindhu R. Johnson
- grid.17063.330000 0001 2157 2938Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON Canada ,Toronto Scleroderma Program, Division of Rheumatology, Toronto Western and Mount Sinai Hospitals, Toronto, ON Canada
| | - Zahi Touma
- grid.17063.330000 0001 2157 2938Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON Canada ,grid.231844.80000 0004 0474 0428University of Toronto Lupus Clinic, Centre for Prognosis Studies in Rheumatic Diseases, Schroeder Arthritis Institute, University Health Network, Toronto, ON Canada
| | - Zareen Ahmad
- grid.17063.330000 0001 2157 2938Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON Canada ,Toronto Scleroderma Program, Division of Rheumatology, Toronto Western and Mount Sinai Hospitals, Toronto, ON Canada
| | - Dennisse Bonilla
- grid.231844.80000 0004 0474 0428Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 0S8 Canada
| | - Eshetu G. Atenafu
- grid.231844.80000 0004 0474 0428Biostatistics Department, Princess Margaret Cancer Center, University Health Network, Toronto, Canada
| | - Linda T. Hiraki
- grid.17063.330000 0001 2157 2938Division of Rheumatology, The Hospital for Sick Children, and Department of Paediatrics, University of Toronto, Toronto, ON Canada
| | - Arthur Bookman
- grid.17063.330000 0001 2157 2938Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON Canada ,grid.231844.80000 0004 0474 0428Division of Rheumatology, Schroeder Arthritis Institute, University Health Network, Toronto, ON Canada
| | - Joan Wither
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada. .,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada. .,Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada. .,Division of Rheumatology, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
78
|
Zheng M, Hu Z, Mei X, Ouyang L, Song Y, Zhou W, Kong Y, Wu R, Rao S, Long H, Shi W, Jing H, Lu S, Wu H, Jia S, Lu Q, Zhao M. Single-cell sequencing shows cellular heterogeneity of cutaneous lesions in lupus erythematosus. Nat Commun 2022; 13:7489. [PMID: 36470882 PMCID: PMC9722937 DOI: 10.1038/s41467-022-35209-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Discoid lupus erythematosus (DLE) and systemic lupus erythematosus (SLE) are both types of lupus, yet the characteristics, and differences between them are not fully understood. Here we show single-cell RNA sequencing data of cutaneous lesions from DLE and SLE patients and skin tissues from healthy controls (HCs). We find significantly higher proportions of T cells, B cells and NK cells in DLE than in SLE. Expanded CCL20+ keratinocyte, CXCL1+ fibroblast, ISGhiCD4/CD8 T cell, ISGhi plasma cell, pDC, and NK subclusters are identified in DLE and SLE compared to HC. In addition, we observe higher cell communication scores between cell types such as fibroblasts and macrophage/dendritic cells in cutaneous lesions of DLE and SLE compared to HC. In summary, we clarify the heterogeneous characteristics in cutaneous lesions between DLE and SLE, and discover some specific cell subtypes and ligand-receptor pairs that indicate possible therapeutic targets of lupus erythematosus.
Collapse
Affiliation(s)
- Meiling Zheng
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Zhi Hu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Xiaole Mei
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China
| | - Lianlian Ouyang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Yang Song
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Wenhui Zhou
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Yi Kong
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Ruifang Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Shijia Rao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Hai Long
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Wei Shi
- Department of Dermatology, Xiangya Hospital, Central South University, 410008, Changsha, China
| | - Hui Jing
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Shuang Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China
| | - Sujie Jia
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, 410011, Changsha, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China.
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China.
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 210042, Nanjing, China.
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, 410011, Changsha, China.
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, 410011, Changsha, China.
| |
Collapse
|
79
|
de Vos L, Guel T, Niebel D, Bald S, ter Steege A, Bieber T, Wenzel J. Characterization of B cells in lupus erythematosus skin biopsies in the context of different immune cell infiltration patterns. Front Med (Lausanne) 2022; 9:1037408. [PMID: 36438026 PMCID: PMC9685332 DOI: 10.3389/fmed.2022.1037408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
Cutaneous lesions in lupus erythematosus (LE) subtypes are heterogenous. In line with the heterogeneity of the clinical presentation, the underlying lesional inflammation in LE skin samples is defined by different immune cell infiltrates. Pathophysiologically, lesional inflammation is driven by autoreactive cytotoxic T cells, targeting keratinocytes; plasmacytoid dendritic cells (pDCs), producing large amounts of interferon (IFN); and B cells, whose function in cutaneous LE is still unclear. This study aims to (a) classify inflammatory patterns with regard to the dominating cell type or cytokine expression and (b) investigating the specific role of B cells in LE skin lesions. Therefore, the immunohistological expression of inflammatory surrogates (CD20, CD123, MXA) in skin samples of n = 119 LE (subtypes: subacute cutaneous LE, chronic discoid LE, chilblain LE, LE tumidus, other LE) and n = 17 patients with inflammatory skin diseases (atopic dermatitis, psoriasis) were assessed. Samples were classified with regard to inflammatory groups. In addition multiplex-immunohistochemical analyses of n = 17 LE skin samples focusing on lesional B cells were conducted. In this study, we show that cutaneous lesions present with eight different inflammatory groups dominated by B cells, pDCs, a strong IFN expression, or overlapping patterns. Altogether, LE subtypes show heterogenous infiltration regardless of LE subtype, certain subtypes display a preference for infiltration groups. Furthermore, lesional B cells either form diffuse infiltrates or pseudofollicular structures, wherein they show antigen-presenting and T cell-activating properties. Altogether, in the light of emerging targeted therapeutic options, we suggest histological assessment in regard to B-cell or pDC preponderance to allow tailored treatment decisions.
Collapse
Affiliation(s)
- Luka de Vos
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Tugce Guel
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Dennis Niebel
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Sandra Bald
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Adrian ter Steege
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Thomas Bieber
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Joerg Wenzel
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
- *Correspondence: Joerg Wenzel,
| |
Collapse
|
80
|
Campbell NO, Davison LM, Banerjee S, Nguyen JK, Krafcik S, Silverman RH, Jorgensen TN. Ablation of SigH+ pDCs in B6.Nba2 mice prevents lupus-like disease development only if started before disease is fully established. Lupus 2022; 31:1619-1629. [PMID: 36134524 PMCID: PMC10466375 DOI: 10.1177/09612033221127561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Systemic lupus erythematosus is characterized by hyper-activation of the immune system, multi-organ inflammation, and end-organ damage. Type I interferons (IFN-I) have been strongly implicated a role in disease etiology as has the main IFN-I-producing cell subset, the plasmacytoid dendritic cell (pDC). The B6.Nba2 mouse model develops a lupus-like disease characterized by elevated IFN-I levels and pDC pathogenicity. We have previously shown that pDC ablation prior to disease development in B6.Nba2 mice effectively prevents disease; however, it remains unclear if a similar protection can be seen if pDC ablation is initiated during later disease stages. This is important as Systemic lupus erythematosus patients are rarely diagnosed until disease is well-established and thus preventative treatment is unlikely to take place. Here we show that ablation of pDCs in the B6.Nba2 mouse model must be initiated early in order to effectively block disease development and that sustained reduction in pDC numbers is necessary for sustained effects. Finally, targeting of pDCs have been hypothesized to affect immunity towards infectious agents, in particular virus and intracellular bacteria. We show here that pDC ablation in B6.Nba2 mice does not affect the anti-viral response to encephalomyocarditic virus or a model T-dependent antigen. In summary, pDC ablation does not affect general immunity, but needs to happen early and be sustained to prevent lupus-like disease development in B6.Nba2 mice.
Collapse
Affiliation(s)
- Nicole O Campbell
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
| | - Laura M Davison
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland OH, USA
- Amgen (Teneobio), Newark, CA, USA
| | - Shuvojit Banerjee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Autonomous Therapeutics, Inc., Rockville, MD, USA
| | - Jane K Nguyen
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sarah Krafcik
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
| | - Robert H Silverman
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
| | - Trine N Jorgensen
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland OH, USA
| |
Collapse
|
81
|
Zheng M, Hu Z, Zhou W, Kong Y, Wu R, Zhang B, Long H, Jia S, Lu Q, Zhao M. Single-cell transcriptome reveals immunopathological cell composition of skin lesions in subacute cutaneous lupus erythematosus. Clin Immunol 2022; 245:109172. [DOI: 10.1016/j.clim.2022.109172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/14/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022]
|
82
|
Maz MP, Martens JWS, Hannoudi A, Reddy AL, Hile GA, Kahlenberg JM. Recent advances in cutaneous lupus. J Autoimmun 2022; 132:102865. [PMID: 35858957 PMCID: PMC10082587 DOI: 10.1016/j.jaut.2022.102865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/25/2022]
Abstract
Cutaneous lupus erythematosus (CLE) is an inflammatory and autoimmune skin condition that affects patients with systemic lupus erythematosus (SLE) and exists as an isolated entity without associated SLE. Flares of CLE, often triggered by exposure to ultraviolet (UV) light result in lost productivity and poor quality of life for patients and can be associated with trigger of systemic inflammation. In the past 10 years, the knowledge of CLE etiopathogenesis has grown, leading to promising targets for better therapies. Development of lesions likely begins in a pro-inflammatory epidermis, conditioned by excess type I interferon (IFN) production to undergo increased cell death and inflammatory cytokine production after UV light exposure. The reasons for this inflammatory predisposition are not well-understood, but may be an early event, as ANA + patients without criteria for autoimmune disease exhibit similar (although less robust) findings. Non-lesional skin of SLE patients also exhibits increased innate immune cell infiltration, conditioned by excess IFNs to release pro-inflammatory cytokines, and potentially increase activation of the adaptive immune system. Plasmacytoid dendritic cells are also found in non-lesional skin and may contribute to type I IFN production, although this finding is now being questioned by new data. Once the inflammatory cycle begins, lesional infiltration by numerous other cell populations ensues, including IFN-educated T cells. The heterogeneity amongst lesional CLE subtypes isn't fully understood, but B cells appear to discriminate discoid lupus erythematosus from other subtypes. Continued discovery will provide novel targets for additional therapeutic pursuits. This review will comprehensively discuss the contributions of tissue-specific and immune cell populations to the initiation and propagation of disease.
Collapse
Affiliation(s)
- Mitra P Maz
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA; Program in Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jacob W S Martens
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA; Program in Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andrew Hannoudi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alayka L Reddy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Grace A Hile
- Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - J Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Dermatology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
83
|
Identification of Significant Genes and Pathways for the Chronic and Subacute Cutaneous Lupus Erythematosus via Bioinformatics Analysis. DISEASE MARKERS 2022; 2022:9891299. [PMID: 36212172 PMCID: PMC9537011 DOI: 10.1155/2022/9891299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/26/2022] [Accepted: 09/10/2022] [Indexed: 11/29/2022]
Abstract
Background Chronic cutaneous lupus erythematosus (CCLE) and subacute cutaneous lupus erythematosus (SCLE) are both common variants of cutaneous lupus erythematosus (CLE) that mainly involve the skin and mucous membrane. Oral mucosal involvement is frequently observed in patients of CLE. Despite that they have different clinicopathological features, whether there is a significant difference in pathogenesis between them remains unclear. Herein, we investigated specific genes and pathways of SCLE and CCLE via bioinformatics analysis. Methods Microarray expression datasets of GSE109248 and GSE112943 were both retrieved from the GEO database. Differentially expressed genes (DEGs) between CCLE or SCLE skin tissues and health controls were selected by GEO2R. Common DEGs were picked out via the Venn diagram software. Then, functional enrichment and PPI network analysis were conducted, and the top 10 key genes were identified via Cytohubba. Results Totally, 176 DEGs of SCLE and 287 DEGs of CCLE were identified. The GO enrichment and KEGG analysis of DEGs of SCLE is significantly enriched in the response to virus, defense response to virus, response to IFN-gamma, cellular response to IFN-γ, type I IFN signaling pathway, chemokine activity, chemokine receptor binding, NOD-like receptor signaling pathway, etc. The GO enrichment and KEGG analysis of DEGs of CCLE is significantly enriched in the response to virus, regulation of multiorganism process, negative regulation of viral process, regulation of lymphocyte activation, chemokine receptor binding, CCR chemokine receptor binding, NOD-like receptor signaling pathway, etc. The top 10 hub genes of SCLE and CCLE, respectively, include STAT1, CXCL10, IRF7, ISG15, and RSAD2 and CXCL10, IRF7, IFIT3, CTLA4, and ISG15. Conclusion Our finding suggests that SCLE and CCLE have the similar potential key genes and pathways and majority of them belong to IFN signatures and IFN signaling pathway. Besides, the NOD-like receptor signaling pathway might also have an essential role in the pathogenesis of SCLE and CCLE. Together, the identified genes and signaling pathways have enhanced our understanding of the mechanism underlying the occurrence and development of both SCLE and CCLE.
Collapse
|
84
|
Gene Profiling of a 3D Psoriatic Skin Model Enriched in T Cells: Downregulation of PTPRM Promotes Keratinocyte Proliferation through Excessive ERK1/2 Signaling. Cells 2022; 11:cells11182904. [PMID: 36139479 PMCID: PMC9497242 DOI: 10.3390/cells11182904] [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: 07/26/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 12/03/2022] Open
Abstract
Psoriasis is a complex, immune-mediated skin disease involving a wide range of epithelial and immune cells. The underlying mechanisms that govern the epidermal defects and immunological dysfunction observed in this condition remain largely unknown. In recent years, the emergence of new, more sophisticated models has allowed the evolution of our knowledge of the pathogenesis of psoriasis. The development of psoriatic skin biomaterials that more closely mimic native psoriatic skin provides advanced preclinical models that will prove relevant in predicting clinical outcomes. In this study, we used a tissue-engineered, two-layered (dermis and epidermis) human skin substitute enriched in T cells as a biomaterial to study both the cellular and molecular mechanisms involved in psoriasis’ pathogenesis. Gene profiling on microarrays revealed significant changes in the profile of genes expressed by the psoriatic skin substitutes compared with the healthy ones. Two genes, namely, PTPRM and NELL2, whose products influence the ERK1/2 signaling pathway have been identified as being deregulated in psoriatic substitutes. Deregulation of these genes supports excessive activation of the ERK1/2 pathway in psoriatic skin substitutes. Most importantly, electrophoresis mobility shift assays provided evidence that the DNA-binding properties of two downstream nuclear targets of ERK1/2, both the NF-κB and Sp1 transcription factors, are increased under psoriatic conditions. Moreover, the results obtained with the inhibition of RSK, a downstream effector of ERK1/2, supported the therapeutic potential of inhibiting this signaling pathway for psoriasis treatment. In conclusion, this two-layered human psoriatic skin substitute enriched in T cells may prove particularly useful in deciphering the mechanistic details of psoriatic pathogenesis and provide a relevant biomaterial for the study of potential therapeutic targets.
Collapse
|
85
|
Soto JA, Melo-González F, Riedel CA, Bueno SM, Kalergis AM. Modulation of Immune Cells as a Therapy for Cutaneous Lupus Erythematosus. Int J Mol Sci 2022; 23:10706. [PMID: 36142624 PMCID: PMC9504747 DOI: 10.3390/ijms231810706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/04/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Cutaneous lupus erythematosus (CLE) is an autoimmune disorder like systemic lupus erythematosus (SLE). Both SLE and CLE characterize autoantibody secretion and immune cell recruitment. In particular, CLE can be divided into three more frequent types, varying in the severity of the skin lesions they present. The role of type I IFN was shown to be one of the leading causes of the development of this pathology in the skin. Different treatments have been developed and tested against these different variants of CLE to decrease the increasing levels of CLE in humans. In this article, a literature revision discussing the similarities between SLE and CLE is carried out. In addition, new advances in understanding the development of CLE and the leading treatments being evaluated in animal models and clinical trials are reviewed.
Collapse
Affiliation(s)
- Jorge A. Soto
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370146, Chile
| | - Felipe Melo-González
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370146, Chile
| | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370146, Chile
| | - Susan M. Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330023, Chile
| |
Collapse
|
86
|
Psarras A, Wittmann M, Vital EM. Emerging concepts of type I interferons in SLE pathogenesis and therapy. Nat Rev Rheumatol 2022; 18:575-590. [PMID: 36097207 DOI: 10.1038/s41584-022-00826-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 11/09/2022]
Abstract
Type I interferons have been suspected for decades to have a crucial role in the pathogenesis of systemic lupus erythematosus (SLE). Evidence has now overturned several long-held assumptions about how type I interferons are regulated and cause pathological conditions, providing a new view of SLE pathogenesis that resolves longstanding clinical dilemmas. This evidence includes data on interferons in relation to genetic predisposition and epigenetic regulation. Importantly, data are now available on the role of interferons in the early phases of the disease and the importance of non-haematopoietic cellular sources of type I interferons, such as keratinocytes, renal tubular cells, glial cells and synovial stromal cells, as well as local responses to type I interferons within these tissues. These local effects are found not only in inflamed target organs in established SLE, but also in histologically normal skin during asymptomatic preclinical phases, suggesting a role in disease initiation. In terms of clinical application, evidence relating to biomarkers to characterize the type I interferon system is complex, and, notably, interferon-blocking therapies are now licensed for the treatment of SLE. Collectively, the available data enable us to propose a model of disease pathogenesis that invokes the unique value of interferon-targeted therapies. Accordingly, future approaches in SLE involving disease reclassification and preventative strategies in preclinical phases should be investigated.
Collapse
Affiliation(s)
- Antonios Psarras
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Miriam Wittmann
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Edward M Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK. .,NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| |
Collapse
|
87
|
Klein B, Kunz M. Current concepts of photosensitivity in cutaneous lupus erythematosus. Front Med (Lausanne) 2022; 9:939594. [PMID: 36091671 PMCID: PMC9452788 DOI: 10.3389/fmed.2022.939594] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Cutaneous lupus erythematosus (CLE) represents a complex autoimmune disease with a broad phenotypic spectrum ranging from acute to chronic destructive cutaneous lesions. Patients with CLE exhibit high photosensitivity and ultraviolet (UV) irradiation can lead to systemic flares in systemic lupus erythematosus. However, the exact mechanisms how UV irradiation enhances cutaneous inflammation in lupus are not fully understood. Recently, new molecular mechanisms of UV-driven immune responses in CLE were identified, offering potential therapeutic approaches. Especially the induction of type I interferons, central cytokines in lupus pathogenesis which are released by various skin cells, have become the focus of current research. In this review, we describe current pathogenic concepts of photosensitivity in lupus erythematosus, including UV-driven activation of intracellular nucleic acid sensors, cellular cytokine production and immune cell activation. Furthermore, we discuss activated pathways contributing to enhanced apoptosis as well as intracellular translocation of autoantigens thereby promoting CLE upon UV light exposure.
Collapse
Affiliation(s)
- Benjamin Klein
- Department of Dermatology, Venereology, and Allergology, University Hospital Leipzig, Leipzig, Germany
| | | |
Collapse
|
88
|
Min X, Zheng M, Yu Y, Wu J, Kuang Q, Hu Z, Ouyang L, Lu S, Zhao M. Ultraviolet light induces HERV expression to activate RIG-I signalling pathway in keratinocytes. Exp Dermatol 2022; 31:1165-1176. [PMID: 35332586 DOI: 10.1111/exd.14568] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/12/2022] [Accepted: 03/21/2022] [Indexed: 11/30/2022]
Abstract
Skin inflammation and photosensitivity are common in lupus erythematosus (LE) patients, and ultraviolet (UV) light is a known trigger of skin and possibly systemic inflammation in systemic lupus erythematosus (SLE) and discoid lupus erythematosus (DLE) patients. Type I interferons (IFN) are upregulated in LE skin after UV exposure; however, the mechanisms to explain UVB-induced inflammation remain unclear. Here, we demonstrated that UVB irradiation-induced activation of human endogenous retroviruses (HERVs) plays a major role in the immune response. UVB-induced HERV-associated dsRNA transcription and subsequent activation of the innate antiviral RIG-I/MDA5/IRF7 pathway led to downstream transcription of interferon-stimulated genes, which promotes UVB-induced apoptosis and proliferation inhibition in keratinocytes through RIG-I and MDA5 pathways. Our findings indicate that UVB irradiation induces HERV-dsRNA overexpression, and the dsRNA-sensing innate immunity pathway promotes type I IFN production, which may be a potential mechanism of skin inflammatory response and skin lesion of SLE/DLE.
Collapse
Affiliation(s)
- Xiaoli Min
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Meiling Zheng
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Yaqin Yu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Jiali Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Qiqi Kuang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Zhi Hu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Lianlian Ouyang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Shuang Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| |
Collapse
|
89
|
IL-33/ST2 Activation Is involved in Ro60-Regulated Photosensitivity in Cutaneous Lupus Erythematosus. Mediators Inflamm 2022; 2022:4955761. [PMID: 35909659 PMCID: PMC9328989 DOI: 10.1155/2022/4955761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
Interleukin- (IL-) 33 contributes to various inflammatory processes. IL-33/ST2 activation participates in systemic lupus erythematous via binding to the receptor of Suppression of Tumorigenicity 2 protein (ST2). However, whether IL-33/ST2 interferes with the nosogenesis of cutaneous lupus erythematosus (CLE) has not been reported so far. Herein, we proposed to disclose the impacts on IL-33/ST2 activation and Ro60 on CLE and their potential implications in the photosensitization of CLE cells. IL-33, ST2, and Ro60 in CLE patients' skin lesions were detected. Murine keratinocytes stimulated with or without IL-33 were irradiated by ultraviolet B (UVB), and the levels of Ro60 and inflammation markers were determined. Keratinocytes were cocultured with J774.2 macrophages and stimulated with IL-33 for analysis of chemostasis. The results identified that IL-33, ST2, and downstream inflammation markers were significantly upregulated in CLE lesions with Ro60 overexpression. Additionally, IL-33 treatment promoted the upregulation of Ro60 induced by UVB treatment in murine keratinocytes. Moreover, IL-33 stimulates keratinocytes to induce macrophage migration via enhancing the generation of the chemokine (C–C motif) ligands 17 and 22. Meanwhile, the silencing of ST2 or nuclear factor-kappa B (NF-κB) suppression abolished IL-33-induced upregulation of Ro60 in keratinocytes. Similarly, the inhibition of SOX17 expression was followed by downregulation of Ro60 in keratinocytes following IL-33 stimulation. In addition, UVB irradiation upregulated SOX17 in keratinocytes. Conclusively, the IL-33/ST2 axis interferes with Ro60-regulated photosensitization via activating the NF-κB- and PI3K/Akt- and SOX17-related pathways.
Collapse
|
90
|
Li W, Zhang C, Wang YY, Xiao L, Feng Y, Huo X, Wang C, Sun Y, Wang F, Sun T. Alterations of RNAs in the insula related to cocaine-induced condition place preference in adolescent mice. Biochem Biophys Res Commun 2022; 621:109-115. [PMID: 35820280 DOI: 10.1016/j.bbrc.2022.06.080] [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: 05/10/2022] [Revised: 05/29/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022]
Abstract
Cocaine as a highly addictive psychostimulant can cause changes in the body at the cellular and molecular levels over a long period of time. It reminds us that cocaine may have a potential role in post-transcriptional regulation, but the alteration of insula-expression profile in adolescent cocaine use disorder (CUD) has not been reported. To reveal the mechanisms underlying the post-transcriptional regulation of cocaine, we investigate the transcriptome in the insula of cocaine-induced mice based on high-throughput strand-specific RNA sequencing. We analyzed the alterations of messenger RNA (mRNA) expression profile in the insula of cocaine-induced condition place preference (CPP) mice and then correlated it with microRNAs to reveal their involvement in the formation of cocaine-induced CPP. In this study, a total of 27786 genes were identified, 5750 new genes (novel expressed transcripts of unannotated in the reference genome) were discovered, among which 1,205 were annotated functionally. A total of 198 differentially expressed genes (DEG) that functioned in synaptic transmission, cholinergic, developmental process, neurotransmitter metabolic process, drug catabolism, cellular response to drug, MAP kinase activity, ceramidase activity, and drug resistance were significantly enriched. Further analysis showed that 26045 mRNAs formed 45,208 network-relationship pairs with 1770 microRNAs. In the current study, our work was the first to reveal that alterations of RNAs in the insula, as a core brain region of the neural circuits of interoception, were involved in the process of cocaine-induced CPP of adolescent mice. These findings enrich the biology and expand the molecular regulatory network related to adolescence CUD. They provided the possibility that some DEGs may be used as novel biomarkers for the diagnosis or evaluation of substance use disorder, and also provided clues for elucidating the neurobiological mechanism of substance use disorder.
Collapse
Affiliation(s)
- Wenchao Li
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan, China
| | - Chun Zhang
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan, China
| | - Yang Yang Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan, China
| | - Lifei Xiao
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan, China
| | - Yan Feng
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan, China
| | - Xianhao Huo
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan, China
| | - Chaofan Wang
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan, China
| | - Yu Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan, China
| | - Feng Wang
- Zhejiang University School of Medicine, Hangzhou, China.
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Disease, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
91
|
Vital EM, Merrill JT, Morand EF, Furie RA, Bruce IN, Tanaka Y, Manzi S, Kalunian KC, Kalyani RN, Streicher K, Abreu G, Tummala R. Anifrolumab efficacy and safety by type I interferon gene signature and clinical subgroups in patients with SLE: post hoc analysis of pooled data from two phase III trials. Ann Rheum Dis 2022; 81:951-961. [PMID: 35338035 PMCID: PMC9213795 DOI: 10.1136/annrheumdis-2021-221425] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/26/2022] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To characterise the efficacy and safety of anifrolumab in patients with systemic lupus erythematosus (SLE) according to interferon gene signature (IFNGS), demographic and clinical subgroups. METHODS We performed post hoc analyses of pooled data from the 52-week phase III TULIP-1/TULIP-2 placebo-controlled trials of intravenous anifrolumab in moderate-to-severe SLE. Outcomes were assessed in predefined subgroups: IFNGS (high/low), age, sex, body mass index, race, geographic region, age of onset, glucocorticoid use, disease activity and serological markers. RESULTS In pooled data, patients received anifrolumab 300 mg (360/726) or placebo (366/726); 82.6% were IFNGS-high. IFNGS-high patients had greater baseline disease activity and were more likely to have abnormal serological markers versus IFNGS-low patients. In the total population, a greater proportion of patients treated with anifrolumab versus placebo achieved British Isles Lupus Assessment Group-based Composite Lupus Assessment (BICLA) response at week 52 (difference 16.6%; nominal p<0.001). BICLA response treatment differences with anifrolumab versus placebo were comparable to the total population across most predefined subgroups, including subgroups for baseline glucocorticoid dosage (<10/≥10 mg/day prednisone/equivalent) and for clinical disease activity (SLE Disease Activity Index 2000 score <10/≥10). Subgroups with larger treatment differences included IFNGS-high patients (18.2%), patients with abnormal baseline serological markers (23.1%) and Asian patients (29.2%). The safety profile of anifrolumab was similar across subgroups. CONCLUSIONS Overall, this study supports the consistent efficacy and safety of anifrolumab across a range of patients with moderate-to-severe SLE. In a few subgroups, small sample sizes limited conclusions from being drawn regarding the treatment benefit with anifrolumab. TRIAL REGISTRATION NUMBER NCT02446912, NCT02446899.
Collapse
Affiliation(s)
- Edward M Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Joan T Merrill
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Eric F Morand
- Centre for Inflammatory Disease Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Richard A Furie
- Division of Rheumatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, New York, USA
| | - Ian N Bruce
- Centre for Epidemiology Versus Arthritis, The University of Manchester, NIHR Manchester Biomedical Research Centre, Manchester, UK
- Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Susan Manzi
- Lupus Center of Excellence, Autoimmunity Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Kenneth C Kalunian
- Division of Rheumatology, Allergy and Immunology, University of California San Diego, La Jolla, California, USA
| | - Rubana N Kalyani
- BioPharmaceuticals R&D, AstraZeneca US, Gaithersburg, Maryland, USA
| | - Katie Streicher
- BioPharmaceuticals R&D, AstraZeneca US, Gaithersburg, Maryland, USA
| | - Gabriel Abreu
- BioPharmaceuticals R&D, AstraZeneca R&D, Gothenburg, Sweden
| | - Raj Tummala
- BioPharmaceuticals R&D, AstraZeneca US, Gaithersburg, Maryland, USA
| |
Collapse
|
92
|
Fetter T, Braegelmann C, de Vos L, Wenzel J. Current Concepts on Pathogenic Mechanisms and Histopathology in Cutaneous Lupus Erythematosus. Front Med (Lausanne) 2022; 9:915828. [PMID: 35712102 PMCID: PMC9196867 DOI: 10.3389/fmed.2022.915828] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Cutaneous lupus erythematosus (CLE) is an interferon (IFN)-driven autoimmune disease that may be limited to the skin or can be associated with systemic lupus erythematosus (SLE). CLE occurs in several morphologic subtypes ranging from isolated, disc-shaped plaques to disseminated skin lesions. The typical histopathologic pattern of skin lesions is named interface dermatitis and characterized by a lymphocytic infiltrate and necroptotic keratinocytes at the dermo-epidermal junction. Other histopathologic patterns primarily involve the dermis or subcutis, depending on the subtype. One critical mechanism in CLE is the chronic reactivation of innate and adaptive immune pathways. An important step in this process is the recognition of endogenous nucleic acids released from dying cells by various pattern recognition receptors (PRRs), including Toll-like receptors (TLRs) and other cytosolic receptors. Crucial cells in CLE pathogenesis comprise plasmacytoid dendritic cells (pDCs) as major producers of type I IFN, T cells exerting cytotoxic effects, and B cells, previously believed to contribute via secretion of autoantibodies. However, B cells are increasingly considered to have additional functions, supported by studies finding them to occur in highest numbers in chronic discoid lupus erythematosus (CDLE), a subtype in which autoantibodies are often absent. More precise knowledge of how CLE subtypes differ pathophysiologically may allow a tailored pharmacotherapy in the future, taking into account the specific molecular signature in relation to the morphologic subtype.
Collapse
Affiliation(s)
- Tanja Fetter
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | | | - Luka de Vos
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Joerg Wenzel
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
93
|
Chen HW, Barber G, Chong BF. The Genetic Landscape of Cutaneous Lupus Erythematosus. Front Med (Lausanne) 2022; 9:916011. [PMID: 35721085 PMCID: PMC9201079 DOI: 10.3389/fmed.2022.916011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Cutaneous lupus erythematosus (CLE) is an autoimmune connective tissue disease that can exist as a disease entity or within the context of systemic lupus erythematosus (SLE). Over the years, efforts to elucidate the genetic underpinnings of CLE and SLE have yielded a wealth of information. This review examines prior studies investigating the genetics of CLE at the DNA and RNA level and identifies future research areas. In this literature review, we examined the English language literature captured within the MEDLINE and Embase databases using pre-defined search terms. First, we surveyed studies investigating various DNA studies of CLE. We identified three predominant areas of focus in HLA profiling, complement deficiencies, and genetic polymorphisms. An increased frequency of HLA-B8 has been strongly linked to CLE. In addition, multiple genes responsible for mediating innate immune response, cell growth, apoptosis, and interferon response confer a higher risk of developing CLE, specifically TREX1 and SAMHD1. There was a strong association between C2 complement deficiency and CLE. Second, we reviewed literature studying aberrations in the transcriptomes of patients with CLE. We reviewed genetic aberrations initiated by environmental insults, and we examined the interplay of dysregulated inflammatory, apoptotic, and fibrotic pathways in the context of the pathomechanism of CLE. These current learnings will serve as the foundation for further advances in integrating personalized medicine into the care of patients with CLE.
Collapse
|
94
|
Werth VP, Fleischmann R, Robern M, Touma Z, Tiamiyu I, Gurtovaya O, Pechonkina A, Mozaffarian A, Downie B, Matzkies F, Wallace D. Filgotinib or lanraplenib in moderate to severe cutaneous lupus erythematosus: a phase 2, randomized, double-blind, placebo-controlled study. Rheumatology (Oxford) 2022; 61:2413-2423. [PMID: 34498056 PMCID: PMC9157055 DOI: 10.1093/rheumatology/keab685] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/01/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To explore the safety and efficacy of filgotinib (FIL), a Janus kinase 1 inhibitor, and lanraplenib (LANRA), a spleen kinase inhibitor, in cutaneous lupus erythematosus (CLE). METHODS This was a phase 2, randomized, double-blind, placebo-controlled, exploratory, proof-of-concept study of LANRA (30 mg), FIL (200 mg) or placebo (PBO) once daily for 12 weeks in patients with active CLE. At week 12, PBO patients were rerandomized 1:1 to receive LANRA or FIL for up to 36 additional weeks. RESULTS Of 47 randomized patients, 45 were treated (PBO, n = 9; LANRA, n = 19; FIL, n = 17). The primary endpoint [change from baseline in Cutaneous Lupus Erythematosus Disease Area and Severity Index Activity (CLASI-A) score at week 12] was not met. The least squares mean CLASI-A score change from baseline was -5.5 (s.e. 2.56) with PBO, -4.5 (1.91) with LANRA and -8.7 (1.85) with FIL. Numerical differences between FIL and PBO were greater in select subgroups. A ≥5-point improvement in the CLASI-A score at week 12 was achieved by 50.0%, 56.3% and 68.8% in the PBO, LANRA and FIL arms, respectively. A numerically greater proportion of patients in the FIL arm (50%) also achieved ≥50% improvement in the CLASI-A score at week 12 (37.5% PBO, 31.3% LANRA). Most adverse events (AEs) were mild or moderate in severity. Two serious AEs were reported with LANRA and one with FIL. CONCLUSION The primary endpoint was not met. Select subgroups displayed a numerically greater treatment response to FIL relative to PBO. LANRA and FIL were generally well tolerated. TRIAL REGISTRATION ClinicalTrials.gov identifier NCT03134222.
Collapse
Affiliation(s)
- Victoria P Werth
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Corporal Michael J. Crescenz VAMC, Philadelphia, PA
| | - Roy Fleischmann
- Department of Internal Medicine, Metroplex Clinical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Zahi Touma
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | - Daniel Wallace
- Rheumatology, Cedars-Sinai/David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
95
|
Patel J, Vazquez T, Chin F, Keyes E, Yan D, Diaz D, Grinnell M, Sharma M, Li Y, Feng R, Sprow G, Dan J, Werth VP. Multidimensional immune profiling of cutaneous lupus erythematosus in vivo stratified by patient responses to antimalarials. Arthritis Rheumatol 2022; 74:1687-1698. [PMID: 35583812 DOI: 10.1002/art.42235] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/14/2022] [Accepted: 05/12/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The pathogenesis of cutaneous lupus erythematous (CLE) is multifactorial and CLE is difficult to treat due to heterogeneity of inflammatory processes between patients. Antimalarials such as hydroxychloroquine (HCQ) and quinacrine (QC) have long been first-line systemic therapy; however, many patients do not respond and require systemic immunosuppressants with undesirable side effects. Given the complexity and unpredictable responses in CLE, we sought to identify the immunologic landscape of CLE patients stratified by subsequent treatment outcomes to identify potential biomarkers of inducible response. METHOD We performed imaging mass cytometry with 48 treatment-naïve skin biopsies of HCQ responders, QC responders, and non-responders (NR) to analyze multiple immune cell types and inflammatory markers in their native environment in CLE skin. Patients were stratified according to their subsequent response to antimalarials to identify baseline immunophenotypes which may predict response to therapy. RESULTS HCQ responders demonstrated increased CD4 T cells compared to QC. NR had decreased Tregs compared to QC and increased central memory T cells compared to HCQ. QC responders expressed increased phosphorylated (p) STING and IFNκ compared to HCQ. pSTING and IFNκ localized to conventional dendritic cells and positively correlated on a tissue and cellular level. Neighborhood analysis revealed decreased regulatory cell interactions in NR patients. Hierarchical clustering revealed NR groups separated based on pSTAT2/3/4/5, pIRF3, Granzyme B, pJAK2, IL4, IL17, and IFNγ. CONCLUSION These findings demonstrate differential immune compositions between CLE patients, guiding the future for precision-based medicine and treatment response.
Collapse
Affiliation(s)
- Jay Patel
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas Vazquez
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104
| | - Felix Chin
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily Keyes
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daisy Yan
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - DeAnna Diaz
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Madison Grinnell
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meena Sharma
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yubin Li
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Feng
- Department of Biostatistics and Epidemiology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Grant Sprow
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Josh Dan
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Victoria P Werth
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104.,Department of Dermatology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
96
|
Wolf SJ, Audu CO, Joshi A, denDekker A, Melvin WJ, Davis FM, Xing X, Wasikowski R, Tsoi LC, Kunkel SL, Gudjonsson JE, O’Riordan MX, Kahlenberg JM, Gallagher KA. IFN-κ is critical for normal wound repair and is decreased in diabetic wounds. JCI Insight 2022; 7:e152765. [PMID: 35358091 PMCID: PMC9090246 DOI: 10.1172/jci.insight.152765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 03/24/2022] [Indexed: 02/01/2023] Open
Abstract
Wound repair following acute injury requires a coordinated inflammatory response. Type I IFN signaling is important for regulating the inflammatory response after skin injury. IFN-κ, a type I IFN, has recently been found to drive skin inflammation in lupus and psoriasis; however, the role of IFN-κ in the context of normal or dysregulated wound healing is unclear. Here, we show that Ifnk expression is upregulated in keratinocytes early after injury and is essential for normal tissue repair. Under diabetic conditions, IFN-κ was decreased in wound keratinocytes, and early inflammation was impaired. Furthermore, we found that the histone methyltransferase mixed-lineage leukemia 1 (MLL1) is upregulated early following injury and regulates Ifnk expression in diabetic wound keratinocytes via an H3K4me3-mediated mechanism. Using a series of in vivo studies with a geneticall y engineered mouse model (Mll1fl/fl K14cre-) and human wound tissues from patients with T2D, we demonstrate that MLL1 controls wound keratinocyte-mediated Ifnk expression and that Mll1 expression is decreased in T2D keratinocytes. Importantly, we found the administration of IFN-κ early following injury improves diabetic tissue repair through increasing early inflammation, collagen deposition, and reepithelialization. These findings have significant implications for understanding the complex role type I IFNs play in keratinocytes in normal and diabetic wound healing. Additionally, they suggest that IFN may be a viable therapeutic target to improve diabetic wound repair.
Collapse
Affiliation(s)
| | | | - Amrita Joshi
- Section of Vascular Surgery, Department of Surgery
| | | | | | | | | | | | | | | | | | | | - J. Michelle Kahlenberg
- Department of Dermatology
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Katherine A. Gallagher
- Section of Vascular Surgery, Department of Surgery
- Department of Microbiology and Immunology, and
| |
Collapse
|
97
|
Halasi M, Nyska A, Rubin L, Tal Y, Tsokos GC, Adini I. Melanocyte-secreted fibromodulin constrains skin inflammation in mice injected with lupus serum. Clin Immunol 2022; 241:109055. [DOI: 10.1016/j.clim.2022.109055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 05/25/2022] [Indexed: 11/03/2022]
|
98
|
Billi AC, Ma F, Plazyo O, Gharaee-Kermani M, Wasikowski R, Hile GA, Xing X, Yee CM, Rizvi SM, Maz MP, Berthier CC, Wen F, Tsoi LC, Pellegrini M, Modlin RL, Gudjonsson JE, Kahlenberg JM. Nonlesional lupus skin contributes to inflammatory education of myeloid cells and primes for cutaneous inflammation. Sci Transl Med 2022; 14:eabn2263. [PMID: 35476593 PMCID: PMC9169615 DOI: 10.1126/scitranslmed.abn2263] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cutaneous lupus erythematosus (CLE) is a disfiguring and poorly understood condition frequently associated with systemic lupus. Previous studies suggest that nonlesional keratinocytes play a role in disease predisposition, but this has not been investigated in a comprehensive manner or in the context of other cell populations. To investigate CLE immunopathogenesis, normal-appearing skin, lesional skin, and circulating immune cells from lupus patients were analyzed via integrated single-cell RNA sequencing and spatial RNA sequencing. We demonstrate that normal-appearing skin of patients with lupus represents a type I interferon-rich, prelesional environment that skews gene transcription in all major skin cell types and markedly distorts predicted cell-cell communication networks. We also show that lupus-enriched CD16+ dendritic cells undergo robust interferon education in the skin, thereby gaining proinflammatory phenotypes. Together, our data provide a comprehensive characterization of lesional and nonlesional skin in lupus and suggest a role for skin education of CD16+ dendritic cells in CLE pathogenesis.
Collapse
Affiliation(s)
| | - Feiyang Ma
- Dept of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Olesya Plazyo
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Mehrnaz Gharaee-Kermani
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Grace A. Hile
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Xianying Xing
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Christine M. Yee
- Dept of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Syed M. Rizvi
- Dept of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mitra P. Maz
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Celine C. Berthier
- Division of Nephrology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Fei Wen
- Dept of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Lam C. Tsoi
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Matteo Pellegrini
- Dept of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Robert L. Modlin
- Dept of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | | | - J. Michelle Kahlenberg
- Dept of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
99
|
Aringer M, Alarcón-Riquelme ME, Clowse M, Pons-Estel GJ, Vital EM, Dall’Era M. A glimpse into the future of systemic lupus erythematosus. Ther Adv Musculoskelet Dis 2022; 14:1759720X221086719. [PMID: 35368371 PMCID: PMC8972918 DOI: 10.1177/1759720x221086719] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/23/2022] [Indexed: 12/17/2022] Open
Abstract
This viewpoint article on a forecast of clinically meaningful changes in the management of systemic lupus erythematosus (SLE) in the next 10 years is based on a review of the current state of the art. The groundwork has been laid by a robust series of classification criteria and treatment recommendations that have all been published since 2019. Building on this strong foundation, SLE management predictably will take significant steps forward. Assessment for lupus arthritis will presumably include musculoskeletal sonography. Large-scale polyomics studies are likely to unravel more of the central immune mechanisms of the disease. Biomarkers predictive of therapeutic success may enter the field; the type I interferon signature, as a companion for use of anifrolumab, an antibody against the common type I interferon receptor, is one serious candidate. Besides anifrolumab for nonrenal SLE and the new calcineurin inhibitor voclosporin in lupus nephritis, both of which are already approved in the United States and likely to become available in the European Union in 2022, several other approaches are in advanced clinical trials. These include advanced B cell depletion, inhibition of costimulation via CD40 and CD40 ligand (CD40L), and Janus kinase 1 (Jak1) and Tyrosine kinase 2 (Tyk2) inhibition. At the same time, essentially all of our conventional therapeutic armamentarium will continue to be used. The ability of patients to have successful SLE pregnancies, which has become much better in the last decades, should further improve, with approaches including tumor necrosis factor blockade and self-monitoring of fetal heart rates. While we hope that the COVID-19 pandemic will soon be controlled, it has highlighted the risk of severe viral infections in SLE, with increased risk tied to certain therapies. Although there are some data that a cure might be achievable, this likely will remain a challenge beyond 10 years from now.
Collapse
Affiliation(s)
- Martin Aringer
- Professor of Medicine (Rheumatology), Division of Rheumatology, Department of Medicine III, University Medical Center and Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Marta E. Alarcón-Riquelme
- Department of Medical Genomics, GENYO, Pfizer-University of Granada-Andalusian Government Center for Genomics and Oncological Research, Granada, Spain
| | - Megan Clowse
- Division of Rheumatology & Immunology, Duke University, Durham, NC, USA
| | - Guillermo J. Pons-Estel
- Department of Rheumatology, Grupo Oroño–Centro Regional de Enfermedades Autoinmunes y Reumáticas (GO-CREAR), Rosario, Argentina
| | - Edward M. Vital
- University of Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Maria Dall’Era
- Lupus Clinic and Rheumatology Clinical Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
100
|
Zeidi M, Chen KL, Patel J, Desai K, Kim HJ, Chakka S, Lim R, Werth VP. Increased CD69+CCR7+ circulating activated T cells and STAT3 expression in cutaneous lupus erythematosus patients recalcitrant to antimalarials. Lupus 2022; 31:472-481. [PMID: 35258358 DOI: 10.1177/09612033221084093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Antimalarials are first-line systemic therapy for cutaneous lupus erythematosus (CLE). While some patients unresponsive to hydroxychloroquine (HCQ) alone benefit from the addition of quinacrine (QC), a subset of patients is refractory to both antimalarials. METHODS We classified CLE patients as HCQ-responders, HCQ+QC-responders, or HCQ+QC-nonresponders to compare immune profiles. Immunohistochemistry, immunofluorescence, and qRT-PCR were used to characterize inflammatory cells and cytokines in lesional skin. RESULTS Immunohistochemistry showed that CD69+ T cells were higher in HCQ+QC-nonresponders compared to HCQ- and HCQ+QC-responders (p < 0.05). Immunofluorescence further identified these cells as CD69+CCR7+ circulating activated T cells. Myeloid dendritic cells were significantly higher in HCQ+QC-responders compared to both HCQ-responders and HCQ+QC-nonresponders. Plasmacytoid dendritic cells were significantly increased in HCQ-responders compared to HCQ- and HCQ+QC-nonresponders. No differences were found in the number of autoreactive T cells, MAC387+ cells, and neutrophils among the groups. CLASI scores of the HCQ+QC-nonresponder group positively correlated with CD69+CCR7+ circulating activated T cells (r = 0.6335, p < 0.05) and MAC387+ cells (r = 0.5726, p < 0.05). IL-17 protein expression was higher in HCQ+QC-responders compared to HCQ-responders or HCQ+QC-nonresponders, while IL-22 protein expression did not differ. mRNA expression demonstrated increased STAT3 expression in a subset of HCQ+QC-nonresponders. CONCLUSION An increased number of CD69+CCR7+ circulating activated T cells and a strong correlation with CLASI scores in the HCQ+QC-nonresponders suggest these cells are involved in antimalarial-refractory skin disease. STAT3 is also increased in HCQ+QC-nonresponders and may also be a potential target for antimalarial-refractory skin disease.
Collapse
Affiliation(s)
- Majid Zeidi
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Kristen L Chen
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Jay Patel
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Krisha Desai
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Hee Joo Kim
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Srita Chakka
- Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Lim
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA
| | - Victoria P Werth
- Corporal Michael J Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, 14640University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|