1
|
Tao Y, Hua G, Min S, Xiao-hong L, Jia-hui J, Biao T, Cai-feng H, Cheng Z, Chao C, Jian-ping W. Verification of biological markers of subacute cutaneous lupus erythematosus via TMT labelling proteomics combined with transcriptome data. Ann Med 2025; 57:2500696. [PMID: 40323689 PMCID: PMC12054568 DOI: 10.1080/07853890.2025.2500696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/25/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025] Open
Abstract
OBJECTIVE This study aimed to investigate biological markers in subacute cutaneous lupus erythematosus (SCLE). METHODS The tandem mass tag (TMT)-labelling proteomics method was used to explore differentially expressed proteins between SCLE lesions and normal skin tissues. The differences in transcriptomic data between SCLE tissues and normal skin tissues were analysed from the GEO database (GSE81071, GSE109248 and GSE112943). The differences in transcriptomic data from peripheral blood mononuclear cells (PBMCs) of patients with systemic lupus erythematosus (SLE) and normal controls were analysed (GSE81622 and GSE154851). The 35 healthy controls, 30 SCLE patients, 35 SLE patients and 30 lupus nephritis (LN) patients were diagnosed and enrolled. The serum expression levels of IFI44 and EPSTI1 were detected. Data were presented as the mean ± standard deviation or frequency and were analysed using Student's t-test, Chi-square test and one-way ANOVA between the groups. Receiver operating characteristic (ROC) curves were used to analyse the clinical efficacy of IFI44 and EPSTI1 in distinguishing SCLE from SLE. RESULTS In a comparative analysis of SCLE lesions and normal skin tissues, proteomics studies identified 376 proteins that exhibited significant differential expression. In GO and KEGG analyses, the enriched terms mainly included the interferon-gamma-mediated signalling pathway (p < .001), immune receptor activity (p < .001) and cell adhesion molecules (p < .001). The top 10 hub genes were screened in SCLE as follows: CD8A, CXCL10, IFI44, CD7, CCL5, TLR4, EPSTI1, ISG15, KLRD1 and SELL using Cytoscape (3.10.1) software. The 15 common proteins/genes between proteomics and three datasets results were found, including CXCL10, OAS1, DDX60L, CFB, IFI6, HERC6, IFI44L, GBP1, EPSTI1, OAS2, CXCL11, TYMP, IFI44, ISG15 and IFIT3. The 61 differentially expressed genes in GSE81622 and the top 100 differentially expressed genes in GSE154851, alongside the 15 identified genes described above through Venn diagram analysis. Four common genes, IFI44L, IFI44, EPSTI1 and OAS1, were identified. Two common genes, IFI44 and EPSTI1, were found in hub genes from the proteomics results. The serum levels of IFI44 and EPSTI1 in LN were significantly higher than those in SLE patients (p < .05). ROC curve analysis demonstrated that serum levels of IFI44 and EPSTI1 could differentiate SCLE from SLE with an area under the curve (AUC) of 0.898 and 0.847, respectively. CONCLUSIONS The IFI44 and EPSTI1 proved to be closely involved in the progression from SCLE to SLE, and can represent new candidate diagnostic molecular markers of occurrence and progression of SCLE.
Collapse
Affiliation(s)
- Yuan Tao
- Department of Dermatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Guo Hua
- Department of Dermatology, The Affiliated Huishan Hospital of Xinglin College, Nantong University, Wuxi Huishan District People’s Hospital, Wuxi, China
| | - Sun Min
- Department of Dermatology, The Affiliated Huishan Hospital of Xinglin College, Nantong University, Wuxi Huishan District People’s Hospital, Wuxi, China
| | - Lu Xiao-hong
- Department of Dermatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Jiang Jia-hui
- Department of Dermatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Tang Biao
- Department of Dermatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - He Cai-feng
- Department of Dermatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Zhang Cheng
- Department of Dermatology, Anhui Provincial Children’s Hospital, Hefei, China
| | - Ci Chao
- Department of Dermatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Wu Jian-ping
- Department of Dermatology, Anhui Provincial Children’s Hospital, Hefei, China
| |
Collapse
|
2
|
Bokor LA, Martyin K, Krebs M, Galajda NÁ, Meznerics FA, Szabó B, Hegyi P, Lőrincz K, Kiss N, Bánvölgyi A, Hidvégi B. Deucravacitinib shows superior efficacy and safety in cutaneous lupus erythematosus compared to various biologics and small molecules - A systematic review and meta-analysis. Autoimmun Rev 2025; 24:103723. [PMID: 39694128 DOI: 10.1016/j.autrev.2024.103723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Novel therapies for cutaneous lupus erythematosus (CLE) and systemic lupus erythematosus (SLE) demonstrated efficacy and safety in previous trials. However, data on the comparison of these treatments is still lacking, limiting their integration into clinical practice. Therefore, our aim is to perform a systematic review and network meta-analysis to compare the efficacy and safety of novel systemic therapies in CLE. METHODS A systematic search was performed across PubMed, Embase, and CENTRAL on November 25, 2023, to identify studies involving patients with CLE or SLE with active skin involvement treated with novel systemic therapies. The primary outcomes assessed were the proportion of patients achieving the Cutaneous Lupus Erythematosus Disease Area and Severity Index-50 (CLASI-50), the change in CLASI-A, the occurrence of adverse events (AEs), and serious adverse events (SAEs). RESULTS 18,280 records were retrieved, of which 53 met the inclusion criteria. Deucravacitinib showed significantly greater efficacy in achieving the CLASI50 compared to placebo (OR: 8.28, 95 % CI: 2.22-30.91). Both litifilimab (OR: 2.54, 95 % CI: 1.20-5.40) and anifrolumab (OR: 2.25, 95 % CI: 1.23-4.14) were also significantly more effective than placebo. No significant differences were observed in the occurrence of AEs and SAEs between these therapeutics and placebo. CONCLUSION Anifrolumab and litifilimab are effective and safe treatment options in CLE. However, deucravacitinib demonstrated superior efficacy and safety with fewer adverse events compared to anifrolumab. CLE patients who have shown an inadequate response to first- and second-line treatments may benefit from the incorporation of deucravacitinib into their treatment regimens.
Collapse
Affiliation(s)
- Laura Anna Bokor
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 41 Mária Street, Budapest 1085, Hungary; Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary
| | - Katalin Martyin
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 41 Mária Street, Budapest 1085, Hungary; Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary
| | - Máté Krebs
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 41 Mária Street, Budapest 1085, Hungary; Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary
| | - Noémi Ágnes Galajda
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 41 Mária Street, Budapest 1085, Hungary; Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary
| | - Fanni Adél Meznerics
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 41 Mária Street, Budapest 1085, Hungary; Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary
| | - Bence Szabó
- Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary; Division of Pancreatic Diseases, Heart and Vascular Centre, Semmelweis University, 25-29 Tömő Street, Budapest 1083, Hungary; Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti Street, Pécs 7624, Hungary
| | - Kende Lőrincz
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 41 Mária Street, Budapest 1085, Hungary; Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary
| | - Norbert Kiss
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 41 Mária Street, Budapest 1085, Hungary; Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary
| | - András Bánvölgyi
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 41 Mária Street, Budapest 1085, Hungary; Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary
| | - Bernadett Hidvégi
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 41 Mária Street, Budapest 1085, Hungary; Centre for Translational Medicine, Semmelweis University, 22 Baross Street, Budapest 1085, Hungary.
| |
Collapse
|
3
|
Jafari AJ, McGee C, Klimas N, Hebert AA. Monoclonal antibodies for the management of cutaneous lupus erythematosus: an update on the current treatment landscape. Clin Exp Dermatol 2025; 50:314-322. [PMID: 39243383 DOI: 10.1093/ced/llae374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/07/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024]
Abstract
Cutaneous lupus erythematosus (CLE) is a complex autoimmune disease often characterized by a multitude of skin findings. CLE is generally classified into three main categories: acute CLE, subacute CLE and chronic CLE. The current therapeutic guidelines for CLE include counselling patients on general measures and medication regimens. Treatment options include optimized photoprotection, avoidance of environmental triggers, corticosteroids, topical and systemic immunomodulators, and antimalarials. To date, no biologic medications (i.e. monoclonal antibodies, mAbs) are approved for CLE. The first mAb for the treatment of both systemic lupus erythematosus (SLE) and active lupus nephritis was belimumab, and was approved for these diseases in 2011 and 2020, respectively. Belimumab is a specific inhibitor of B-lymphocyte stimulator. Anifrolumab, a type I interferon receptor antagonist, was approved in 2021 for SLE. Other mAbs with different targets, including a novel biologic that inhibits blood dendritic cell antigen 2, are currently under investigation for CLE. This review will describe the general treatment landscape for CLE. Selected studies related to these various mAbs will be discussed, as well as their safety profiles and efficacies demonstrated in clinical trials. Biologic medications can potentially augment the number of treatment options for patients living with CLE.
Collapse
Affiliation(s)
- Alexander J Jafari
- Department of Dermatology, UTHealth McGovern Medical School, Houston, TX, USA
| | | | - Natasha Klimas
- Department of Dermatology, UTHealth McGovern Medical School, Houston, TX, USA
| | - Adelaide A Hebert
- Department of Dermatology, UTHealth McGovern Medical School, Houston, TX, USA
- Department of Pediatrics, UTHealth McGovern Medical School, Houston, TX, USA
- Department of Dermatology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
4
|
Eshaq AM, Flanagan TW, Hassan SY, Al Asheikh SA, Al-Amoudi WA, Santourlidis S, Hassan SL, Alamodi MO, Bendhack ML, Alamodi MO, Haikel Y, Megahed M, Hassan M. Non-Receptor Tyrosine Kinases: Their Structure and Mechanistic Role in Tumor Progression and Resistance. Cancers (Basel) 2024; 16:2754. [PMID: 39123481 PMCID: PMC11311543 DOI: 10.3390/cancers16152754] [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: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Protein tyrosine kinases (PTKs) function as key molecules in the signaling pathways in addition to their impact as a therapeutic target for the treatment of many human diseases, including cancer. PTKs are characterized by their ability to phosphorylate serine, threonine, or tyrosine residues and can thereby rapidly and reversibly alter the function of their protein substrates in the form of significant changes in protein confirmation and affinity for their interaction with protein partners to drive cellular functions under normal and pathological conditions. PTKs are classified into two groups: one of which represents tyrosine kinases, while the other one includes the members of the serine/threonine kinases. The group of tyrosine kinases is subdivided into subgroups: one of them includes the member of receptor tyrosine kinases (RTKs), while the other subgroup includes the member of non-receptor tyrosine kinases (NRTKs). Both these kinase groups function as an "on" or "off" switch in many cellular functions. NRTKs are enzymes which are overexpressed and activated in many cancer types and regulate variable cellular functions in response to extracellular signaling-dependent mechanisms. NRTK-mediated different cellular functions are regulated by kinase-dependent and kinase-independent mechanisms either in the cytoplasm or in the nucleus. Thus, targeting NRTKs is of great interest to improve the treatment strategy of different tumor types. This review deals with the structure and mechanistic role of NRTKs in tumor progression and resistance and their importance as therapeutic targets in tumor therapy.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA;
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Sofie-Yasmin Hassan
- Department of Pharmacy, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Simeon Santourlidis
- Institute of Cell Therapeutics and Diagnostics, University Medical Center of Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Maryam O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Marcelo L. Bendhack
- Department of Urology, Red Cross University Hospital, Positivo University, Rua Mauá 1111, Curitiba 80030-200, Brazil;
| | - Mohammed O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
5
|
Liu M, Wang S, Liang Y, Fan Y, Wang W. Genetic polymorphisms in genes involved in the type I interferon system (STAT4 and IRF5): association with Asian SLE patients. Clin Rheumatol 2024; 43:2403-2416. [PMID: 38963465 DOI: 10.1007/s10067-024-07046-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 05/21/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Systemic lupus erythematosus (SLE) is a common autoimmune disease with a polymorphic clinical presentation involving multisystem damages with significant differences in prevalence and disease severity among different ethnic groups. Although genetic, hormonal, and environmental factors have been demonstrated to contribute a lot to SLE, the pathogenesis of SLE is still unknown. Numerous evidence revealed that gene variants within the type I interferons (IFN) signaling pathway performed the great genetic associations with autoimmune diseases including SLE. To date, through genome-wide association studies (GWAS), genetic association studies showed that more than 100 susceptibility genes have been linked to the pathogenesis of SLE, among which TYK2, STAT1, STAT4, and IRF5 are important molecules directly connected to the type I interferon signaling system. The review summarized the genetic associations and the detailed risk loci of STAT4 and IRF5 with Asian SLE patients, explored the genotype distributions associated with the main clinical manifestations of SLE, and sorted out the potential reasons for the differences in susceptibility in Asia and Europe. Moreover, the therapies targeting STAT4 and IRF5 were also evaluated in order to propose more personalized and targeted treatment plans in SLE.
Collapse
Affiliation(s)
- Mengyao Liu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shenglong Wang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yujiao Liang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yongsheng Fan
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Weijie Wang
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| |
Collapse
|
6
|
Ezeh N, Vleugels RA, Shahriari N. Discoid lupus erythematosus successfully treated with deucravacitinib. JAAD Case Rep 2024; 49:59-61. [PMID: 38883165 PMCID: PMC11176598 DOI: 10.1016/j.jdcr.2024.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024] Open
Affiliation(s)
- Nnenna Ezeh
- Harvard Combined Dermatology Residency, Massachusetts General Hospital, Boston, Massachusetts
| | - Ruth Ann Vleugels
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Neda Shahriari
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
7
|
Pandey SP, Bhaskar R, Han SS, Narayanan KB. Autoimmune Responses and Therapeutic Interventions for Systemic Lupus Erythematosus: A Comprehensive Review. Endocr Metab Immune Disord Drug Targets 2024; 24:499-518. [PMID: 37718519 DOI: 10.2174/1871530323666230915112642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/05/2023] [Accepted: 07/22/2023] [Indexed: 09/19/2023]
Abstract
Systemic Lupus Erythematosus (SLE) or Lupus is a multifactorial autoimmune disease of multiorgan malfunctioning of extremely heterogeneous and unclear etiology that affects multiple organs and physiological systems. Some racial groups and women of childbearing age are more susceptible to SLE pathogenesis. Impressive progress has been made towards a better understanding of different immune components contributing to SLE pathogenesis. Recent investigations have uncovered the detailed mechanisms of inflammatory responses and organ damage. Various environmental factors, pathogens, and toxicants, including ultraviolet light, drugs, viral pathogens, gut microbiome metabolites, and sex hormones trigger the onset of SLE pathogenesis in genetically susceptible individuals and result in the disruption of immune homeostasis of cytokines, macrophages, T cells, and B cells. Diagnosis and clinical investigations of SLE remain challenging due to its clinical heterogeneity and hitherto only a few approved antimalarials, glucocorticoids, immunosuppressants, and some nonsteroidal anti-inflammatory drugs (NSAIDs) are available for treatment. However, the adverse effects of renal and neuropsychiatric lupus and late diagnosis make therapy challenging. Additionally, SLE is also linked to an increased risk of cardiovascular diseases due to inflammatory responses and the risk of infection from immunosuppressive treatment. Due to the diversity of symptoms and treatment-resistant diseases, SLE management remains a challenging issue. Nevertheless, the use of next-generation therapeutics with stem cell and gene therapy may bring better outcomes to SLE treatment in the future. This review highlights the autoimmune responses as well as potential therapeutic interventions for SLE particularly focusing on the recent therapeutic advancements and challenges.
Collapse
Affiliation(s)
- Surya Prakash Pandey
- Aarogya Institute of Healthcare and Research, Jaipur, Rajasthan, 302033, India
- Department of Zoology, School of Science, IFTM University, Moradabad, Uttar Pradesh, 244102, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| | - Kannan Badri Narayanan
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| |
Collapse
|
8
|
McCormick ET, Draganski A, Chalmers S, Zahn J, Garcia S, Nussbaum D, Friedman A, Putterman C, Friedman J. Nano-encapsulated anandamide reduces inflammatory cytokines in vitro and lesion severity in a murine model of cutaneous lupus erythematosus. Exp Dermatol 2023; 32:2072-2083. [PMID: 37726950 DOI: 10.1111/exd.14935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023]
Abstract
Cutaneous lupus erythematosus (CLE) is a heterogeneous autoimmune skin disease which occurs independently and in conjunction with systemic lupus erythematosus. Drug development for CLE is severely lacking. Anandamide (AEA) is a primary endocannabinoid which exhibits immunomodulatory effects through mixed cannabinoid receptor agonism. We evaluated AEA as topical treatment for CLE and assessed benefits of nanoparticle encapsulation (AEA-NP) on cutaneous drug penetration, delivery and biological activity. Compared to untreated controls, AEA-NP decreased IL-6 and MCP-1 in UVB-stimulated keratinocytes (p < 0.05) in vitro. In BALB/c mice, AEA-NP displayed improved cutaneous penetration, extended release and persistence of AEA in the follicular unit extending to the base after 24 h. Utilizing the MRL-lpr lupus murine model, twice weekly treatment of lesions with topical AEA-NP for 10 weeks led to decreased clinical and histologic lesion scores compared to unencapsulated AEA and untreated controls (p < 0.05). Prophylactic application of AEA-NP to commonly involved areas on MRL-lpr mice similarly resulted in decreased clinical and histologic scores when compared to controls (p < 0.05), and reduced C3 and IBA-1 in lesional tissue (p < 0.05). The demonstrated clinical and immunomodulatory effects of treatment with AEA support its potential as therapy for CLE. This work also suggests that encapsulation of AEA improves penetration and treatment efficacy. Future studies will be conducted to assess full therapeutic potential.
Collapse
Affiliation(s)
- Erika T McCormick
- George Washington University Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | | | - Samantha Chalmers
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, Bronx, USA
| | - Joseph Zahn
- George Washington University Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Sayra Garcia
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, Bronx, USA
| | - Dillon Nussbaum
- George Washington University Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Adam Friedman
- George Washington University Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Chaim Putterman
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, Bronx, USA
- Azrieli Faculty of Medicine of Bar-Ilan University, Zefat, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
| | - Joel Friedman
- Division of Rheumatology, Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, Bronx, USA
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
Fernandez-Ruiz R, Niewold TB. Type I Interferons in Autoimmunity. J Invest Dermatol 2022; 142:793-803. [PMID: 35016780 PMCID: PMC8860872 DOI: 10.1016/j.jid.2021.11.031] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/30/2022]
Abstract
Dysregulated IFN-1 responses play crucial roles in the development of multiple forms of autoimmunity. Many patients with lupus, systemic sclerosis, Sjogren's syndrome, and dermatomyositis demonstrate enhanced IFN-1 signaling. IFN-1 excess is associated with disease severity and autoantibodies and could potentially predict response to newer therapies targeting IFN-1 pathways. In this review, we provide an overview of the signaling pathway and immune functions of IFN-1s in health and disease. We also review the systemic autoimmune diseases classically associated with IFN-1 upregulation and current therapeutic strategies targeting the IFN-1 system.
Collapse
Affiliation(s)
- Ruth Fernandez-Ruiz
- Division of Rheumatology, Department of Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Timothy B Niewold
- Judith & Stewart Colton Center for Autoimmunity, Department of Medicine Research, NYU Grossman School of Medicine, New York, New York, USA.
| |
Collapse
|
15
|
Tan G, Baby B, Zhou Y, Wu T. Emerging Molecular Markers Towards Potential Diagnostic Panels for Lupus. Front Immunol 2022; 12:808839. [PMID: 35095896 PMCID: PMC8792845 DOI: 10.3389/fimmu.2021.808839] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/22/2021] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease which can affect various tissues and organs, posing significant challenges for clinical diagnosis and treatment. The etiology of SLE is highly complex with contributions from environmental factors, stochastic factors as well as genetic susceptibility. The current criteria for diagnosing SLE is based primarily on a combination of clinical presentations and traditional lab testing. However, these tests have suboptimal sensitivity and specificity. They are unable to indicate disease cause or guide physicians in decision-making for treatment. Therefore, there is an urgent need to develop a more accurate and robust tool for effective clinical management and drug development in lupus patients. It is fortunate that the emerging Omics have empowered scientists in the discovery and identification of potential novel biomarkers of SLE, especially the markers from blood, urine, cerebrospinal fluids (CSF), and other bodily fluids. However, many of these markers have not been carefully validated for clinical use. In addition, it is apparent that individual biomarkers lack sensitivity or specificity. This review summarizes the sensitivity, specificity and diagnostic value of emerging biomarkers from recent studies, and discusses the potential of these markers in the development of biomarker panel based diagnostics or disease monitoring system in SLE.
Collapse
Affiliation(s)
- Gongjun Tan
- Department of Clinical Laboratory, Zhuhai Maternal and Child Healthcare Hospital, Zhuhai, China
| | - Binila Baby
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Yuqiu Zhou
- Department of Clinical Laboratory, Zhuhai Maternal and Child Healthcare Hospital, Zhuhai, China
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| |
Collapse
|
16
|
Immunogenetics of Lupus Erythematosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:213-257. [DOI: 10.1007/978-3-030-92616-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
17
|
Gao ZY, Su LC, Wu QC, Sheng JE, Wang YL, Dai YF, Chen AP, He SS, Huang X, Yan GQ. Bioinformatics analyses of gene expression profile identify key genes and functional pathways involved in cutaneous lupus erythematosus. Clin Rheumatol 2021; 41:437-452. [PMID: 34553293 DOI: 10.1007/s10067-021-05913-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/05/2021] [Accepted: 09/05/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Lupus erythematosus is an autoimmune disease that causes damage to multiple organs ranging from skin lesions to systemic manifestations. Cutaneous lupus erythematosus (CLE) is a common type of lupus erythematosus (LE), but its molecular mechanisms are currently unknown. The study aimed to explore changes in the gene expression profiles and identify key genes involved in CLE, hoping to uncover its molecular mechanism and identify new targets for CLE. METHOD We analyzed the microarray dataset (GSE109248) derived from the Gene Expression Omnibus (GEO) database, which was a transcriptome profiling of CLE cutaneous lesions. The differentially expressed genes (DEGs) were identified, and the functional annotation of DEGs was performed with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Protein-protein interaction (PPI) network was also constructed to identify hub genes involved in CLE. RESULT A total of 755 up-regulated DEGs and 405 down-regulated DEGs were identified. GO enrichment analysis showed that defense response to virus, immune response, and type I interferon signaling pathway were the most significant enrichment items in DEGs. The KEGG pathway analysis identified 51 significant enrichment pathways, which mainly included systemic lupus erythematosus, osteoclast differentiation, cytokine-cytokine receptor interaction, and primary immunodeficiency. Based on the PPI network, the study identified the top 10 hub genes involved in CLE, which were CXCL10, CCR7, FPR3, PPARGC1A, MMP9, IRF7, IL2RG, SOCS1, ISG15, and GSTM3. By comparison between subtypes, the results showed that ACLE had the least DEGs, while CCLE showed the most gene and functional changes. CONCLUSION The identified hub genes and functional pathways found in this study may expand our understanding on the underlying pathogenesis of CLE and provide new insights into potential biomarkers or targets for the diagnosis and treatment of CLE. Key Points • The bioinformatics analysis based on CLE patients and healthy controls was performed and 1160 DEGs were identified • The 1160 DEGs were mainly enriched in biological processes related to immune responses, including innate immune response, type I interferon signaling pathway, interferon-γ-mediated signaling pathway, positive regulation of T cell proliferation, regulation of immune response, antigen processing, and presentation via MHC class Ib and so on • KEGG pathway enrichment analysis indicated that DEGs were mainly enriched in several immune-related diseases and virus infection, including systemic lupus erythematosus, primary immunodeficiency, herpes simplex infection, measles, influenza A, and so on • The hub genes such as CXCL10, IRF7, MMP9, CCR7, and SOCS1 may become new markers or targets for the diagnosis and treatment of CLE.
Collapse
Affiliation(s)
- Zhen-Yu Gao
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China. .,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.
| | - Lin-Chong Su
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Qing-Chao Wu
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Jiao-E Sheng
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Yun-Long Wang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Yu-Fang Dai
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - An-Ping Chen
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - San-Shan He
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Xia Huang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| | - Guo-Qing Yan
- Department of Rheumatology and Immunology, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China.,Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, The Affiliated Hospital of Hubei Minzu University, Enshi, Hubei Province, China
| |
Collapse
|
18
|
Gonciarz M, Pawlak-Buś K, Leszczyński P, Owczarek W. TYK2 as a therapeutic target in the treatment of autoimmune and inflammatory diseases. Immunotherapy 2021; 13:1135-1150. [PMID: 34235974 DOI: 10.2217/imt-2021-0096] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
JAKs are intracellular protein tyrosine kinases that, through activation of STATs, are responsible for signal transduction pathways that regulate cellular responses to numerous cytokines, growth factors and hormones in many different cells. JAK-STAT signaling plays a key role in regulating immune function, and cytokines - such as IL-23, IL-12 and type I interferons - are central to the pathogenesis of autoimmune diseases, including psoriasis, inflammatory bowel disease and systemic lupus erythematosus. Here the authors review the evidence for targeting TYK2 as a more specific approach to treating these conditions. TYK2 inhibitors are clinically effective in autoimmune and inflammatory diseases and may avoid some of the complications reported with nonselective JAK inhibitors.
Collapse
Affiliation(s)
- Maciej Gonciarz
- Department of Gastroenterology & Internal Medicine, Military Institute of Medicine, Warsaw, Poland
| | - Katarzyna Pawlak-Buś
- Department of Rheumatology, Rehabilitation & Internal Medicine, University of Medical Sciences, Poznan, Poland
| | - Piotr Leszczyński
- Department of Rheumatology, Rehabilitation & Internal Medicine, University of Medical Sciences, Poznan, Poland
| | - Witold Owczarek
- Department of Dermatology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
19
|
Pellenz FM, Dieter C, Lemos NE, Bauer AC, Souza BMD, Crispim D. Association of TYK2 polymorphisms with autoimmune diseases: A comprehensive and updated systematic review with meta-analysis. Genet Mol Biol 2021; 44:e20200425. [PMID: 33949620 PMCID: PMC8097517 DOI: 10.1590/1678-4685-gmb-2020-0425] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/09/2021] [Indexed: 12/05/2022] Open
Abstract
Autoimmune diseases are characterized by the loss of self-tolerance, leading to
immune-mediated tissue destruction and chronic inflammation. Tyrosine kinase 2
(TYK2) protein plays a key role in immunity and apoptosis pathways. Studies have
reported associations between single nucleotide polymorphisms (SNPs) in the
TYK2 gene and autoimmune diseases; however, results are
still inconclusive. Thus, we conducted a systematic review followed by
meta-analysis. A literature search was performed to find studies that
investigated associations between TYK2 SNPs and autoimmune
diseases (multiple sclerosis, systemic lupus erythematosus, Crohn’s disease,
ulcerative colitis, psoriasis, rheumatoid arthritis, type 1 diabetes, and
inflammatory bowel disease). Pooled odds ratios (OR) with 95 % CI were
calculated using random (REM) or fixed (FEM) effects models in the Stata 11.0
Software. Thirty-four articles were eligible for inclusion in the meta-analyses,
comprising 9 different SNPs: rs280496, rs280500, rs280523, rs280519, rs2304256,
rs12720270, rs12720356, rs34536443, and rs35018800. Meta-analysis results showed
the minor alleles of rs2304256, rs12720270, rs12720356, rs34536443, and
rs35018800 SNPs were associated with protection against autoimmune diseases.
Moreover, the A allele of the rs280519 SNP was associated with risk for systemic
lupus erythematosus. Our meta-analyses demonstrated that the rs2304256,
rs12720270, rs12720356, rs34536443, rs35018800, and rs280519 SNPs in the
TYK2 gene are associated with different autoimmune
diseases.
Collapse
Affiliation(s)
- Felipe Mateus Pellenz
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| | - Cristine Dieter
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| | - Natália Emerim Lemos
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| | - Andrea Carla Bauer
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil.,Hospital de Clínicas de Porto Alegre, Serviço de Nefrologia, Porto Alegre, RS, Brazil
| | - Bianca Marmontel de Souza
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| | - Daisy Crispim
- Hospital de Clínicas de Porto Alegre, Serviço de Endocrinologia, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| |
Collapse
|
20
|
Proteome study of cutaneous lupus erythematosus (CLE) and dermatomyositis skin lesions reveals IL-16 is differentially upregulated in CLE. Arthritis Res Ther 2021; 23:132. [PMID: 33931094 PMCID: PMC8086067 DOI: 10.1186/s13075-021-02511-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 04/12/2021] [Indexed: 11/30/2022] Open
Abstract
Background The objective of the study was to explore the disease pathways activated in the inflammatory foci of skin lesions in cutaneous lupus erythematosus (CLE) and dermatomyositis (DM). Methods Skin biopsies acquired from active CLE and DM lesions, patient (PC), and also healthy controls (HC) were investigated. Biopsy sections were examined by a pathologist, inflammatory foci were laser micro-dissected and captured, and proteins within captured tissue were detected in an unbiased manner by mass spectrometry. Protein pathway analysis was performed by the string-db.org platform. Findings of interest were confirmed by immunohistochemistry (IHC). Results Proteome investigation identified abundant expression of interferon-regulated proteins (IRP) as a common feature of CLE and DM. Interleukin (IL)-16 was the only abundant cytokine differentially expressed in CLE compared to DM. Caspase-3, an enzyme that cleaves IL-16 into its active form, was detected in low levels. Significantly higher proportion of IL-16- and caspase-3-positive cells was identified in CLE lesions in comparison with DM, PC, and HC. Proteomic results indicate more abundant complement deposition in CLE skin lesions. Conclusions Using unbiased mass spectrometry investigation of CLE and DM inflammatory infiltrates, we confirmed that high IRP expression is a common feature of both CLE and DM, while IL-16 is the only differentially expressed cytokine in CLE. IHC confirmed high expression of IL-16 and caspase-3 in CLE. Our novel molecular findings indicate that IL-16 detection could be useful in differential diagnostics between the two conditions that can display similar histopathological appearance. IL-16 could be of interest as a future therapeutic target for CLE.
Collapse
|
21
|
Patel J, Borucki R, Werth VP. An Update on the Pathogenesis of Cutaneous Lupus Erythematosus and Its Role in Clinical Practice. Curr Rheumatol Rep 2020; 22:69. [PMID: 32845411 DOI: 10.1007/s11926-020-00946-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Understanding the pathogenesis of cutaneous lupus erythematosus (CLE) is an important step in developing new medications and providing effective treatment to patients. This review focuses on novel research within CLE pathogenesis, as well as some of the medications being developed based on this knowledge. RECENT FINDINGS The subtle differences between systemic lupus erythematosus (SLE) and CLE pathogenesis are highlighted by differences in the circulating immune cells found in each disease, as well as the specific pathways activated by ultraviolet light. Plasmacytoid dendritic cells and the related type I interferon pathway are major components of CLE pathogenesis, and as such, therapies targeting components of this pathway have been successful in recent clinical trials. B cell-depleting therapies have shown success in SLE; however, their role in CLE is less clear. Understanding the differences between these manifestations of lupus allows for the development of therapies that are more effective in skin-specific disease. Discovering key pathways in CLE pathogenesis is critical for understanding the clinical features of the disease and ultimately developing new and effective therapies.
Collapse
Affiliation(s)
- Jay Patel
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert Borucki
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, USA.,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Victoria P Werth
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, USA. .,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. .,Department of Dermatology, Perelman Center for Advanced Medicine, Suite 1-330A, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
| |
Collapse
|
22
|
Garelli CJ, Refat MA, Nanaware PP, Ramirez-Ortiz ZG, Rashighi M, Richmond JM. Current Insights in Cutaneous Lupus Erythematosus Immunopathogenesis. Front Immunol 2020; 11:1353. [PMID: 32714331 PMCID: PMC7343764 DOI: 10.3389/fimmu.2020.01353] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/27/2020] [Indexed: 12/25/2022] Open
Abstract
Cutaneous Lupus Erythematosus (CLE) is a clinically diverse group of autoimmune skin diseases with shared histological features of interface dermatitis and autoantibodies deposited at the dermal-epidermal junction. Various genetic and environmental triggers of CLE promote infiltration of T cells, B cells, neutrophils, antigen presenting cells, and NK cells into lesional skin. In this mini-review, we will discuss the clinical features of CLE, insights into CLE immunopathogenesis, and novel treatment approaches.
Collapse
Affiliation(s)
- Colton J. Garelli
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Maggi Ahmed Refat
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Padma P. Nanaware
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Zaida G. Ramirez-Ortiz
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Mehdi Rashighi
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Jillian M. Richmond
- Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
23
|
Kahlenberg JM. Rethinking the Pathogenesis of Cutaneous Lupus. J Invest Dermatol 2020; 141:32-35. [PMID: 32605817 DOI: 10.1016/j.jid.2020.05.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 10/24/2022]
Abstract
Knowledge of the etiology of cutaneous lupus is rapidly evolving. Dissection of the pathologic events in lesional skin has led to knowledge of important cell populations and transcriptional changes contributing to disease. Recently, the study of nonlesional skin in patients with systemic lupus has also identified key abnormalities that likely contribute to a propensity for inflammation. These include an elevated type I IFN signature, overproduction of IFNs, and an absence of Langerhans cells. These changes promote aberrant inflammation in response to known triggers of disease, such as UV light. Further research will undoubtedly accelerate our understanding of this disfiguring disease.
Collapse
Affiliation(s)
- J Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Lupus erythematosus (LE) is characterized by broad and varied clinical forms ranging from a localized skin lesion to a life-threatening form with severe systemic manifestations. The overlapping between cutaneous LE (CLE) and systemic LE (SLE) brings difficulties to physicians for early accurate diagnosis and sometimes may lead to delayed treatment for patients. We comprehensively review recent progress about the similarities and differences of the main three subsets of LE in pathogenesis and immunological mechanisms, with a particular focus on the skin damage. RECENT FINDINGS Recent studies on the mechanisms contributing to the skin damage in lupus have shown a close association of abnormal circulating inflammatory cells and abundant production of IgG autoantibodies with the skin damage of SLE, whereas few evidences if serum autoantibodies and circulating inflammatory cells are involved in the pathogenesis of CLE, especially for the discoid LE (DLE). Till now, the pathogenesis and molecular/cellular mechanism for the progress from CLE to SLE are far from clear. But more and more factors correlated with the differences among the subsets of LE and progression from CLE to SLE have been found, such as the mutation of IRF5, IFN regulatory factors and abnormalities of plasmacytoid dendritic cells (PDCs), Th1 cells, and B cells, which could be the potential biomarkers for the interventions in the development of LE. A further understanding in pathogenesis and immunological mechanisms for skin damage in different subsets of LE makes us think more about the differences and cross-links in the pathogenic mechanism of CLE and SLE, which will shed a light in predictive biomarkers and therapies in LE.
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW The review provides an update on the diagnosis, pathogenesis, and treatment of cutaneous lupus erythematosus (CLE). RECENT FINDINGS Diagnostic challenges exist in better defining CLE as an independent disease distinct from systemic lupus erythematosus with cutaneous features and further classifying CLE based on clinical, histological, and laboratory features. Recent mechanistic studies revealed more genetic variations, environmental triggers, and immunologic dysfunctions that are associated with CLE. Drug induction specifically has emerged as one of the most important triggers for CLE. Treatment options include topical agents and systemic therapies, including newer biologics such as belimumab, rituximab, ustekinumab, anifrolumab, and BIIB059 that have shown good clinical efficacy in trials. CLE is a group of complex and heterogenous diseases. Future studies are warranted to better define CLE within the spectrum of lupus erythematosus. Better insight into the pathogenesis of CLE could facilitate the design of more targeted therapies.
Collapse
Affiliation(s)
- Amy J Petty
- School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Lauren Floyd
- Department of Dermatology, Duke University, Durham, NC, 27710, USA
| | | | | |
Collapse
|
26
|
Carrozzo M, Porter S, Mercadante V, Fedele S. Oral lichen planus: A disease or a spectrum of tissue reactions? Types, causes, diagnostic algorhythms, prognosis, management strategies. Periodontol 2000 2019; 80:105-125. [PMID: 31090143 DOI: 10.1111/prd.12260] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oral lichen planus and lichenoid lesions comprise a group of disorders of the oral mucosa that likely represent a common reaction pattern to 1 or more unknown antigens. The coexistence of hyperkeratotic striation/reticulation, varying degrees of mucosal inflammation from mild erythema to severe widespread ulceration, and a band-like infiltrate of mononuclear inflammatory cells including activated T lymphocytes, macrophages, and dendritic cells, are considered suggestive of oral lichen planus and lichenoid lesions. Several classification systems of oral lichen planus and lichenoid lesions have been attempted, although none seem to be comprehensive. In this paper, we present a classification of oral lichen planus and lichenoid lesions that includes oral lichen planus, oral lichenoid contact lesions, oral lichenoid drug reactions, oral lichenoid lesions of graft vs. host disease, discoid lupus erythematosus, and systemic lupus erythematosus, lichen planus-like variant of paraneoplastic pemphigus/paraneoplastic autoimmune multiorgan syndrome, chronic ulcerative stomatitis, lichen planus pemphigoides, solitary fixed drug eruptions, and lichen sclerosus. We present the clinical and diagnostic aspects of oral lichen planus and lichenoid lesions, and discuss related treatment options.
Collapse
Affiliation(s)
- Marco Carrozzo
- Centre for Oral Health Research, Oral Medicine Department, Newcastle University, Newcastle upon Tyne, UK
| | - Stephen Porter
- UCL Eastman Dental Institute, University College London, London, UK
| | | | - Stefano Fedele
- UCL Eastman Dental Institute, University College London, London, UK.,NIHR University College London Hospitals Biomedical Research Centre, London, UK
| |
Collapse
|
27
|
Catalytically Impaired TYK2 Variants are Protective Against Childhood- and Adult-Onset Systemic Lupus Erythematosus in Mexicans. Sci Rep 2019; 9:12165. [PMID: 31434951 PMCID: PMC6704113 DOI: 10.1038/s41598-019-48451-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 08/06/2019] [Indexed: 01/30/2023] Open
Abstract
Type I interferon (IFN-I) pathway plays a central role in the systemic lupus erythematosus (SLE) pathogenesis. Recent data suggest that SLE is associated with variants in IFN-I genes, such as tyrosine kinase 2 (TYK2), which is crucial in anti-viral immunity. Here, five TYK2 single nucleotide polymorphisms (SNPs) were genotyped in 368 childhood-onset SLE Mexican patients and 516 sex-matched healthy controls. Allele frequencies were also estimated in four indigenous groups. SLE protection was associated with TYK2 risk infection variants affecting residually its catalytic domain, rs12720356 (OR = 0.308; p = 0.041) and rs34536443 (OR = 0.370; p = 0.034), but not with rs2304256, rs12720270, and rs280500. This association was replicated in a 506 adult-onset SLE patients sample (OR = 0.250; p = 0.005, and OR = 0.277; p = 0.008, respectively). The minor alleles of both associated SNPs had a lower frequency in Mestizos than in Spaniards and were absent or rare in indigenous, suggesting that the presence of these alleles in the Mexican Mestizo population was derived from the Spaniards. For the first time, we report genetic variants with a protective effect in childhood- and adult-onset SLE Mexican population. Our results suggest that the frequency of IFN-I alleles associated with SLE, may have been shaped in populations exposed to infectious diseases for long periods, and this could be an explanation why Native American ancestry is associated with a higher SLE prevalence and an earlier onset.
Collapse
|
28
|
Cutaneous lupus erythematosus: new insights into pathogenesis and therapeutic strategies. Nat Rev Rheumatol 2019; 15:519-532. [PMID: 31399711 DOI: 10.1038/s41584-019-0272-0] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2019] [Indexed: 01/07/2023]
Abstract
Cutaneous lupus erythematosus (CLE) is an autoimmune disease that can present as an isolated skin disease or as a manifestation within the spectrum of systemic lupus erythematosus. The clinical spectrum of CLE is broad, ranging from isolated discoid plaques to widespread skin lesions. Histologically, skin lesions present as interface dermatitis (inflammation of the skin mediated by anti-epidermal responses), which is orchestrated by type I and type III interferon-regulated cytokines and chemokines. Both innate and adaptive immune pathways are strongly activated in the formation of skin lesions owing to continuous re-activation of innate pathways via pattern recognition receptors (PRRs). These insights into the molecular pathogenesis of skin lesions in CLE have improved our understanding of the mechanisms underlying established therapies and have triggered the development of targeted treatment strategies that focus on immune cells (for example, B cells, T cells or plasmacytoid dendritic cells), as well as immune response pathways (for example, PRR signalling, Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signalling and nuclear factor-κB signalling) and their cytokines and chemokines (for example, type I interferons, CXC-chemokine ligand 10 (CXCL10), IL-6 and IL-12).
Collapse
|
29
|
Jarrett P, Werth VP. A review of cutaneous lupus erythematosus: improving outcomes with a multidisciplinary approach. J Multidiscip Healthc 2019; 12:419-428. [PMID: 31213824 PMCID: PMC6549666 DOI: 10.2147/jmdh.s179623] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/03/2019] [Indexed: 12/20/2022] Open
Abstract
Cutaneous lupus erythematosus encompasses a spectrum of cutaneous disease with different phenotypes, and it potentially intersects with many disciplines in medicine. This review examines the epidemiology, clinical subtypes, pathology, psychology and intervention options for this disorder. It is important to understand the psychological distress that cutaneous lupus can cause and if needed actively investigate this possibility with the patient. Careful liaison between disciplines will achieve the optimum outcome.
Collapse
Affiliation(s)
- Paul Jarrett
- Department of Dermatology, Middlemore Hospital, Auckland, New Zealand.,Department of Medicine, The University of Auckland, Auckland, New Zealand
| | - Victoria P Werth
- Corporal Michael J. Crescenz Veteran Affairs Medical Center, Philadelphia, PA, USA.,Department of Dermatology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
30
|
Essential Kinases and Transcriptional Regulators and Their Roles in Autoimmunity. Biomolecules 2019; 9:biom9040145. [PMID: 30974919 PMCID: PMC6523499 DOI: 10.3390/biom9040145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022] Open
Abstract
Kinases and transcriptional regulators are fundamental components of cell signaling that are expressed on many types of immune cells which are involved in secretion of cytokines, cell proliferation, differentiation, and apoptosis. Both play important roles in biological responses in health as well as in illnesses such as the autoimmune diseases which comprise at least 80 disorders. These diseases are caused by complex genetic and environmental interactions that lead to a breakage of immunologic tolerance and a disruption of the balance between self-reactive cells and regulatory cells. Kinases or transcriptional regulatory factors often have an abnormal expression in the autoimmune cells that participate in the pathogenesis of autoimmune disease. These abnormally expressed kinases or transcriptional regulators can over-activate the function of self-reactive cells to produce inflammatory cytokines or down-regulate the activity of regulatory cells, thus causing autoimmune diseases. In this review we introduce five kinds of kinase and transcriptional regulator related to autoimmune diseases, namely, members of the Janus kinase (JAK) family (JAK3 and/or tyrosine kinase 2 (TYK2)), fork head box protein 3 (Foxp3), the retinoic acid-related orphan receptor gamma t (RORγt), and T-box expressed in T cells (T-bet) factors. We also provide a mechanistic insight into how these kinases and transcriptional regulators affect the function of the immune cells related to autoimmune diseases, as well as a description of a current drug design targeting these kinases and transcriptional regulators. Understanding their exact role helps offer new therapies for control of the inflammatory responses that could lead to clinical improvement of the autoimmune diseases.
Collapse
|
31
|
Gardet A, Pellerin A, McCarl CA, Diwanji R, Wang W, Donaldson D, Franchimont N, Werth VP, Rabah D. Effect of in vivo Hydroxychloroquine and ex vivo Anti-BDCA2 mAb Treatment on pDC IFNα Production From Patients Affected With Cutaneous Lupus Erythematosus. Front Immunol 2019; 10:275. [PMID: 30846987 PMCID: PMC6394354 DOI: 10.3389/fimmu.2019.00275] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 01/31/2019] [Indexed: 12/11/2022] Open
Abstract
Objective: Plasmacytoid dendritic cells (pDCs) are a major source of Type-I Interferon (IFN-I), a key driver in cutaneous lupus erythematosus (CLE). Currently evaluated in Phase II clinical trial, 24F4A (BIIB059) is an antibody targeting BDCA2, an inhibitory receptor expressed on pDCs. Given that Hydroxychloroquine (HCQ), a widely-used CLE therapy, and 24F4A are both able to inhibit pDC-derived IFN-I production; this study aimed to determine whether 24F4A would show an additional inhibitory effect on pDC response after ex vivo or in vivo treatment with HCQ. Methods: The effect of 24F4A on pDC-derived IFNα was measured from peripheral blood mononuclear cells (PBMC) either from healthy donors in presence or absence of HCQ or from CLE patients clinically exposed to various levels of HCQ. TLR7, TLR7/8, and TLR9 agonists (ssRNA, R848, and CpG-A) were used for pDC stimulation. Results: PDCs were the only producers of IFNα in response to CpG-A, R848, and ssRNA stimulation in PBMC cultures. CLE patients with higher levels of blood HCQ showed lower ex vivo pDC responses to CpG-A, but not R848 or ssRNA. In contrast, 24F4A reduced the amount of IFNα produced by pDCs from CLE patients in response to all TLR agonists, irrespective of the blood HCQ level. Conclusion: Our findings reveal that clinically-relevant HCQ concentrations partially inhibit the pDC response to TLR9 and weakly affect the response to TLR7/8 stimulation. 24F4A robustly inhibits pDC responses even in the presence of HCQ, highlighting its unique potential to disrupt pDC disease relevant biology, which could provide additional therapeutic benefit for CLE patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Victoria P Werth
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Corporal Michael J. Crescenz VAMC, Philadelphia, PA, United States
| | | |
Collapse
|
32
|
Qiu H, Wu H, Chan V, Lau CS, Lu Q. Transcriptional and epigenetic regulation of follicular T-helper cells and their role in autoimmunity. Autoimmunity 2017; 50:71-81. [PMID: 28263097 DOI: 10.1080/08916934.2017.1284821] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Hong Qiu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China and
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China and
| | - Vera Chan
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong
| | - Chak-Sing Lau
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China and
| |
Collapse
|
33
|
Wu H, Zeng J, Yin J, Peng Q, Zhao M, Lu Q. Organ-specific biomarkers in lupus. Autoimmun Rev 2017; 16:391-397. [PMID: 28212922 DOI: 10.1016/j.autrev.2017.02.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/16/2017] [Indexed: 12/12/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex and highly heterogeneous disease, which affects multiple organs, including joints, skin, kidneys, heart, hematopoietic system, and nerve system. While the etiopathogenesis of SLE still remains unclear, genetic susceptibilities and aberrant epigenetic modifications are believed to be involved. For precision therapy, it is necessary to assess accurately and objectively organ involvements and disease activity, which is difficult by current clinical laboratory tests. Biomarkers, which are a biologic, genetic, epigenetic or a chemical characteristic and conveniently detectable, serve as measures of disease diagnosis, activity, prognosis, and manifestation prediction, thereby providing instruction for individualized therapy. In addition, biomarkers differ according to different manifestations, since the disease activity index and treatments vary significantly. For example, unlike other non-renal SLE, lupus nephritis requires significant immunosuppressive drugs. Over the past decades, the research on biomarkers in lupus has been strengthened and numerous promising biomarkers have been identified at levels of genomics, transcriptomics and proteomics. In this review, we summarize the conventional and novel biomarkers in the tissue-specific manner, and discuss their roles in specific organ diagnosis, future manifestation prediction, disease activity assessment and their correlation with histology results. By doing so, it aims to shed a light on individualized treatment.
Collapse
Affiliation(s)
- Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Jinrong Zeng
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Jinghua Yin
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Qiao Peng
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China.
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is the prototypic autoimmune condition, often affecting multiple organ systems, including the skin. Cutaneous lupus erythematosus (CLE) is distinct from SLE and may be skin limited or associated with systemic disease. Histopathologically, the hallmark of lupus-specific manifestations of SLE and CLE is an interface dermatitis. The cause of SLE and CLE is likely multifactorial and may include shared genetic factors. In this review, we will discuss the genetic findings related to the cutaneous manifestations of SLE and isolated CLE, with a particular focus on the lupus-specific CLE subtypes. RECENT FINDINGS Several major histocompatibility complex and nonmajor histocompatibility complex genetic polymorphisms have been identified which may contribute to the cutaneous manifestations of SLE and to CLE. Most of these genetic variants are associated with mechanisms attributed to the pathogenesis of SLE, including pathways involved in interferon and vitamin D regulation and ultraviolet light exposure. Although there is overlap between the genetic factors associated with SLE and CLE, there appear to be unique genetic factors specific for CLE. SUMMARY Improved understanding of the genetics of CLE may lead to the creation of targeted therapies, improving outcomes for patients with this challenging dermatologic condition.
Collapse
|
35
|
Hejazi EZ, Werth VP. Cutaneous Lupus Erythematosus: An Update on Pathogenesis, Diagnosis and Treatment. Am J Clin Dermatol 2016; 17:135-46. [PMID: 26872954 DOI: 10.1007/s40257-016-0173-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Cutaneous lupus erythematosus (CLE) includes a broad range of dermatologic manifestations, which may or may not be associated with systemic disease. Recent studies in this area continue to shape our understanding of this disease and treatment options. Epidemiologic studies have found an incidence of CLE of 4.30 per 100,000, which approaches similar analysis for systemic lupus erythematosus (SLE). Although there have been extensive efforts to define SLE, the classification of CLE and its subgroups remains a challenge. Currently, diagnosis relies on clinical and laboratory findings as well as skin histology. The Cutaneous Lupus Area and Severity Index™ (CLASI™) is a validated measure of disease activity and damage. CLE pathogenesis is multifactorial and includes genetic contributions as well as effects of ultraviolet (UV) light. Immune dysregulation and aberrant cell signaling pathways through cytokine cascades are also implicated. Patient education and avoidance of triggers are key to disease prevention. Antimalarials and topical steroids continue to be the standard of care; however, immunosuppressants, thalidomide analogs and monoclonal antibodies are possible systemic therapies for the treatment of recalcitrant disease.
Collapse
|
36
|
Genetic association study of systemic lupus erythematosus and disease subphenotypes in European populations. Clin Rheumatol 2016; 35:1161-8. [DOI: 10.1007/s10067-016-3235-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 03/11/2016] [Accepted: 03/12/2016] [Indexed: 01/10/2023]
|
37
|
Tang L, Wan P, Wang Y, Pan J, Wang Y, Chen B. Genetic association and interaction between the IRF5 and TYK2 genes and systemic lupus erythematosus in the Han Chinese population. Inflamm Res 2015; 64:817-24. [DOI: 10.1007/s00011-015-0865-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/17/2015] [Accepted: 08/04/2015] [Indexed: 10/23/2022] Open
|
38
|
Abstract
The pathophysiology of cutaneous lupus erythematosus (CLE) encompasses the complex interactions between genetics, the environment, and cells and their products. Recent data have provided enhanced understanding of these interactions and the mechanism by which they cause disease. A number of candidate genes have been identified which increase the risk of developing CLE. Ultraviolet radiation, the predominant environmental exposure associated with CLE, appears to initiate CLE lesion formation by inducing apoptosis, precipitating autoantigen presentation, and promoting cellular production of specific cytokines. Autoantibodies are a well-known entity in CLE, but their exact role remains unclear. Finally, cells ranging from native skin cells to innate and adaptive immune cells produce cytokines and other molecules and play specific roles in lesion formation and perpetuation. Native skin cells implicated in CLE include keratinocytes and endothelial cells. Innate immune cells crucial to CLE pathophysiology include dendritic cells and neutrophils. The primary adaptive immune cells thought to be involved include Th1 cells, Th17 cells, cytotoxic T cells, and invariant natural killer T cells. Though the pathophysiology of CLE has yet to be fully characterized, current research provides direction for future research and therapies.
Collapse
Affiliation(s)
- Jordan C Achtman
- Philadelphia VA Medical Center, 3900 Woodland Avenue, Philadelphia, PA, 19104, USA. .,Department of Dermatology, Perelman Center for Advanced Medicine, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
| | - Victoria P Werth
- Philadelphia VA Medical Center, 3900 Woodland Avenue, Philadelphia, PA, 19104, USA. .,Department of Dermatology, Perelman Center for Advanced Medicine, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
| |
Collapse
|
39
|
Robinson E, Werth V. The role of cytokines in the pathogenesis of cutaneous lupus erythematosus. Cytokine 2015; 73:326-34. [DOI: 10.1016/j.cyto.2015.01.031] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/24/2015] [Accepted: 01/28/2015] [Indexed: 02/07/2023]
|
40
|
Kunz M, König IR, Schillert A, Kruppa J, Ziegler A, Grallert H, Müller-Nurasyid M, Lieb W, Franke A, Ranki A, Panelius J, Koskenmies S, Hasan T, Kere J, Rönn AC, Simon JC, Schmidt E, Wenzel J, Tüting T, Landsberg J, Zeller T, Blankenberg S, Gläser R, Patsinakidis N, Kuhn A, Ibrahim SM. Genome-wide association study identifies new susceptibility loci for cutaneous lupus erythematosus. Exp Dermatol 2015; 24:510-5. [PMID: 25827949 DOI: 10.1111/exd.12708] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2015] [Indexed: 12/11/2022]
Abstract
Cutaneous lupus erythematosus (CLE) is a chronic autoimmune disease of the skin with typical clinical manifestations. Here, we genotyped 906 600 single nucleotide polymorphisms (SNPs) in 183 CLE cases and 1288 controls of Central European ancestry. Replication was performed for 13 SNPs in 219 case subjects and 262 controls from Finland. Association was particularly pronounced at 4 loci, all with genomewide significance (P < 5 × 10(-8) ): rs2187668 (PGWAS = 1.4 × 10(-12) ), rs9267531 (PGWAS = 4.7 × 10(-10) ), rs4410767 (PGWAS = 1.0 × 10(-9) ) and rs3094084 (PGWAS = 1.1 × 10(-9) ). All mentioned SNPs are located within the major histocompatibility complex (MHC) region of chromosome 6 and near genes of known immune functions or associations with other autoimmune diseases such as HLA-DQ alpha chain 1 (HLA-DQA1), MICA, MICB, MSH5, TRIM39 and RPP21. For example, TRIM39/RPP21 read through transcript is a known mediator of the interferon response, a central pathway involved in the pathogenesis of CLE and systemic lupus erythematosus (SLE). Taken together, this genomewide analysis of disease association of CLE identified candidate genes and genomic regions that may contribute to pathogenic mechanisms in CLE via dysregulated antigen presentation (HLA-DQA1), apoptosis regulation, RNA processing and interferon response (MICA, MICB, MSH5, TRIM39 and RPP21).
Collapse
Affiliation(s)
- Manfred Kunz
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany
| | - Inke R König
- Institut für Medizinische Biometrie und Statistik, und Zentrum für Klinische Studien, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Lübeck, Germany
| | - Arne Schillert
- Institut für Medizinische Biometrie und Statistik, und Zentrum für Klinische Studien, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Lübeck, Germany
| | - Jochen Kruppa
- Institut für Medizinische Biometrie und Statistik, und Zentrum für Klinische Studien, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Lübeck, Germany
| | - Andreas Ziegler
- Institut für Medizinische Biometrie und Statistik, und Zentrum für Klinische Studien, Universität zu Lübeck, Universitätsklinikum Schleswig-Holstein, Lübeck, Germany
| | - Harald Grallert
- Research unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,German Center for Diabetes Research, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Department of Medicine I, Ludwig Maximilian University Munich, Munich, Germany.,German Center for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
| | - Wolfgang Lieb
- Institute for Epidemiology and Biobank popgen, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Annamari Ranki
- Department of Dermatology and Allergology, Skin and Allergy Hospital, Helsinki University Central Hospital, Helsinki, Finland
| | - Jaana Panelius
- Department of Dermatology and Allergology, Skin and Allergy Hospital, Helsinki University Central Hospital, Helsinki, Finland
| | - Sari Koskenmies
- Department of Dermatology and Allergology, Skin and Allergy Hospital, Helsinki University Central Hospital, Helsinki, Finland
| | - Taina Hasan
- Department of Dermatology, Tampere University Central Hospital, University of Tampere, Tampere, Finland
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden.,Department of Medical Genetics, Folkhälsan Institute of Genetics, University of Helsinki, Helsinki, Finland
| | - Ann-Charlotte Rönn
- Clinical Research Center, Karolinska University Hospital, Huddinge, Sweden
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany
| | - Enno Schmidt
- Department of Dermatology, Allergology and Venereology, University of Schleswig-Holstein, Lübeck, Germany
| | - Joerg Wenzel
- Department of Dermatology and Allergy, University of Bonn, Bonn, Germany
| | - Thomas Tüting
- Department of Dermatology and Allergy, University of Bonn, Bonn, Germany
| | - Jennifer Landsberg
- Department of Dermatology and Allergy, University of Bonn, Bonn, Germany
| | - Tanja Zeller
- University Heart Center Hamburg, Clinic for General and Interventional Cardiology, German Center for Cardiovascular Research (DZHK), Hamburg, Germany
| | - Stefan Blankenberg
- University Heart Center Hamburg, Clinic for General and Interventional Cardiology, German Center for Cardiovascular Research (DZHK), Hamburg, Germany
| | - Regine Gläser
- Department of Dermatology and Allergology, University of Schleswig-Holstein, Kiel, Germany
| | - Nikolaos Patsinakidis
- Department of Dermatology, Venereology and Allergology, Ruhr-University of Bochum, Bochum, Germany
| | - Annegret Kuhn
- Division of Immunogenetics, German Cancer Research Center, Heidelberg, Germany
| | - Saleh M Ibrahim
- Department of Dermatology, Allergology and Venereology, University of Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
41
|
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that is characterized by the development of autoantibodies and immunologic attack of different organ systems, including the skin. This review aims to provide an overview of some of the pathogenic processes that may be important in the development of SLE, specifically cutaneous lupus erythematosus, and then illustrates how therapies might be tailored to modify these processes and treat disease.
Collapse
Affiliation(s)
- Mark G Kirchhof
- Department of Dermatology and Skin Science, University of British Columbia, 835 West 10th Avenue, Vancouver, British Columbia V5Z 4E8, Canada
| | - Jan P Dutz
- Department of Dermatology and Skin Science, University of British Columbia, 835 West 10th Avenue, Vancouver, British Columbia V5Z 4E8, Canada; Child and Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, British Columbia V5Z 4H4, Canada.
| |
Collapse
|
42
|
Kubanov AA, Mineyeva AA. Molecular and genetic studies of the predisposition to the development of psoriasis among the population of the Russian Federation: a study of polymorphisms of TNFAIP3, TNIP1, TYK2 and REL genes. VESTNIK DERMATOLOGII I VENEROLOGII 2014. [DOI: 10.25208/0042-4609-2014-90-5-73-80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Goal. To study the role of polymorphic variants of genes encoding proteins of the signaling pathway of the nuclear transcription factor kappa-B, NF-kB (TNFAIP3 (rs610604), TNIP1 (rs17728338), TYK2 (rs12720356) and REL (rs702873)) in the predisposition to psoriasis among the Caucasian population of the Russian Federation. Materials and methods. The authors studied markers of the predisposition to the development of psoriasis such as TNFAIP3 (rs610604), TNIP1 (rs17728338), TYK2 (rs12720356) and REL (rs702873) using Real-Time PCR methods and allele-specific oligonucleotide probes and PFLP (Restriction Fragment Length Polymorphism) assay in whole blood samples taken from psoriatic patients (n = 301). To compare the frequency of genotypes, the authors used data obtained as a result of the genotyping of DNAs taken from healthy volunteers (n = 109). Results. The authors analyzed particular features of the distribution of polymorphic variants of TNFAIP3 (rs610604), TNIP1 (rs17728338), TYK2 (rs1272035) и REL (rs702873) genes in a sample of 301 psoriatic patients and 109 healthy volunteers from the Russian Federation. They revealed reliable differences in the frequency of registration of genotypes of these genes. The authors also determined genotypes considered as risk factors with regard to the development of psoriasis (TNFAIP3-A/C, TNIP1-A/G, TNIP1-A/A and TYK2-T/T) as well as protector genotypes (TNFAIP3-A/Ä, REL-A/A and TYK2-T/G)
Collapse
|
43
|
Dey-Rao R, Smith J, Chow S, Sinha A. Differential gene expression analysis in CCLE lesions provides new insights regarding the genetics basis of skin vs. systemic disease. Genomics 2014; 104:144-55. [DOI: 10.1016/j.ygeno.2014.06.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 06/15/2014] [Indexed: 01/06/2023]
|
44
|
Kreuter A, Lehmann P. Relevant new insights into the effects of photoprotection in cutaneous lupus erythematosus. Exp Dermatol 2014; 23:712-3. [PMID: 24910084 DOI: 10.1111/exd.12466] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2014] [Indexed: 11/27/2022]
Abstract
Cutaneous lupus erythematosus (CLE) denotes a heterogeneous spectrum of autoimmune diseases that primarily affect the skin. Ultraviolet irradiation (UV) is one of the most important environmental factors that can trigger skin lesions in CLE or even induce systemic organ manifestation. It has been shown that broad-spectrum sunscreen with high sun protection factors can effectively prevent UV-induced skin lesions in patients with different subtypes of CLE. In a recent issue of Experimental Dermatology, Zahn and colleagues demonstrate that potent photoprotection blocks disease-specific skin lesions in CLE patients by reducing lesional tissue damage and interferon-driven inflammation.
Collapse
Affiliation(s)
- Alexander Kreuter
- Department of Dermatology, Venereology and Allergology, HELIOS St. Elisabeth Hospital Oberhausen, Oberhausen, Germany
| | | |
Collapse
|
45
|
Shao WH, Cohen PL. The role of tyrosine kinases in systemic lupus erythematosus and their potential as therapeutic targets. Expert Rev Clin Immunol 2014; 10:573-82. [PMID: 24678775 DOI: 10.1586/1744666x.2014.893827] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The autoimmune disease systemic lupus erythematosus is characterized by loss of tolerance to nuclear antigens. Breakdown of tolerance is associated with alterations in T-cell and B-cell receptor signal transduction, including increased protein phosphorylation that may underlie pathogenesis and explain the characteristic hyperactivity of T and B cells and other immune cells in active disease. Tyrosine kinases play a central role in signaling processes in cells known to be important in the pathogenesis of autoimmune diseases. Considerable progress has been made in understanding the function of tyrosine kinases in immune cell signaling pathways. In this review, we will summarize the function of tyrosine kinases and their novel inhibitors from studies made in animal lupus models and systemic lupus erythematosus patients.
Collapse
Affiliation(s)
- Wen-Hai Shao
- Department of Medicine and Temple Autoimmunity Center, Section of Rheumatology, Temple University, Philadelphia, PA, USA
| | | |
Collapse
|
46
|
J DAS, C A, P SG, S C. Systemic Lupus Erythematosus: Old and New Susceptibility Genes versus Clinical Manifestations. Curr Genomics 2014; 15:52-65. [PMID: 24653663 PMCID: PMC3958959 DOI: 10.2174/138920291501140306113715] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/31/2013] [Accepted: 11/01/2013] [Indexed: 12/19/2022] Open
Abstract
Systemic Lupus Erythematosus (SLE) is one of the most relevant world-wide autoimmune disorders. The formation of autoantibodies and the deposition of antibody-containing immune complexes in blood vessels throughout the body is the main pathogenic mechanism of SLE leading to heterogeneous clinical manifestations and target tissue damage. The complexity of etiology and pathogenesis in SLE, enclosing genetic and environmental factors, apparently is one of the greatest challenges for both researchers and clinicians. Strong indications for a genetic background in SLE come from studies in families as well as in monozygotic and dizygotic twins, discovering several SLE-associated loci and genes (e.g. IRF5, PTPN22, CTLA4, STAT4 and BANK1). As SLE has a complex genetic background, none of these genes is likely to be entirely responsible for triggering autoimmune response in SLE even if they disclosure a potentially novel molecular mechanisms in the pathogenesis' disease. The clinical manifestations and disease severity varies greatly among patients, thus several studies try to associate clinical heterogeneity and prognosis with specific genetic polymorphisms in SLE associated genes. The continue effort to describe new predisposing or modulating genes in SLE is justified by the limited knowledge about the pathogenesis, assorted clinical manifestation and the possible prevention strategies. In this review we describe newly discovered, as well as the most studied genes associated to SLE susceptibility, and relate them to clinical manifestations of the disease.
Collapse
Affiliation(s)
- De Azevêdo Silva J
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Addobbati C
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco, Recife, Pernambuco, Brazil ; Department of Genetics, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Sandrin-Garcia P
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco, Recife, Pernambuco, Brazil ; Department of Genetics, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Crovella S
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco, Recife, Pernambuco, Brazil ; Department of Genetics, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| |
Collapse
|
47
|
Liang Y, Zhu Y, Xia Y, Peng H, Yang XK, Liu YY, Xu WD, Pan HF, Ye DQ. Therapeutic potential of tyrosine kinase 2 in autoimmunity. Expert Opin Ther Targets 2014; 18:571-80. [PMID: 24654603 DOI: 10.1517/14728222.2014.892925] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Tyrosine kinase 2 (Tyk2) is a Janus kinase family member that is crucial for signaling transduction in response to a wide variety of cytokines, including type I IFNs, IL-6, IL-10, IL-12 and IL-23. An appropriate expression of Tyk2-mediated signaling might be essential for maintaining normal immune responses. AREAS COVERED This review summarizes that Tyk2 is essential for the differentiation and function of a wide variety of immune cells, including natural killer cells, B cells, as well as T helper cells. In addition, Tyk2-mediated signaling promoted the production of autoimmune-associated components, which is implicated in the pathogenesis of autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis. Aberrant expression of Tyk2 was observed in many autoimmune conditions. EXPERT OPINION Until recently, no patent filings had claimed selective inhibitors of Tyk2. Both CP-690,500 and CMP6 failed to be used in clinical treatment due to the difficulties of finding suitable selective leads or due to detrimental toxicities. Although the result of Cmpd1 is promising, it remains to be seen how specific the Tyk2 inhibitor is and how they are working. Currently, structure-based drug design (SBDD) technology has provided us with a quite useful window for SBDD of Tyk2 inhibitors.
Collapse
Affiliation(s)
- Yan Liang
- Anhui Medical University, School of Public Health, Department of Epidemiology and Biostatistics , 81 Meishan Road, Hefei, Anhui, 230032 , PR China +86 551 65167726 ; +86 551 65161171 ;
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Genetics unquestionably contributes to systemic lupus erythematosus (SLE) predisposition, progression and outcome. Nevertheless, single-gene defects causing lupus-like phenotypes have been infrequently documented. The majority of the identified genetic SLE risk factors are, therefore, common variants, responsible for a small effect on the global risk. Recently, genome wide association studies led to the identification of a growing number of gene variants associated with SLE susceptibility, particular disease phenotypes, and antibody profiles. Further studies addressed the biological effects of these variants. In addition, the role of epigenetics has recently been revealed. These combined efforts contributed to a better understanding of SLE pathogenesis and to the characterization of clinically relevant pathways. In this review, we describe SLE-associated single-gene defects, common variants, and epigenetic changes. We also discuss the limitations of current methods and the challenges that we still have to face in order to incorporate genomic and epigenomic data into clinical practice.
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Cutaneous Lupus Erythematous (CLE) is an autoimmune disease in which patients may present with isolated skin findings or have CLE associated with underlying systemic disease. The most significant recent studies on its pathogenesis and therapeutic management are reviewed here. RECENT FINDINGS Patients with subacute and Discoid Lupus Erythematous had elevated Interferon score, about a third of all cases of SCLE could be attributed to previous drug exposure, and smoking may be more closely associated with CLE than Systemic Lupus Erythematous (SLE). An underlying genetic defect in some subsets of CLE patients may also be shared with SLE. Efficacy of antimalarial therapy is enhanced by increasing treatment duration or maintaining higher blood drug concentrations. Combination antimalarials that include quinacrine, thalidomide analogs, and Mycophenalate Mofetil may also be effective in refractory CLE. SUMMARY The pathogenesis of CLE remains unclear, and is likely multifactorial. Identified associations with subsets of CLE suggest future research questions in CLE pathogenesis. Subsets of CLE associated with interface dermatitis may share an underlying genetic defect in interferon signaling with SLE. The Cutaneous Lupus Disease Area and Severity Index is a valuable and widely used tool allowing standardized assessment and reporting of cutaneous disease activity and damage. More evidence is available to guide treatment of refractory CLE, but larger studies are needed. VIDEO ABSTRACT http://links.lww.com/COR/A4.
Collapse
|
50
|
Khudhur AS, Di Zenzo G, Carrozzo M. Oral lichenoid tissue reactions: diagnosis and classification. Expert Rev Mol Diagn 2014; 14:169-84. [DOI: 10.1586/14737159.2014.888953] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|