1
|
Saran A, Nishizaki D, Lippman SM, Kato S, Kurzrock R. Interleukin-17: A pleiotropic cytokine implicated in inflammatory, infectious, and malignant disorders. Cytokine Growth Factor Rev 2025:S1359-6101(25)00002-4. [PMID: 39875232 DOI: 10.1016/j.cytogfr.2025.01.002] [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: 12/05/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025]
Abstract
IL-17A, referred to as IL-17, is the founding member of a family of pro-inflammatory cytokines, including IL-17B, IL-17C, IL-17D, IL-17E (or IL-25), and IL-17F, which act via receptors IL-17RA to IL-17RE, and elicit potent cellular responses that impact diverse diseases. IL-17's interactions with various cytokines include forming a heterodimer with IL-17F and being stimulated by IL-23's activation of Th17 cells, which can lead to inflammation and autoimmunity. IL-17 is implicated in infectious diseases and inflammatory disorders such as rheumatoid arthritis and psoriasis, promoting neutrophil recruitment and anti-bacterial immunity, but potentially exacerbating fungal and viral infections, revealing its dual role as protective and pathologic. IL-17 is also involved in various cancers, including breast, colon, cervical, prostate, and skin cancer, contributing to proliferation, immune invasion, and metastases, but also playing a protective role in certain instances. Four FDA-approved drugs-secukinumab (for ankylosing spondylitis, enthesitis-related arthritis, hidradenitis suppurativa, non-radiographic axial spondyloarthritis, plaque psoriasis, and psoriatic arthritis), ixekizumab (for ankylosing spondylitis, non-radiographic axial spondyloarthritis, plaque psoriasis, and psoriatic arthritis), brodalumab (for plaque psoriasis), and bimekizumab (for plaque psoriasis)-suppress the IL-17 pathway, with more in development, including netakimab, sonelokimab, izokibep, and CJM112. These agents and others are being studied across a spectrum of disorders. Understanding the complicated interplay between IL-17 and other immune mediators may yield new treatments for inflammatory/autoimmune conditions and malignancies.
Collapse
Affiliation(s)
| | - Daisuke Nishizaki
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA; Center for Personalized Cancer Therapy, University of California, San Diego, La Jolla, CA, USA; Division of Hematology Oncology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Scott M Lippman
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA; Center for Personalized Cancer Therapy, University of California, San Diego, La Jolla, CA, USA; Division of Hematology Oncology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Shumei Kato
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA; Center for Personalized Cancer Therapy, University of California, San Diego, La Jolla, CA, USA; Division of Hematology Oncology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| | - Razelle Kurzrock
- MCW Cancer Center and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA; WIN Consortium, Paris, France; University of Nebraska, Lincoln, NE, USA.
| |
Collapse
|
2
|
Selvakumar B, Rah B, Jagal J, Sekar P, Moustafa R, Ramakrishnan RK, Haider M, Ibrahim SM, Samsudin R. Modulation of Plet1 expression by N-Acetylglucosamine through the IL-17 A-MAPK pathway in an imiquimod-induced psoriasis mouse model. Inflamm Res 2024; 73:2217-2230. [PMID: 39407062 DOI: 10.1007/s00011-024-01958-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 12/11/2024] Open
Abstract
Psoriasis (Ps) is a chronic inflammatory disorder marked by skin plaque formation, driven by immune dysregulation and genetic factors. Despite the available treatments, incidence of Ps is increasing in the dermatology patients. Novel strategies are crucial due to current treatment limitations. The interleukin 17 (IL-17) pathway is pivotal in Ps pathogenesis, however the expression of its putative target gene placenta expressed transcript 1 (Plet1) remains unstudied in Ps. Considering the potential anti-inflammatory properties of N-Acetylglucosamine (GlcNAc), our study explored its role in modulating Plet1 expression in an imiquimod (IMQ)-induced Ps mouse model. Our data demonstarted a significant reduction of inflammation and Psoriasis Area and Severity Index (PASI) scores, downregulation of growth factors (GFs), IL-17 A, and MAPK expression after GlcNAc treatment. In addition, GlcNAc treatment reduced neutrophils, monocyte-dendritic cells (Mo-DC) and conventional T cells (Tcons) while increasing monocyte-macrophages (Mo-Macs) and regulatory T cells (Tregs). GlcNAc treatment also downregulated Plet1 overexpression in psoriatic mouse skin and in vitro, reduced proliferation and apoptosis in IL-17 A stimulated human dermal fibroblasts (HDF), along with IL-17 A and TGF-β mRNA expression. Together, these data suggest that, GlcNAc interferes with downstream mechanisms in IL-17 pathway and downregulating Plet1 expression, presenting a promising strategy for Ps treatment.
Collapse
Affiliation(s)
- Balachandar Selvakumar
- Microbiota research group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| | - Bilal Rah
- Iron Biology research Group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Jayalakshmi Jagal
- Drug Delivery research group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Priyadarshini Sekar
- Microbiota research group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Raneem Moustafa
- Microbiota research group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Rakhee Kizhuvappat Ramakrishnan
- Tissue injury and repair research group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Mohamed Haider
- Drug Delivery research group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Saleh Mohamed Ibrahim
- College of Medicine and Health Science Research, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates
- Institute of Experimental Dermatology, University of Lübeck, 2562, Lübeck, Germany
| | - Rani Samsudin
- Microbiota research group, Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
- College of Dental Medicine, University of sharjah, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
3
|
Elkordy AA, Hill D, Attia M, Chaw CS. Liposomes and Their Therapeutic Applications in Enhancing Psoriasis and Breast Cancer Treatments. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1760. [PMID: 39513840 PMCID: PMC11547384 DOI: 10.3390/nano14211760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Psoriasis and breast cancer are two examples of diseases where associated inflammatory pathways within the body's immune system are implicated. Psoriasis is a complex, chronic and incurable inflammatory skin disorder that is primarily recognized by thick, scaly plaques on the skin. The most noticeable pathophysiological effect of psoriasis is the abnormal proliferation of keratinocytes. Breast cancer is currently the most diagnosed cancer and the leading cause of cancer-related death among women globally. While treatments targeting the primary tumor have significantly improved, preventing metastasis with systemic treatments is less effective. Nanocarriers such as liposomes and lipid nanoparticles have emerged as promising drug delivery systems for drug targeting and specificity. Advances in technologies and drug combinations have emerged to develop more efficient lipid nanocarriers to include more than one drug in combinational therapy to enhance treatment outcomes and/or relief symptoms for better patients' quality of life. Although there are FDA-approved liposomes with anti-cancer drugs for breast cancer, there are still unmet clinical needs to reduce the side effects associated with those nanomedicines. Hence, combinational nano-therapy may eliminate some of the issues and challenges. Furthermore, there are no nanomedicines yet clinically available for psoriasis. Hence, this review will focus on liposomes encapsulated single and/or combinational therapy to augment treatment outcomes with an emphasis on the effectiveness of combinational therapy within liposomal-based nanoparticulate drug delivery systems to tackle psoriasis and breast cancer. This review will also include an overview of both diseases, challenges in delivering drug therapy and the roles of nanomedicines as well as psoriasis and breast cancer models used for testing therapeutic interventions to pave the way for effective in vivo testing prior to the clinical trials.
Collapse
Affiliation(s)
- Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| | - David Hill
- School of Nursing and Health Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK;
| | - Mohamed Attia
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| | - Cheng Shu Chaw
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| |
Collapse
|
4
|
Varela Martins T, Silva de Melo BM, Toller-Kawahisa JE, da Silva GVL, Aníbal Silva CE, Paiva IM, Públio GA, Rosa MH, da Silva Souza C, Zamboni DS, Cunha FQ, Cunha TM, Ryffel B, Riteau N, Alves-Filho JC. The DNA sensor AIM2 mediates psoriasiform inflammation by inducing type 3 immunity. JCI Insight 2024; 9:e171894. [PMID: 39352743 PMCID: PMC11601563 DOI: 10.1172/jci.insight.171894] [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: 04/28/2023] [Accepted: 09/18/2024] [Indexed: 11/09/2024] Open
Abstract
Psoriasis is a chronic and recurrent inflammatory skin disease characterized by abnormal proliferation and differentiation of keratinocytes and activation of immune cells. However, the molecular driver that triggers this immune response in psoriatic skin remains unclear. The inflammation-related gene absent in melanoma 2 (AIM2) was identified as a susceptibility gene/locus associated with psoriasis. In this study, we investigated the role of AIM2 in the pathophysiology of psoriasis. We found elevated levels of mitochondrial DNA in patients with psoriasis, along with high expression of AIM2 in both the human psoriatic epidermis and a mouse model of psoriasis induced by topical imiquimod (IMQ) application. Genetic ablation of AIM2 reduced the development of IMQ-induced psoriasis by decreasing the production of type 3 cytokines (such as IL-17A and IL-23) and infiltration of immune cells into the inflammatory site. Furthermore, we demonstrate that IL-17A induced AIM2 expression in keratinocytes. Finally, the genetic absence of inflammasome components downstream AIM2, ASC, and caspase-1 alleviated IMQ-induced skin inflammation. Collectively, our data show that AIM2 is involved in developing psoriasis through its canonical activation.
Collapse
Affiliation(s)
- Timna Varela Martins
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
- Immune Health Laboratory, Regulation of host responses and immune health, IRL2029, French National Centre for Scientific Research (CNRS) and Ribeirão Preto Medical School (FMRP) of the Sao Paulo University (USP), Sao Paulo, Brazil
- INEM, CNRS, UMR7355 and University, Orleans, France
| | - Bruno Marcel Silva de Melo
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Juliana Escher Toller-Kawahisa
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Gabriel Victor Lucena da Silva
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Conceição Elidianne Aníbal Silva
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Isadora Marques Paiva
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Gabriel Azevedo Públio
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Marcos Henrique Rosa
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | | | - Dario Simões Zamboni
- Department of Cell Biology, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Fernando Q. Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
| | | | - Nicolas Riteau
- Immune Health Laboratory, Regulation of host responses and immune health, IRL2029, French National Centre for Scientific Research (CNRS) and Ribeirão Preto Medical School (FMRP) of the Sao Paulo University (USP), Sao Paulo, Brazil
- INEM, CNRS, UMR7355 and University, Orleans, France
| | - José C. Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, and
- Center for Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, Sao Paulo, Brazil
- Immune Health Laboratory, Regulation of host responses and immune health, IRL2029, French National Centre for Scientific Research (CNRS) and Ribeirão Preto Medical School (FMRP) of the Sao Paulo University (USP), Sao Paulo, Brazil
| |
Collapse
|
5
|
Masuda-Kuroki K, Alimohammadi S, Lowry S, Di Nardo A. Sphingosine 1-phosphate receptor 2 in keratinocytes plays a key role in reducing inflammation in psoriasis. Front Immunol 2024; 15:1469829. [PMID: 39391307 PMCID: PMC11464331 DOI: 10.3389/fimmu.2024.1469829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024] Open
Abstract
Background Psoriasis is an inflammatory skin condition where immune cells play a significant role. The importance of the cross-talk between keratinocytes and immune cells in the pathogenesis of psoriasis has recently been reaffirmed. Recent studies have found that several S1PR functional antagonists, other than S1PR2, are effective in improving psoriasis. This study aims to investigate the role of S1PR2 in psoriasis, that has not been investigated before. Methods Spatial transcriptomics, RT-qPCR, and flow cytometry were used to map the immune cell landscape and its association with metabolic pathways in an imiquimod (IMQ)-induced psoriasis-like inflammation in S1pr2fl/fl K14-Cre mice that could not sense sphingosine-1-phosphate (S1P) in the epidermis through the S1PR2 receptor. Results Our analysis suggests that S1PR2 in keratinocytes plays a major role in psoriasis-like inflammation compared to other S1PRs. It acts as a down-regulator, inhibiting the recruitment of Th17 cells into the skin. In IMQ-induced psoriasis skin, both S1pr2-/- and S1pr2fl/fl K14-Cre mice showed higher expressions of proinflammatory cytokines such as TNF-α, IL-17A, and IL-1β together with higher expressions of MyD88/NF-κB pathway compared to the wild-type mice. Remarkably, in IMQ-treated mice, the deletion of S1pr2 in keratinocytes only resulted in a larger population of Th17 cells in skin-draining lymph nodes. Other S1PR modulators did not improve the worsening of psoriasis-like inflammation caused by S1PR2 deficiency in keratinocytes. Conclusion This study reaches two main conclusions: signals from keratinocytes play a central role in creating an immune environment that promotes the development of psoriasis, and stimulating S1PR2, instead of suppressing it, represents a potential therapeutic approach for psoriasis.
Collapse
Affiliation(s)
| | | | | | - Anna Di Nardo
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
6
|
Gujarathi PP, Korat RH, Gujarathi PS. Preclinical techniques for drug discovery in psoriasis. Int Immunopharmacol 2024; 137:112378. [PMID: 38852518 DOI: 10.1016/j.intimp.2024.112378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
Psoriasis is a chronic, inflammatory, papulosquamous, noncontagious disease characterized by scaly, demarcated erythematous plaque, affecting skin, nails, and scalp. The IL-23/Th17 axis is the main operator in the development of psoriasis. Psoriasis is affecting worldwide, and new treatment options are urgently needed. Various local and systemic treatments are available for psoriasis but they only provide symptomatic relief because of numerous unknown mechanisms. Clinical trials demand overwhelming resources; therefore, drug development predominantly depends on the in-vivo, in-vitro, and ex-vivo techniques. Immediate attention is required to develop experimental techniques that completely imitate human psoriasis to assist drug development. This review portrays the various in-vivo, in-vitro, and ex-vivo techniques used in psoriasis research. It describes these techniques' characteristics, pathological presentations, and mechanisms. The experimental techniques of psoriasis provide significant information on disease progression mechanisms and possible therapeutic targets. However, until now, it has been challenging to invent a timely, affordable model that precisely imitates a human disease. Only the xenotransplantation model is reckoned as the closer, that mimics the complete genetic, and immunopathogenic event. Imiquimod-induced psoriasis and HaCat cell lines are popular among researchers because of their convenience, ease of use, and cost-effectiveness. There need to further improve the experimental techniques to best serve the disease imitation and meet the research goal.
Collapse
Affiliation(s)
- Pranjal P Gujarathi
- Department of Pharmacology, Vidhyadeep Institute of Pharmacy, Vidhyadeep University, Anita, Surat, Gujarat, India; Bhagwan Mahavir Centre for Advance Research, Bhagwan Mahavir College of Pharmacy, Bhagwan Mahavir University, Vesu, Surat, Gujarat, India.
| | - Rashmi H Korat
- Department of Pharmacognosy, Bhagwan Mahavir College of Pharmacy, Bhagwan Mahavir Univeristy, Vesu, Surat, Gujarat, India
| | - Piyush S Gujarathi
- Department of Community Medicine, Vidhyadeep Homeopathic Medical College and Research Centre, Vidhyadeep University, Anita, Surat, Gujarat, India
| |
Collapse
|
7
|
Zheng T, Deng J, Wen J, Xiao S, Huang H, Shang J, Zhang L, Chen H, Li J, Wang Y, Ouyang S, Yang M, Otsu K, Liu X, Huang G. p38α deficiency ameliorates psoriasis development by downregulating STAT3-mediated keratinocyte proliferation and cytokine production. Commun Biol 2024; 7:999. [PMID: 39147860 PMCID: PMC11327308 DOI: 10.1038/s42003-024-06700-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
Psoriasis is characterized by keratinocyte (KC) hyperproliferation and inflammatory cell infiltration, but the mechanisms remain unclear. In an imiquimod-induced mouse psoriasiform model, p38 activity is significantly elevated in KCs and p38α specific deletion in KCs ameliorates skin inflammation. p38α signaling promotes KC proliferation and psoriasis-related proinflammatory gene expression during psoriasis development. Mechanistically, p38α enhances KC proliferation and production of inflammatory cytokines and chemokines by activating STAT3. While p38α signaling in KCs does not affect the expression of IL-23 and IL-17, it substantially amplifies the IL-23/IL-17 pathogenic axis in psoriasis. The therapeutic effect of IL-17 neutralization is associated with decreased p38 and STAT3 activities in KCs and targeting the p38α-STAT3 axis in KCs ameliorates the severity of psoriasis. As IL-17 also highly activates p38 and STAT3 in KCs, our findings reveal a sustained signaling circuit important for psoriasis development, highlighting p38α-STAT3 axis as an important target for psoriasis treatment.
Collapse
Affiliation(s)
- Tingting Zheng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
| | - Jiaqi Deng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Jiahong Wen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Shuxiu Xiao
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyong Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Jiawen Shang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Luwen Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Huan Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Jingyu Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yanyan Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Suidong Ouyang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Meng Yang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Kinya Otsu
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
- Cardiovascular Division, King's College London, London, UK
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, Institute of Biochemistry & Molecular Biology, Guangdong Medical University, Dongguan, China
| | - Gonghua Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
Xiao X, Qiu T, Cheng Q, Wang W, Fan C, Zuo F. Uridine phosphorylase-1 promotes cell viability and cell-cycle progression in human epidermal keratinocytes via the glycolytic pathway. Clin Exp Pharmacol Physiol 2024; 51:e13874. [PMID: 38797519 DOI: 10.1111/1440-1681.13874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024]
Abstract
Glycolysis is vital for the excessive proliferation of keratinocytes in psoriasis, and uridine phosphorylase-1 (UPP1) functions as an enhancer of cancer cell proliferation. However, little is known about whether UPP1 promotes keratinocyte proliferation and accelerates psoriasis development. This study revealed that UPP1 facilitates cell viability and cell-cycle progression in human epidermal keratinocytes (HEKs) by modulating the glycolytic pathway. Bioinformatics analysis of UPP1 gene expression and its correlation with the Reactome revealed that UPP1 mRNA expression, cell-cycle progression, the interleukin-6 (IL-6)/Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathway and glycolysis were positively associated with psoriasis. Cell proliferation, the cell cycle and glycolysis were evaluated after UPP1 was silenced or overexpressed. The results showed that UPP1 overexpression increased cell proliferation, cell-cycle progression and glycolysis, which was contrary to the effects of UPP1 silencing. However, the STAT3 inhibitor diminished UPP1 expression because STAT3 can bind to the UPP1 promoter. In conclusion, UPP1 was significantly activated by the IL-6/STAT3 pathway and could modulate glycolysis to regulate cell proliferation and cell-cycle progression in keratinocytes during the development of psoriasis.
Collapse
Affiliation(s)
- Xiaoqing Xiao
- Department of Dermatology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tianwen Qiu
- Department of Dermatology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qiong Cheng
- Department of Dermatology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenyu Wang
- Department of Dermatology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunyan Fan
- Department of Dermatology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fuguo Zuo
- Department of Dermatology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
9
|
Wei J, Zhang J, Hu F, Zhang W, Wu Y, Liu B, Lu Y, Li L, Han L, Lu C. Anti-psoriasis effect of 18β-glycyrrhetinic acid by breaking CCL20/CCR6 axis through its vital active group targeting GUSB/ATF2 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155524. [PMID: 38552435 DOI: 10.1016/j.phymed.2024.155524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 02/02/2024] [Accepted: 03/07/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Psoriasis is an immune-mediated chronic inflammatory skin disease. Current research suggests that the long-term persistence and recurrence of psoriasis are closely related to the feedback loop formed between keratinocytes and immune cells, especially in Th 17 or DC cells expressing CCR6. CCL20 is the ligand of CCR6. Therefore, drugs that block the expression of CCL20 or CCR6 may have a certain therapeutic effect on psoriasis. Glycyrrhetinic acid (GA) is the main active ingredient of the plant drug licorice and is often used to treat autoimmune diseases, including psoriasis. However, its mechanism of action is still unclear. METHODS Psoriasis like skin lesion model was established by continuously applying imiquimod on the back skin of normal mice and CCR6-/- mice for 7 days. The therapeutic and preventive effects of glycyrrhetinic acid (GA) on the model were observed and compared. The severity of skin injury is estimated through clinical PASI scores and histopathological examination. qRT-PCR and multiple cytoline assay were explored to detect the expression levels of cytokines in animal dorsal skin lesions and keratinocyte line HaCaT cells, respectively. The dermis and epidermis of the mouse back were separated for the detection of CCL20 expression. Transcription factor assay was applied to screen, and luciferase activity assay to validate transcription factors regulated by GA. Technology of surface plasmon laser resonance with LC-MS (SPR-MS), molecular docking, and enzyme activity assay were used to identified the target proteins for GA. Finally, we synthesized different derivatives of 18beta-GA and compared their effects, as well as glycyrrhetinic acid (GL), on the skin lesion of imiquimod-induced mice to evaluate the active groups of 18beta-GA. RESULTS 18β-glycyrrhetinic acid (GA) improved IMQ-induced psoriatic lesions, and could specifically reduce the chemokine CCL20 level of the epidermis in lesion area, especially in therapeutic administration manner. The process was mainly regulated by transcription factor ATF2 in the keratinocytes. In addition, GUSB was identified as the primary target of 18βGA. Our findings indicated that the subject on molecular target research of glycyrrhizin should be glycyrrhetinic acid (GA) instead of glycyrrhizic acid (GL), because GL showed little activity in vitro or in vivo. Apart from that, α, β, -unsaturated carbonyl in C11/12 positions was crucial or unchangeable to its activity of 18βGA, while proper modification of C3 or C30 position of 18βGA may vastly increase its activity. CONCLUSION Our research indicates that 18βGA exerted its anti-psoriasis effect mainly by suppressing ATF2 and downstream molecule CCL20 predominately through α, β, -unsaturated carbonyl at C11/12 position binding to GUSB in the keratinocytes, and then broke the feedback loop between keratinocytes and CCR6-expressing immune cells. GA has more advantages than GL in the external treatment of psoriasis. A highlight of this study is to investigate the influence of special active groups on the pharmacological action of a natural product, inspired by the molecular docking result.
Collapse
Affiliation(s)
- Jianan Wei
- Research Team of Molecular and Systems Biology of Chinese medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Junhong Zhang
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Fengju Hu
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Wenjuan Zhang
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Yunshan Wu
- Laboratory of Chinese Medicine Quality Standard, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Bo Liu
- Laboratory of Chinese Medicine Quality Standard, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Yue Lu
- Research Team of Molecular and Systems Biology of Chinese medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Li Li
- Research Team of Molecular and Systems Biology of Chinese medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Ling Han
- Research Team of Molecular and Systems Biology of Chinese medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China; State Key laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou 510120, China.
| | - Chuanjian Lu
- State Key laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou 510120, China.
| |
Collapse
|
10
|
Leong HF, Wang WH, Peng F. Biologics as a novel treatment option for palmoplantar pustulosis: a comprehensive review. Postepy Dermatol Alergol 2024; 41:262-269. [PMID: 39027700 PMCID: PMC11253319 DOI: 10.5114/ada.2024.141128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 05/13/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Palmoplantar pustulosis (PPP) is a complex inflammatory skin disease. Currently, no standardized treatments exist, and traditional systemic therapies often display limited effectiveness and substantial adverse effects. Biologics, however, have shown potential for enhanced clinical outcomes in psoriasis patients, thereby prompting this investigation into their applicability in PPP treatment. Aim This study constitutes the first comprehensive review to assess the effectiveness and underlying mechanisms of biologics for PPP. Material and methods We conducted a PubMed search to identify studies on biologics for PPP from 1992 onward. The review focused on assessing the efficacy of biologics targeting cytokines like IL-1, IL-8, IL-17, IL-12/23, IL-36, and TNF-α. Results Biologics for PPP are generally less effective than for psoriasis. Secukinumab and guselkumab, IL-17 and IL-23 inhibitors respectively, have shown better results compared to other biologics in trials. However, the effectiveness of other biologics remains uncertain due to limited data. Conclusions More research is needed to find effective treatments for PPP, and selecting the right biologic for each patient is challenging.
Collapse
Affiliation(s)
- Hio Fong Leong
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Wen-Hui Wang
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Fen Peng
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
11
|
Hansen RL, Jørgensen TS, Egeberg A, Rosenø NAL, Skougaard M, Stisen ZR, Dreyer L, Kristensen LE. Adherence to therapy of ixekizumab and secukinumab in psoriatic arthritis patients using first- or second-line IL-17A inhibitor treatment: a Danish population-based cohort study. Rheumatology (Oxford) 2024; 63:1593-1598. [PMID: 37647648 DOI: 10.1093/rheumatology/kead434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/20/2023] [Accepted: 08/02/2023] [Indexed: 09/01/2023] Open
Abstract
OBJECTIVES To assess the effectiveness and tolerability of first- and second-line interleukin (IL)-17A inhibitor treatment in PsA patients from 2014 to 2021 using data from the Danish Rheumatology Registry (DANBIO) by investigating adherence to therapy. METHOD PsA patients recorded in the DANBIO who received a first- or second-line IL-17A inhibitor treatment were included in this study. All patients included had previously received one or more TNF inhibitor treatment. Baseline characteristics were analysed in subgroups as first-line IL-17A inhibitor treatment and second-line IL-17A inhibitor treatment. Adherence to therapy of first- or second-line IL-17A inhibitor treatments was reported as Kaplan-Meier plots. RESULTS A total of 534 patients were included in the study, with 534 first-line switchers (secukinumab: 510, ixekizumab: 24) and 102 second-line switchers (secukinumab: 35, ixekizumab: 67). Baseline characteristics showed a similar HAQ and visual analogue scale (VAS) for pain. VAS global, 28-joint DAS with CRP and the previous number of biologic DMARD treatments were similar, with a greater value for second-line switchers. First-line ixekizumab-treated patients present a younger age, greater percentage of females, a lower disease duration and a lower CRP value. Concomitant MTX use was greater for the first-line secukinumab-treated patients. First- and second-line switchers had a similar adherence to therapy. Second-line secukinumab and second-line ixekizumab switchers showed a similar adherence to treatment. CONCLUSION PsA patients receiving first- or second-line IL-17A inhibitors showed homogeneous baseline characteristics and similar adherence to therapy. Treatment failure of the first IL-17A inhibitor treatment should not preclude a second-line IL-17A inhibitor treatment.
Collapse
Affiliation(s)
- Rebekka L Hansen
- Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Frederiksberg, Denmark
| | - Tanja S Jørgensen
- Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Frederiksberg, Denmark
| | - Alexander Egeberg
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Nana A L Rosenø
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Marie Skougaard
- Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Frederiksberg, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Center of Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Rheumatology, Center of Rheumatic Research Aalborg, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
| | - Zara R Stisen
- Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Frederiksberg, Denmark
| | - Lene Dreyer
- Center of Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Rheumatology, Center of Rheumatic Research Aalborg, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
- Danish Rheumatologic Database, Copenhagen, Denmark
| | - Lars Erik Kristensen
- Parker Institute, Copenhagen University Hospital, Bispebjerg and Frederiksberg, Frederiksberg, Denmark
| |
Collapse
|
12
|
Swaroop AK, Negi P, Kar A, Mariappan E, Natarajan J, Namboori P K K, Selvaraj J. Navigating IL-6: From molecular mechanisms to therapeutic breakthroughs. Cytokine Growth Factor Rev 2024; 76:48-76. [PMID: 38220583 DOI: 10.1016/j.cytogfr.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/28/2023] [Indexed: 01/16/2024]
Abstract
This concise review navigates the intricate realm of Interleukin-6 (IL-6), an important member of the cytokine family. Beginning with an introduction to cytokines, this narrative review unfolds with the historical journey of IL-6, illuminating its evolving significance. A crucial section unravels the three distinct signaling modes employed by IL-6, providing a foundational understanding of its versatile interactions within cellular landscapes. Moving deeper, the review meticulously dissects IL-6's signaling mechanisms, unraveling the complexities of its pleiotropic effects in both physiological responses and pathological conditions. A significant focus is dedicated to the essential role IL-6 plays in inflammatory diseases, offering insights into its associations and implications for various health conditions. The review also takes a therapeutic turn by exploring the emergence of anti-IL-6 monoclonal inhibitors, marking a profound stride in treatment modalities. Diving into the molecular realm, the review explores small molecules as agents for IL-6 inhibition, providing a nuanced perspective on diverse intervention strategies. As the review embarks on the final chapters, it contemplates future aspects, offering glimpses into potential research trajectories and the evolving landscape of IL-6-related studies.
Collapse
Affiliation(s)
- Akey Krishna Swaroop
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Preeya Negi
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Ayushi Kar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Esakkimuthukumar Mariappan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Jawahar Natarajan
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India
| | - Krishnan Namboori P K
- Amrita Molecular Modeling and Synthesis (AMMAS) Research lab, Amrita Vishwavidyapeetham, Amrita Nagar, Ettimadai, Coimbatore, Tamil Nadu, India
| | - Jubie Selvaraj
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Tamil Nadu, India.
| |
Collapse
|
13
|
Toplu SA, Altunisik N. Comment on 'Immune response to COVID-19 mRNA vaccination in patients with psoriasis undergoing treatment with biologics'. Clin Exp Dermatol 2024; 49:394. [PMID: 37966357 DOI: 10.1093/ced/llad392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 11/21/2023] [Indexed: 11/16/2023]
Affiliation(s)
| | - Nihal Altunisik
- Department of Dermatology, Inonu University, Malatya, Turkey
| |
Collapse
|
14
|
Greenzaid J, Feldman S. Clinical Pharmacokinetic and Pharmacodynamic Considerations in the Treatment of Moderate-to-Severe Psoriasis. Clin Pharmacokinet 2024; 63:137-153. [PMID: 38280146 DOI: 10.1007/s40262-023-01341-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/29/2024]
Abstract
Psoriasis is a common inflammatory immune disorder due to chronic activation of the adaptive and innate immune responses. Therapies for psoriasis target reducing inflammatory cytokines such as tumor necrosis factor-alpha, interleukin-17, and interleukin-22. Patients with inflammatory disorders have reduced metabolism by cytochrome P450 enzymes in the liver. The pharmacokinetic and pharmacodynamic changes due to psoriasis also have an impact on reaching therapeutic concentrations of the drug. Pharmacokinetic and pharmacodynamic data help determine the safety and clinical considerations necessary when utilizing drugs for plaque psoriasis. A literature search was performed on PubMed and Ovid MEDLINE for the pharmacokinetic and pharmacodynamic data of oral therapies and biologics utilized for moderate-to-severe plaque psoriasis. The findings from the literature search were organized into two sections: oral therapies and biologics. The pharmacokinetic and pharmacodynamic parameters in healthy patients, patients with psoriasis, and special populations are discussed in each section. The oral therapies described in this review include methotrexate, cyclosporine, apremilast, tofacitinib, and deucravacitinib. Biologics include tumor necrosis factor-alpha inhibitors, interleukin-17 inhibitors, ustekinumab, and interleukin-23 inhibitors. Clinical considerations for these therapies include drug toxicities, dosing frequency, and anti-drug antibodies. Methotrexate and cyclosporine have a risk for hepatoxicity and renal impairment, respectively. Moreover, drugs metabolized via cytochrome P450, including tofacitinib and apremilast have decreased clearance in patients with psoriasis, requiring dose adjustments. Patients treated with therapies such as adalimumab can develop anti-drug antibodies that reduce the long-term efficacy of the drug. Additionally, overweight patients benefit from more frequent dosing to achieve better psoriasis clearance.
Collapse
Affiliation(s)
- Jonathan Greenzaid
- Department of Dermatology, Center for Dermatology Research, Wake Forest University School of Medicine, 475 Vine St, Winston-Salem, NC, 27101, USA.
| | - Steven Feldman
- Department of Dermatology, Center for Dermatology Research, Wake Forest University School of Medicine, 475 Vine St, Winston-Salem, NC, 27101, USA
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Social Sciences & Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
15
|
Aixue W, Feng W, Huanhuan Z, Xixing M, Yanling L. Cosentyx alleviates psoriasis-induced podocyte injury by inhibiting the tlr/nf-κb signaling pathway. Skin Res Technol 2024; 30:e13562. [PMID: 38279604 PMCID: PMC10818124 DOI: 10.1111/srt.13562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/19/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Pathological studies have shown an association between psoriasis and renal podocyte injury, and the specific mechanism of podocyte injury in psoriasis remains unclear, with no effective treatments currently available. This study aimed to investigate the underlying mechanisms of podocyte and epidermal cell injury in psoriasis and evaluate the therapeutic effect of Cosentyx. MATERIALS AND METHODS A psoriasis-like mouse model was established using BALB/C mice, and Cosentyx treatment was administered via intraperitoneal injection. Various parameters, including skin lesions, urinary protein, kidney/serum inflammatory cytokines, kidney function, podocyte membrane proteins, and Toll-like receptors/nuclear factor kappa-b (TLR/NF-κB) pathway-associated proteins, were analyzed to explore the mechanisms of podocyte and epidermal cell injury in psoriasis and the potential ameliorative effects of Cosentyx. RESULT Treatment with Cosentyx significantly reduced the increased levels of urinary protein, creatinine, and blood urea nitrogen caused by psoriasis. Cosentyx inhibited the upregulation of kidney/serum inflammatory factors (IL-17, IL-1β, IL-6, TNF-α, and IL-22) and TLR/NF-κB-related proteins (TLR2, TLR4, MyD88, and NF-κBp65) in both psoriatic skin and kidney tissues, while also reducing the accumulation of oxidative products. Moreover, Cosentyx treatment suppressed podocyte apoptosis and promoted epidermal cell apoptosis. The experimental data demonstrated that psoriasis-like inflammation impaired renal podocytes through the TLR/NF-κB signaling pathway. CONCLUSION Cosentyx treatment effectively inhibited the expression of TLR/NF-κB-related proteins, providing a therapeutic effect for psoriasis-induced kidney and skin injuries.
Collapse
Affiliation(s)
- Wang Aixue
- DermatologyThe Second Hospital of Hebei Medical UniversityShijiazhuang HebeiChina
| | - Wei Feng
- DermatologyThe Second Hospital of Hebei Medical UniversityShijiazhuang HebeiChina
| | - Zhang Huanhuan
- DermatologyThe Second Hospital of Hebei Medical UniversityShijiazhuang HebeiChina
| | - M Xixing
- DermatologyThe Second Hospital of Hebei Medical UniversityShijiazhuang HebeiChina
| | - L Yanling
- DermatologyThe Second Hospital of Hebei Medical UniversityShijiazhuang HebeiChina
| |
Collapse
|
16
|
Wang H, Pan F, Liu J, Zhang J, Fuli Zhang, Wang Y. Huayuwendan decoction ameliorates inflammation via IL-17/NF-κB signaling pathway in diabetic rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117328. [PMID: 37865275 DOI: 10.1016/j.jep.2023.117328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/30/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huayuwendan decoction (HYWD) is a broad used traditional Chinese medicine and therapeutic effects against type 2 diabetes mellitus (T2DM). The mechanism of HYWD on the treatment of T2DM is still unclear. AIM OF THE STUDY For this reason, this study was performed to uncover the effects and mechanism of action of HYWD on T2DM. MATERIALS AND METHODS Male Wistar rats were chosen to set up the T2DM model. This study was randomly divided into six groups: CON (control), MOD (model), HYWDL (Huayuwendan decoction Low Dose), HYWDM (Huayuwendan decoction Middle Dose), HYWDH (Huayuwendan decoction High Dose), and MET (Metformin). Body weight gains were estimated. Using H&E staining, pathological alterations was explored. The serums of fasting plasma glucose (FPG), 2-h postprandial plasma glucose (2 h PG) were detected by Roche blood glucose meter. LDL-C and HDL-C were analyzed by automatic biochemical analyzer. Network pharmacology analyzed the active ingredients, drug targets, and key pathways of HYWD in T2DM treatment. The islet function and inflammation related factors were determined by ELISA. NF-κB signaling pathway or IL-17 signaling pathway related proteins were analyzed by Western blotting. IL-17RA were determined by immunohistochemistry analyze. RESULTS HYWD inhibited weight gain in T2DM rats. Histopathological results showed that HYWD inhibits liver injury. HYWD suppressed LDL-C and enhanced HDL-C in serum of T2DM rats. HYWD reduce FPG and 2 h PG, inhibit Fins, GSP and IRI, but enhance IAI in serum of T2DM rats. In addition, the network pharmacology results identified 292 chemical compounds in HYWD. 279 candidate targets were recognized, including IL-17A, IL-1β, NFкB, stats, mmp3, and cxcl2. The pathways revealed that the possible target of HYWD related with the regulation of IL-17 signaling pathway and NF-κB pathway. Then in vivo study, HYWD reduced the levels of IL-6, TNF-α, IL-17 and IL-1β in serum and inhibit the protein expression involved in IL-17/NF-κB signaling pathway. CONCLUSIONS The study demonstrates that HYWD may improve T2DM by repressing with the IL-17/NF-κB signaling pathway, which offer encouraging support for using alternative medicine of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Hongyang Wang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China; Heilongjiang Univ Tradit Chinese Med, Harbin, Heilongjiang, 150000, China
| | - Fuzhen Pan
- Heilongjiang Univ Tradit Chinese Med, Harbin, Heilongjiang, 150000, China
| | - Jie Liu
- Heilongjiang Univ Tradit Chinese Med, Harbin, Heilongjiang, 150000, China
| | - Juncheng Zhang
- Heilongjiang Univ Tradit Chinese Med, Harbin, Heilongjiang, 150000, China
| | - Fuli Zhang
- Heilongjiang Univ Tradit Chinese Med, Harbin, Heilongjiang, 150000, China
| | - Yu Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, 563006, China; Inner Mongolia Minzu University, Tongliao, Inner Mongolia, 028000, China.
| |
Collapse
|
17
|
Li L, Lu J, Liu J, Wu J, Zhang X, Meng Y, Wu X, Tai Z, Zhu Q, Chen Z. Immune cells in the epithelial immune microenvironment of psoriasis: emerging therapeutic targets. Front Immunol 2024; 14:1340677. [PMID: 38239345 PMCID: PMC10794746 DOI: 10.3389/fimmu.2023.1340677] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/15/2023] [Indexed: 01/22/2024] Open
Abstract
Psoriasis is a chronic autoimmune inflammatory disease characterized by erroneous metabolism of keratinocytes. The development of psoriasis is closely related to abnormal activation and disorders of the immune system. Dysregulated skin protective mechanisms can activate inflammatory pathways within the epithelial immune microenvironment (EIME), leading to the development of autoimmune-related and inflammatory skin diseases. In this review, we initially emphasized the pathogenesis of psoriasis, paying particular attention to the interactions between the abnormal activation of immune cells and the production of cytokines in psoriasis. Subsequently, we delved into the significance of the interactions between EIME and immune cells in the emergence of psoriasis. A thorough understanding of these immune processes is crucial to the development of targeted therapies for psoriasis. Finally, we discussed the potential novel targeted therapies aimed at modulating the EIME in psoriasis. This comprehensive examination sheds light on the intricate underlying immune mechanisms and provides insights into potential therapeutic avenues of immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Lisha Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Jiaye Lu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Jun Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Junchao Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Yu Meng
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Xiying Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| |
Collapse
|
18
|
Yang Y, Zheng X, Lv H, Tang B, Bi Y, Luo Q, Yao D, Chen H, Lu C. A bibliometrics study on the status quo and hot topics of pathogenesis of psoriasis based on Web of Science. Skin Res Technol 2024; 30:e13538. [PMID: 38174774 PMCID: PMC10765367 DOI: 10.1111/srt.13538] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/06/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Psoriasis is an immune-mediated chronic inflammatory skin disease. Great progress has been made in the pathogenesis of psoriasis in recent years, but there is no bibliometric study on the pathogenesis of psoriasis. The purpose of this study was to use bibliometrics method to analyze the research overview and hot spots of pathogenesis of psoriasis in recent 10 years, so as to further understand the development trend and frontier of this field. METHODS The core literatures on the pathogenesis of psoriasis were searched in the Web of Science database, and analyzed by VOSviewer, CiteSpace, and Bibliometrix in terms of the annual publication volume, country, institution, author, journal, keywords, and so on. RESULTS A total of 3570 literatures were included. China and the United States were the main research countries in this field, and Rockefeller University was the main research institution. Krueger JG, the author, had the highest number of publications and the greatest influence, and Boehncke (2015) was the most cited local literature. J INVEST DERMATOL takes the top spot in terms of the number of Dermatol articles and citation frequency. The main research hotspots in the pathogenesis of psoriasis are as follows: (1) The interaction between innate and adaptive immunity and the related inflammatory loop dominated by Th17 cells and IL-23/IL-17 axis are still the key mechanisms of psoriasis; (2) molecular genetic studies represented by Long Non-Coding RNA (LncRNA); (3) integrated research of multi-omics techniques represented by gut microbiota; and (4) Exploring the comorbidity mechanism of psoriasis represented by Metabolic Syndrome (MetS). CONCLUSION This study is a summary of the current research status and hot trend of the pathogenesis of psoriasis, which will provide some reference for the scholars studying the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Yujie Yang
- The Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Xuwei Zheng
- The Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Haiying Lv
- The Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Bin Tang
- The Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouChina
- State Key Laboratory of Dampness Syndrome of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine)GuangzhouChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangzhouChina
- Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine DermatologyGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yang Bi
- The Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Qianqian Luo
- The Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Danni Yao
- The Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouChina
- State Key Laboratory of Dampness Syndrome of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine)GuangzhouChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangzhouChina
- Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine DermatologyGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouChina
| | - Haiming Chen
- The Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouChina
- State Key Laboratory of Dampness Syndrome of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine)GuangzhouChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangzhouChina
- Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine DermatologyGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouChina
| | - Chuanjian Lu
- The Second Clinical College of Guangzhou University of Chinese MedicineGuangzhouChina
- State Key Laboratory of Dampness Syndrome of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine)GuangzhouChina
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine SyndromeGuangzhouChina
- Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine DermatologyGuangzhouChina
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchGuangzhou University of Chinese MedicineGuangzhouChina
| |
Collapse
|
19
|
Russo F, Galluzzo M, Stingeni L, Persechino S, Zichichi L, Conti A, Giofrè C, Dini V, Vispi M, Atzori L, Cattaneo A, Parodi A, Bardazzi F, Stinco G, Dapavo P, Girolomoni G, Musumeci ML, Papini M, Venturini M, Dastoli S, Di Nuzzo S, Fargnoli MC, Pagnanelli G, Bernardini N, Gambini DM, Malagoli P, Mazzatenta C, Peris K, Zalaudek I, Fabbrocini G, Loconsole F, Vassallo C, Pietroleonardo L, Prignano F, Franchi C, Offidani AM, Bonifati C, Di Lernia V, Gigante G, Bartezaghi MS, Franchi M, Ursoleo P, Aloisi E. Long-Term Drug Survival and Effectiveness of Secukinumab in Patients with Moderate to Severe Chronic Plaque Psoriasis: 42-Month Results from the SUPREME 2.0 Study. Clin Cosmet Investig Dermatol 2023; 16:3561-3574. [PMID: 38107670 PMCID: PMC10725693 DOI: 10.2147/ccid.s416149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 11/01/2023] [Indexed: 12/19/2023]
Abstract
Purpose SUPREME, a phase IIIb study conducted in Italy, demonstrated safety and high efficacy of secukinumab for up to 72 weeks in patients with moderate-to-severe plaque-type psoriasis. SUPREME 2.0 study aimed to provide real-world data on the long-term drug survival and effectiveness of secukinumab beyond 72 weeks. Patients and Methods SUPREME 2.0 is a retrospective observational chart review study conducted in patients previously enrolled in SUPREME study. After the end of the SUPREME study, eligible patients continued treatment as per clinical practice, and their effectiveness and drug survival data were retrieved from medical charts. Results Of the 415 patients enrolled in the SUPREME study, 297 were included in SUPREME 2.0; of which, 210 (70.7%) continued secukinumab treatment throughout the 42-month observation period. Patients in the biologic-naïve cohort had higher drug survival than those in the biologic-experienced cohort (74.9% vs 61.7%), while HLA-Cw6-positive and HLA-Cw6-negative patients showed similar drug survival (69.3% and 71.9%). After 42 months, Psoriasis Area and Severity Index (PASI) 90 was achieved by 79.6% of patients overall; with a similar proportion of biologic-naïve and biologic-experienced patients achieving PASI90 (79.8% and 79.1%). The mean absolute PASI score reduced from 21.94 to 1.38 in the overall population, 21.90 to 1.24 in biologic-naïve and 22.03 to 1.77 in biologic-experienced patients after 42 months. The decrease in the absolute PASI score was comparable between HLA-Cw6-positive and HLA-Cw6-negative patients. The baseline Dermatology Life Quality Index scores also decreased in the overall patients (10.5 to 2.32) and across all study sub-groups after 42 months. Safety was consistent with the known profile of secukinumab, with no new findings. Conclusion In this real-world cohort study, secukinumab showed consistently high long-term drug survival and effectiveness with a favourable safety profile.
Collapse
Affiliation(s)
- Filomena Russo
- Dermatology Section, Department of Medical, Surgical and Neurological Science, University of Siena, S. Maria Alle Scotte Hospital, Siena, Italy
| | - Marco Galluzzo
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
- Dermatology Unit, Fondazione Policlinico “Tor Vergata”, Rome, Italy
| | - Luca Stingeni
- Section of Dermatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Severino Persechino
- Dermatology Unit, NESMOS Department, Faculty of Medicine & Psychology, Sapienza University of Rome, Sant’Andrea University Hospital, Rome, Italy
| | | | - Andrea Conti
- Section of Dermatology, Department of Specialized Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Valentina Dini
- Section of Dermatology, Department of Medicine and Oncology, University of Pisa, Pisa, Italy
| | - Martina Vispi
- Dermatology Unit, Misericordia Hospital, Grosseto, Italy
| | - Laura Atzori
- Dermatology Unit, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Angelo Cattaneo
- Dermatology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Aurora Parodi
- Section of Dermatology, DiSSal University of Genoa, Ospedale-Policlinico San Martino IRCCS, Genova, Italy
| | - Federico Bardazzi
- Dermatology Division, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Giuseppe Stinco
- Section of Dermatology, Department of Medicine, University of Udine, Udine, Italy
| | - Paolo Dapavo
- Section of Dermatology, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giampiero Girolomoni
- Section of Dermatology, Department of Medicine, University of Verona, Verona, Italy
| | - Maria Letizia Musumeci
- Section of Dermatology, Department of Medical and Surgical Specialties, University of Catania, Catania, Italy
| | - Manuela Papini
- Dermatology Clinic of Terni, University of Perugia, Perugia, Italy
| | - Marina Venturini
- Section of Dermatology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Dastoli
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Sergio Di Nuzzo
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maria Concetta Fargnoli
- Section of Dermatology, Department of Biotechnological and Applied Clinical Science, University of L’Aquila, L’Aquila, Italy
| | - Gianluca Pagnanelli
- Department of Dermatology, Istituto Dermopatico dell’Immacolata - IRCCS, Roma, Italy
| | - Nicoletta Bernardini
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Dermatology Unit, “Daniele Innocenzi”, Asl Latina, Italy
| | | | | | - Carlo Mazzatenta
- Dermatology Unit, Lucca Azienda USL Toscana Nord Ovest, Pisa, Italy
| | - Ketty Peris
- Department of Translational Medicine and Surgery, IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Iris Zalaudek
- Department of Dermatology, University of Trieste, Trieste, Italy
| | - Gabriella Fabbrocini
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Camilla Vassallo
- Institute of Dermatology, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy
| | | | - Francesca Prignano
- Dermatology Clinic, Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Anna Maria Offidani
- Dermatological Clinic, Polytechnic University of the Marche Region, Ancona, Italy
| | - Claudio Bonifati
- Department of Dermatology, Istituto Dermatologico San Gallicano - IRCCS, Roma, Italy
| | - Vito Di Lernia
- Dermatology Unit, Arcispedale S. Maria Nuova IRCCS, Reggio Emilia, Italy
| | | | | | - Matteo Franchi
- National Centre for Healthcare Research and Pharmacoepidemiology, Milan, Italy
- Laboratory of Healthcare Research and Pharmacoepidemiology, Department of Statistics and Quantitative Methods, University of Milano Bicocca, Milan, Italy
| | | | | |
Collapse
|
20
|
Kanekura T. CD147/Basigin Is Involved in the Development of Malignant Tumors and T-Cell-Mediated Immunological Disorders via Regulation of Glycolysis. Int J Mol Sci 2023; 24:17344. [PMID: 38139173 PMCID: PMC10743398 DOI: 10.3390/ijms242417344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
CD147/Basigin, a transmembrane glycoprotein belonging to the immunoglobulin superfamily, is a multifunctional molecule with various binding partners. CD147 binds to monocarboxylate transporters (MCTs) and supports their expression on plasma membranes. MTC-1 and MCT-4 export the lactic acid that is converted from pyruvate in glycolysis to maintain the intracellular pH level and a stable metabolic state. Under physiological conditions, cellular energy production is induced by mitochondrial oxidative phosphorylation. Glycolysis usually occurs under anaerobic conditions, whereas cancer cells depend on glycolysis under aerobic conditions. T cells also require glycolysis for differentiation, proliferation, and activation. Human malignant melanoma cells expressed higher levels of MCT-1 and MCT-4, co-localized with CD147 on the plasma membrane, and showed an increased glycolysis rate compared to normal human melanocytes. CD147 silencing by siRNA abrogated MCT-1 and MCT-4 membrane expression and disrupted glycolysis, inhibiting cancer cell activity. Furthermore, CD147 is involved in psoriasis. MCT-1 was absent on CD4+ T cells in CD147-deficient mice. The naïve CD4+ T cells from CD147-deficient mice exhibited a low capacity to differentiate into Th17 cells. Imiquimod-induced skin inflammation was significantly milder in the CD147-deficient mice than in the wild-type mice. Overall, CD147/Basigin is involved in the development of malignant tumors and T-cell-mediated immunological disorders via glycolysis regulation.
Collapse
Affiliation(s)
- Takuro Kanekura
- Department of Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| |
Collapse
|
21
|
Carlsson E, Cowell-McGlory T, Hedrich CM. cAMP responsive element modulator α promotes effector T cells in systemic autoimmune diseases. Immunology 2023; 170:470-482. [PMID: 37435993 DOI: 10.1111/imm.13680] [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: 04/15/2023] [Accepted: 07/01/2023] [Indexed: 07/13/2023] Open
Abstract
T lymphocytes play a crucial role in adaptive immunity. Dysregulation of T cell-derived inflammatory cytokine expression and loss of self-tolerance promote inflammation and tissue damage in several autoimmune/inflammatory diseases, including systemic lupus erythematosus (SLE) and psoriasis. The transcription factor cAMP responsive element modulator α (CREMα) plays a key role in the regulation of T cell homeostasis. Increased expression of CREMα is a hallmark of the T cell-mediated inflammatory diseases SLE and psoriasis. Notably, CREMα regulates the expression of effector molecules through trans-regulation and/or the co-recruitment of epigenetic modifiers, including DNA methyltransferases (DNMT3a), histone-methyltransferases (G9a) and histone acetyltransferases (p300). Thus, CREMα may be used as a biomarker for disease activity and/or target for future targeted therapeutic interventions.
Collapse
Affiliation(s)
- Emil Carlsson
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Taylor Cowell-McGlory
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Christian M Hedrich
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
- Paediatric Excellence Initiative, NIHR Great Ormond Street Biomedical Research Centre, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
| |
Collapse
|
22
|
Kerut CK, Wagner MJ, Daniel CP, Fisher C, Henderson EJ, Burroughs CR, Amarasinghe S, Willett O, Ahmadzadeh S, Varrassi G, Shekoohi S, Kaye AD. Guselkumab, a Novel Monoclonal Antibody Inhibitor of the p19 Subunit of IL-23, for Psoriatic Arthritis and Plaque Psoriasis: A Review of Its Mechanism, Use, and Clinical Effectiveness. Cureus 2023; 15:e51405. [PMID: 38292958 PMCID: PMC10825382 DOI: 10.7759/cureus.51405] [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: 12/08/2023] [Accepted: 12/30/2023] [Indexed: 02/01/2024] Open
Abstract
Psoriatic arthritis and plaque psoriasis are autoimmune conditions affecting multiple organs, including the skin. The pathophysiology and etiology of these conditions are not fully understood; however, numerous factors are believed to play a critical role, including genetics and environmental risk factors. Furthermore, research suggests the IL-23/IL-17 pathway partially mediates these diseases. Once the IL-23 receptor is bound and activated, two subunits, p19, and p40, act through different signaling pathways. Ultimately, inflammation is produced through the effector molecule, IL-17, other cytokines, and tumor necrosis factor (TNF). Traditionally, these chronic conditions have been treated with TNF-α inhibitors and methotrexate, a dihydrofolate reductase inhibitor. Although successful in inhibiting the immune system, these drugs can have many adverse effects due to their broad targets. In recent years, more targeted therapy has become popular. Guselkumab is a monoclonal antibody that inhibits the p19 subunit of IL-23. It has been FDA-approved to treat both plaque psoriasis and psoriatic arthritis. Clinical trials showing guselkumab's efficacy have been promising, even showing improvement in symptoms of plaque psoriasis patients resistant to adalimumab, a TNF-α inhibitor. Guselkumab has also been shown to be well tolerated with a similar safety profile as other biologics inhibiting the immune system. In addition to its efficacy in treating plaque psoriasis and psoriatic arthritis, the mechanism of action offers a targeted approach that may minimize the broad immunosuppressive effects often associated with traditional therapies, providing a potential advantage in the long-term management of these autoimmune conditions.
Collapse
Affiliation(s)
- Christian K Kerut
- School of Medicine, Louisiana State University Health Sciences Center at New Orleans, New Orleans, USA
| | - Maxwell J Wagner
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Charles P Daniel
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Claire Fisher
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Emmilee J Henderson
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Caroline R Burroughs
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Sam Amarasinghe
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Olga Willett
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | | | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| |
Collapse
|
23
|
Guo J, Zhang H, Lin W, Lu L, Su J, Chen X. Signaling pathways and targeted therapies for psoriasis. Signal Transduct Target Ther 2023; 8:437. [PMID: 38008779 PMCID: PMC10679229 DOI: 10.1038/s41392-023-01655-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 11/28/2023] Open
Abstract
Psoriasis is a common, chronic, and inflammatory skin disease with a high burden on individuals, health systems, and society worldwide. With the immunological pathologies and pathogenesis of psoriasis becoming gradually revealed, the therapeutic approaches for this disease have gained revolutionary progress. Nevertheless, the mechanisms of less common forms of psoriasis remain elusive. Furthermore, severe adverse effects and the recurrence of disease upon treatment cessation should be noted and addressed during the treatment, which, however, has been rarely explored with the integration of preliminary findings. Therefore, it is crucial to have a comprehensive understanding of the mechanisms behind psoriasis pathogenesis, which might offer new insights for research and lead to more substantive progress in therapeutic approaches and expand clinical options for psoriasis treatment. In this review, we looked to briefly introduce the epidemiology, clinical subtypes, pathophysiology, and comorbidities of psoriasis and systematically discuss the signaling pathways involving extracellular cytokines and intracellular transmission, as well as the cross-talk between them. In the discussion, we also paid more attention to the potential metabolic and epigenetic mechanisms of psoriasis and the molecular mechanistic cascades related to its comorbidities. This review also outlined current treatment for psoriasis, especially targeted therapies and novel therapeutic strategies, as well as the potential mechanism of disease recurrence.
Collapse
Affiliation(s)
- Jia Guo
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Hanyi Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Wenrui Lin
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Lixia Lu
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China
| | - Juan Su
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha, 410008, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, 410008, Hunan, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, 410008, Hunan, China.
| |
Collapse
|
24
|
Zhang P, Su Y, Li S, Chen H, Wu R, Wu H. The roles of T cells in psoriasis. Front Immunol 2023; 14:1081256. [PMID: 37942312 PMCID: PMC10628572 DOI: 10.3389/fimmu.2023.1081256] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 09/29/2023] [Indexed: 11/10/2023] Open
Abstract
Psoriasis is a recurring inflammatory skin condition characterized by scaly, red patches on the skin. It affects approximately 3% of the US population and is associated with histological changes such as epidermal hyperplasia, increased blood vessel proliferation, and infiltration of leukocytes into the skin's dermis. T cells, which are classified into various subtypes, have been found to play significant roles in immune-mediated diseases, particularly psoriasis. This paper provides a review of the different T lymphocyte subtypes and their functions in psoriasis, as well as an overview of targeted therapies for treating psoriasis.
Collapse
Affiliation(s)
| | | | | | | | - Ruifang Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
25
|
Qu Y, Li D, Liu W, Shi D. Molecular consideration relevant to the mechanism of the comorbidity between psoriasis and systemic lupus erythematosus (Review). Exp Ther Med 2023; 26:482. [PMID: 37745036 PMCID: PMC10515117 DOI: 10.3892/etm.2023.12181] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/03/2023] [Indexed: 09/26/2023] Open
Abstract
Systemic lupus erythematosus (SLE), a common autoimmune disease with a global incidence and newly diagnosed population estimated at 5.14 (range, 1.4-15.13) per 100,000 person-years and 0.40 million people annually, respectively, affects multiple tissues and organs; for example, skin, blood system, heart and kidneys. Accumulating data has also demonstrated that psoriasis (PS) can be a systemic inflammatory disease, which can affect organs other than the skin and occur alongside other autoimmune diseases, such as inflammatory bowel disease, multiple sclerosis, rheumatoid arthritis and SLE. The current explanations for the possible comorbidity of PS and SLE include: i) The two diseases share susceptible gene loci; ii) they share a common IL-23/T helper 17 (Th17) axis inflammatory pathway; and iii) the immunopathogenesis of the two conditions is a consequence of the interactions between IL-17 cytokines with effector Th17 cells, T regulatory cells, as well as B cells. In addition, the therapeutic efficacy of IL-17 or TNF-α inhibitors has been demonstrated in PS, and has also become evident in SLE. However, the mechanisms have not been investigated. To the best of our knowledge, there remains a lack of substantial studies on the correlation between PS and SLE. In the present review, the literature, with regards to the epidemiology, genetic predisposition, inflammatory mechanisms and treatment of the patients with both PS and SLE, has been reviewed. Further investigations into the molecular pathogenic mechanism may provide drug targets that could benefit the patients with concomitant PS and SLE.
Collapse
Affiliation(s)
- Yuying Qu
- Department of Dermatology, College of Clinical Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Weida Liu
- Department of Medical Mycology, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, Jiangsu 272002, P.R. China
| | - Dongmei Shi
- Department of Dermatology, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
26
|
Silva de Melo BM, Veras FP, Zwicky P, Lima D, Ingelfinger F, Martins TV, da Silva Prado D, Schärli S, Publio G, Hiroki CH, Melo PH, Saraiva A, Norbiato T, Lima L, Ryffel B, Vogl T, Roth J, Waisman A, Nakaya HI, da Silva Souza C, Cunha FQ, Cunha TM, Becher B, Alves-Filho JC. S100A9 Drives the Chronification of Psoriasiform Inflammation by Inducing IL-23/Type 3 Immunity. J Invest Dermatol 2023; 143:1678-1688.e8. [PMID: 36921684 DOI: 10.1016/j.jid.2023.02.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 01/13/2023] [Accepted: 02/09/2023] [Indexed: 03/14/2023]
Abstract
Psoriasis is a chronic inflammatory skin disorder driven by the IL-23/type 3 immune response. However, molecular mechanisms sustaining the chronicity of inflammation and psoriatic lesions remain elusive. Combining systematic analyses of several transcriptomic datasets, we delineated gene signatures across human psoriatic skin, identifying S100A9 as one of the most up-regulated genes, which was confirmed in lesioned skin from patients with psoriasis and preclinical psoriasiform skin inflammation models. Genetic ablation or pharmacologic inhibition of S100A9 alleviated Aldara-induced skin inflammation. By single-cell mapping of human psoriatic skin and bone marrow chimeric mice experiments, we identified keratinocytes as the major source of S100A9. Mechanistically, S100A9 induced IL-23 production by dendritic cells, driving the IL-23/type 3 immunity in psoriasiform skin inflammation. In addition, the cutaneous IL-23/IL-17 axis induced epidermal S100A9 expression in human and experimental psoriasis. Thus, we showed an autoregulatory circuit between keratinocyte-derived S100A9 and IL-23/type 3 immunity during psoriasiform inflammation, identifying a crucial function of S100A9 in the chronification of psoriasis.
Collapse
Affiliation(s)
- Bruno Marcel Silva de Melo
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Flávio Protásio Veras
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Diógenes Lima
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Timna Varela Martins
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Douglas da Silva Prado
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Stefanie Schärli
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Gabriel Publio
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Carlos Hiroji Hiroki
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Paulo Henrique Melo
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - André Saraiva
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thainá Norbiato
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Leonardo Lima
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Department of Cell Biology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Thomas Vogl
- Institute of Immunology, University of Münster, Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, Münster, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University of Mainz, Mainz, Germany
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil; Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Cacilda da Silva Souza
- Department of Internal Medicine, Dermatology Division, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil, Ribeirao Preto, Sao Paulo, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - José C Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
27
|
Lee JY, Lee JH, Lim HJ, Kim E, Kim DK, Choi JK. Aminooxy acetic acid suppresses Th17-mediated psoriasis-like skin inflammation by inhibiting serine metabolism. Front Pharmacol 2023; 14:1215861. [PMID: 37649889 PMCID: PMC10464615 DOI: 10.3389/fphar.2023.1215861] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023] Open
Abstract
Background: Psoriasis is a common chronic inflammatory skin disease characterized by an external red rash that is caused by abnormal proliferation and differentiation of keratinocytes and immune T cells. This study aimed to elucidate the role of aminooxy acetic acid (AOA) in alleviating psoriasis from the perspective of immunology and metabolomics. Therefore, contributing to the development of new drugs as candidates for psoriasis treatment. Methods: To investigate the symptom-alleviating effects and the related mechanisms of AOA on the treatment of psoriasis, we used a 12-O-tetradecanoylphorbol-13-acetate-induced psoriasis-like skin mouse model and interleukin (IL)-17-stimulated human keratinocytes. Results: The results showed that AOA ameliorated psoriasis-related symptoms and decreased inflammation-associated antimicrobial peptides and T-helper 17 (Th17)-associated cytokines in a mouse model of psoriasis. Furthermore, AOA inhibited the activation of mechanistic target of rapamycin (mTOR) by suppressing serine metabolism-related genes. Importantly, mTOR inhibition ameliorated psoriatic disease by affecting the differentiation of various T cells and normalizing the Th17/regulatory T (Treg) cell balance. In addition, IL-17-stimulated human keratinocytes showed the same results as in the in vivo experiments. Conclusion: Taken together, these results suggest that targeting the serine metabolism pathway in the treatment of psoriasis is a novel strategy, and that AOA could be utilized as a novel biologic to treat psoriasis.
Collapse
Affiliation(s)
- Jong Yeong Lee
- Department of Immunology, Jeonbuk National University Medical School, Jeonju-si, Republic of Korea
| | - Ji-Hyun Lee
- Department of Immunology, Jeonbuk National University Medical School, Jeonju-si, Republic of Korea
| | - Hyo Jung Lim
- Department of Immunology, Jeonbuk National University Medical School, Jeonju-si, Republic of Korea
| | - Eonho Kim
- Department of Physical Education, Dongguk University, Seoul, Republic of Korea
| | - Dae-Ki Kim
- Department of Immunology, Jeonbuk National University Medical School, Jeonju-si, Republic of Korea
| | - Jin Kyeong Choi
- Department of Immunology, Jeonbuk National University Medical School, Jeonju-si, Republic of Korea
| |
Collapse
|
28
|
Navarro-Compán V, Puig L, Vidal S, Ramírez J, Llamas-Velasco M, Fernández-Carballido C, Almodóvar R, Pinto JA, Galíndez-Aguirregoikoa E, Zarco P, Joven B, Gratacós J, Juanola X, Blanco R, Arias-Santiago S, Sanz Sanz J, Queiro R, Cañete JD. The paradigm of IL-23-independent production of IL-17F and IL-17A and their role in chronic inflammatory diseases. Front Immunol 2023; 14:1191782. [PMID: 37600764 PMCID: PMC10437113 DOI: 10.3389/fimmu.2023.1191782] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/05/2023] [Indexed: 08/22/2023] Open
Abstract
Interleukin-17 family (IL-17s) comprises six structurally related members (IL-17A to IL-17F); sequence homology is highest between IL-17A and IL-17F, displaying certain overlapping functions. In general, IL-17A and IL-17F play important roles in chronic inflammation and autoimmunity, controlling bacterial and fungal infections, and signaling mainly through activation of the nuclear factor-kappa B (NF-κB) pathway. The role of IL-17A and IL-17F has been established in chronic immune-mediated inflammatory diseases (IMIDs), such as psoriasis (PsO), psoriatic arthritis (PsA), axial spondylarthritis (axSpA), hidradenitis suppurativa (HS), inflammatory bowel disease (IBD), multiple sclerosis (MS), and asthma. CD4+ helper T cells (Th17) activated by IL-23 are well-studied sources of IL-17A and IL-17F. However, other cellular subtypes can also produce IL-17A and IL-17F, including gamma delta (γδ) T cells, alpha beta (αβ) T cells, type 3 innate lymphoid cells (ILC3), natural killer T cells (NKT), or mucosal associated invariant T cells (MAIT). Interestingly, the production of IL-17A and IL-17F by innate and innate-like lymphocytes can take place in an IL-23 independent manner in addition to IL-23 classical pathway. This would explain the limitations of the inhibition of IL-23 in the treatment of patients with certain rheumatic immune-mediated conditions such as axSpA. Despite their coincident functions, IL-17A and IL-17F contribute independently to chronic tissue inflammation having somehow non-redundant roles. Although IL-17A has been more widely studied, both IL-17A and IL-17F are overexpressed in PsO, PsA, axSpA and HS. Therefore, dual inhibition of IL-17A and IL-17F could provide better outcomes than IL-23 or IL-17A blockade.
Collapse
Affiliation(s)
| | - Luis Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Silvia Vidal
- Immunology-Inflammatory Diseases, Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Julio Ramírez
- Arthritis Unit, Department of Rheumatology, Hospital Clínic and Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mar Llamas-Velasco
- Department of Dermatology, Hospital Universitario La Princesa, Madrid, Spain
| | | | - Raquel Almodóvar
- Department of Rheumatology, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - José Antonio Pinto
- Department of Rheumatology, Complejo Hospitalario Universitario de A Coruña, Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
| | | | - Pedro Zarco
- Department of Rheumatology, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Beatriz Joven
- Department of Rheumatology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jordi Gratacós
- Department of Rheumatology, Medicine Department Autonomus University of Barcelona (UAB), I3PT, University Hospital Parc Taulí Sabadell, Barcelona, Spain
| | - Xavier Juanola
- Department of Rheumatology, University Hospital Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Ricardo Blanco
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Salvador Arias-Santiago
- Department of Dermatology, Hospital Universitario Virgen de las Nieves, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
- Department of Dermatology, Facultad de Medicina, Universidad de Granada, Spain
| | - Jesús Sanz Sanz
- Department of Rheumatology, Hospital Universitario Puerta del Hierro Majadahonda, Madrid, Spain
| | - Rubén Queiro
- Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Juan D. Cañete
- Arthritis Unit, Department of Rheumatology, Hospital Clínic and Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
29
|
Hipp AV, Bengsch B, Globig AM. Friend or Foe - Tc17 cell generation and current evidence for their importance in human disease. DISCOVERY IMMUNOLOGY 2023; 2:kyad010. [PMID: 38567057 PMCID: PMC10917240 DOI: 10.1093/discim/kyad010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/12/2023] [Accepted: 07/19/2023] [Indexed: 04/04/2024]
Abstract
The term Tc17 cells refers to interleukin 17 (IL-17)-producing CD8+ T cells. While IL-17 is an important mediator of mucosal defense, it is also centrally involved in driving the inflammatory response in immune-mediated diseases, such as psoriasis, multiple sclerosis, and inflammatory bowel disease. In this review, we aim to gather the current knowledge on the phenotypic and transcriptional profile, the in vitro and in vivo generation of Tc17 cells, and the evidence pointing towards a relevant role of Tc17 cells in human diseases such as infectious diseases, cancer, and immune-mediated diseases.
Collapse
Affiliation(s)
- Anna Veronika Hipp
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Bertram Bengsch
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Anna-Maria Globig
- Clinic for Internal Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, University Medical Center Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
30
|
Singh VK, Sahoo D, Agrahari K, Khan A, Mukhopadhyay P, Chanda D, Yadav NP. Anti-inflammatory, anti-proliferative and anti-psoriatic potential of apigenin in RAW264.7 cells, HaCaT cells and psoriasis like dermatitis in BALB/c mice. Life Sci 2023:121909. [PMID: 37414141 DOI: 10.1016/j.lfs.2023.121909] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/30/2023] [Accepted: 07/02/2023] [Indexed: 07/08/2023]
Abstract
AIMS Psoriasis is an immune-mediated skin disease characterized by keratinocytes hyperproliferation, abnormal differentiation and inflammation. Therefore, this study aimed to investigate in-vitro and in-vivo anti-inflammatory and anti-proliferative activity to evaluate anti-psoriatic potential of apigenin. MAIN METHODS For in-vivo study, 5 % imiquimod cream was used to induce psoriasis-like skin inflammation in BALB/c mice to mimic human psoriatic conditions. PASI scores, CosCam score, histopathology, immunohistochemistry, qRT-PCR, and ELISA were done to evaluate the anti-psoriatic potential of topically applied apigenin. For in-vitro studies, LPS-induced inflammation in RAW264.7 was done, and qRT-PCR, ELISA, and immunofluorescence were conducted to evaluate the anti-inflammatory activity of apigenin. Migration and cell doubling assay in HaCaT cells were performed to assess the anti-proliferative effect of apigenin. Acute dermal toxicity profile of apigenin has also been done as per OECD guidelines. KEY FINDINGS Results showed that apigenin significantly reduce the PASI and CosCam scores, ameliorate the deteriorating histopathology, and effectively downregulated the expression of CCR 6, IL-17A, and NF-κB. Apigenin effectively downregulated the expression and secretion of pro-inflammatory cytokines through IL-23/IL-17/IL-22 axis. Apigenin suppressed nuclear translocation of NF-κB in LPS-induced RAW 264.7 cells. Cell migration and cell doubling assay in HaCaT cells showing the anti-proliferative potential of apigenin. Apigenin was found safe in acute dermal toxicity study. SIGNIFICANCE Apigenin was found effective against psoriasis in both in-vitro and in-vivo models suggesting apigenin as a potential candidate for the development of anti-psoriatic agent.
Collapse
Affiliation(s)
- Vipin Kumar Singh
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Debasish Sahoo
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India
| | - Kirti Agrahari
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India
| | - Ammar Khan
- Plant Biotechnology division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Pradipto Mukhopadhyay
- Plant Biotechnology division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Debabrata Chanda
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Narayan Prasad Yadav
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
31
|
Cannizzaro MV, Coscarella G, Chiricozzi A. Brodalumab in the Treatment of Plaque Psoriasis Localized in Difficult-to-Treat Areas: A Narrative Review. Dermatol Pract Concept 2023; 13:e2023245. [PMID: 37557129 PMCID: PMC10412049 DOI: 10.5826/dpc.1303a245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 08/11/2023] Open
Abstract
INTRODUCTION Psoriasis is a common chronic, immune-mediated, inflammatory skin disease that in certain localization results difficult to treat. Psoriatic lesions in difficult-to-treat areas might be hardly managed as no standardized therapeutic approach and the application of topical treatments might have great limitations. Systemic agents, including biologic therapies, have been proven effective in treating this subgroup of patients. In particular, current evidence has shown beneficial effects with the use of brodalumab, a fully human IgG2 monoclonal antibody antagonizing the IL-17 receptor A subunit (IL-17RA). OBJECTIVES The aim of this narrative review was to collect published data about efficacy and safety of brodalumab in the treatment of psoriasis occurring in difficult-to-treat areas. METHODS Data on brodalumab effectiveness and safety deriving from both trials and real-world setting that had been published in the last 15 years were collected for this review, together with clinical findings issued during international meetings. RESULTS In phase 3 trials, brodalumab demonstrated to be effective in promoting a rapid response in scalp psoriasis as well as in generalized pustular psoriasis and erythrodermic psoriasis. Nail psoriasis demonstrated marked clinical improvement after treatment with brodalumab. Amelioration of palmoplantar psoriasis was also described in brodalumab-treated patients. Various retrospective real-world studies reported a complete clearance of psoriatic lesions in difficult-to-treat areas, including genitalia, through short-term brodalumab treatment. CONCLUSIONS Brodalumab, combining rapid and sustained efficacy with a favorable safety profile, may be a valid therapeutic option for severe variants of psoriasis as well as for psoriasis localized in difficult-to-treat areas.
Collapse
Affiliation(s)
- Maria Vittoria Cannizzaro
- Dermatologia, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giulia Coscarella
- Dermatologia, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Dermatologia, Dipartimento Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Andrea Chiricozzi
- Dermatologia, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Dermatologia, Dipartimento Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
32
|
Schaller T, Ringen J, Fischer B, Bieler T, Perius K, Knopp T, Kommoss KS, Korn T, Heikenwälder M, Oelze M, Daiber A, Münzel T, Kramer D, Wenzel P, Wild J, Karbach S, Waisman A. Reactive oxygen species produced by myeloid cells in psoriasis as a potential biofactor contributing to the development of vascular inflammation. Biofactors 2023; 49:861-874. [PMID: 37139784 DOI: 10.1002/biof.1949] [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: 10/31/2022] [Accepted: 03/04/2023] [Indexed: 05/05/2023]
Abstract
Psoriasis is an immune-mediated inflammatory skin disease driven by interleukin-17A (IL-17A) and associated with cardiovascular dysfunction. We used a severe psoriasis mouse model of keratinocyte IL-17A overexpression (K14-IL-17Aind/+ , IL-17Aind/+ control mice) to investigate the activity of neutrophils and a potential cellular interconnection between skin and vasculature. Levels of dermal reactive oxygen species (ROS) and their release by neutrophils were measured by lucigenin-/luminol-based assays, respectively. Quantitative RT-PCR determined neutrophilic activity and inflammation-related markers in skin and aorta. To track skin-derived immune cells, we used PhAM-K14-IL-17Aind/+ mice allowing us to mark all cells in the skin by photoconversion of a fluorescent protein to analyze their migration into spleen, aorta, and lymph nodes by flow cytometry. Compared to controls, K14-IL-17Aind/+ mice exhibited elevated ROS levels in the skin and a higher neutrophilic oxidative burst accompanied by the upregulation of several activation markers. In line with these results psoriatic mice displayed elevated expression of genes involved in neutrophil migration (e.g., Cxcl2 and S100a9) in skin and aorta. However, no direct immune cell migration from the psoriatic skin into the aortic vessel wall was observed. Neutrophils of psoriatic mice showed an activated phenotype, but no direct cellular migration from the skin to the vasculature was observed. This suggests that highly active vasculature-invading neutrophils must originate directly from the bone marrow. Hence, the skin-vasculature crosstalk in psoriasis is most likely based on the systemic effects of the autoimmune skin disease, emphasizing the importance of a systemic therapeutic approach for psoriasis patients.
Collapse
Affiliation(s)
- Theresa Schaller
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Julia Ringen
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center of Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Berenice Fischer
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Tabea Bieler
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), University of Heidelberg, Heidelberg, Germany
| | - Katharina Perius
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center of Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Tanja Knopp
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center of Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Department of Hematology and Central Hematology Laboratory, Inselspital University Hospital Bern, Bern, Switzerland
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Katharina S Kommoss
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University of Heidelberg, Heidelberg, Germany
| | - Thomas Korn
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- The M3 Research Institute, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Matthias Oelze
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK) - Partner Site Rhine-Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK) - Partner Site Rhine-Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center of Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK) - Partner Site Rhine-Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Daniela Kramer
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Philip Wenzel
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center of Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK) - Partner Site Rhine-Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Johannes Wild
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center of Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK) - Partner Site Rhine-Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Susanne Karbach
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Center of Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK) - Partner Site Rhine-Main, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
33
|
Zhan YP, Chen BS. Drug Target Identification and Drug Repurposing in Psoriasis through Systems Biology Approach, DNN-Based DTI Model and Genome-Wide Microarray Data. Int J Mol Sci 2023; 24:10033. [PMID: 37373186 DOI: 10.3390/ijms241210033] [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: 05/30/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Psoriasis is a chronic skin disease that affects millions of people worldwide. In 2014, psoriasis was recognized by the World Health Organization (WHO) as a serious non-communicable disease. In this study, a systems biology approach was used to investigate the underlying pathogenic mechanism of psoriasis and identify the potential drug targets for therapeutic treatment. The study involved the construction of a candidate genome-wide genetic and epigenetic network (GWGEN) through big data mining, followed by the identification of real GWGENs of psoriatic and non-psoriatic using system identification and system order detection methods. Core GWGENs were extracted from real GWGENs using the Principal Network Projection (PNP) method, and the corresponding core signaling pathways were annotated using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Comparing core signaling pathways of psoriasis and non-psoriasis and their downstream cellular dysfunctions, STAT3, CEBPB, NF-κB, and FOXO1 are identified as significant biomarkers of pathogenic mechanism and considered as drug targets for the therapeutic treatment of psoriasis. Then, a deep neural network (DNN)-based drug-target interaction (DTI) model was trained by the DTI dataset to predict candidate molecular drugs. By considering adequate regulatory ability, toxicity, and sensitivity as drug design specifications, Naringin, Butein, and Betulinic acid were selected from the candidate molecular drugs and combined into potential multi-molecule drugs for the treatment of psoriasis.
Collapse
Affiliation(s)
- Yu-Ping Zhan
- Laboratory of Automatic Control, Signal Processing and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Bor-Sen Chen
- Laboratory of Automatic Control, Signal Processing and Systems Biology, Department of Electrical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
34
|
Fargnoli MC, Bardazzi F, Bianchi L, Dapavo P, Fabbrocini G, Gisondi P, Micali G, Offidani AM, Pellacani G, Skroza N, Angileri RG, Burlando M, Campanati A, Carrera CG, Chiricozzi A, Conti A, Simone CD, Di Lernia V, Errichetti E, Galluzzo M, Guarneri C, Lasagni C, Lembo S, Loconsole F, Megna M, Musumeci ML, Prignano F, Richetta AG, Trovato E, Venturini M, Peris K, Pinton PC. Brodalumab for the Treatment of Moderate-to-Severe Psoriasis: An Expert Delphi Consensus Statement. J Clin Med 2023; 12:jcm12103545. [PMID: 37240650 DOI: 10.3390/jcm12103545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/05/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Brodalumab is a recombinant, fully human immunoglobulin IgG2 monoclonal antibody specifically targeted against interleukin-17RA that has been approved for the treatment of moderate-to-severe psoriasis in Europe. We developed a Delphi consensus document focused on brodalumab for the treatment of moderate-to-severe psoriasis. Based on published literature and their clinical experience a steering committee drafted 17 statements covering 7 domains specific to the treatment of moderate-to-severe psoriasis with brodalumab. A panel of 32 Italian dermatologists indicated their level of agreement using a 5-point Likert scale (from 1 = "strongly disagree" to 5 = "strongly agree") using an online modified Delphi method. After the first round of voting (32 participants), positive consensus was reached for 15/17 (88.2%) of the proposed statements. Following a face-to-face virtual meeting, the steering committee decided that 5 statements would form "main principles" and 10 statements formed the final list. After a second round of voting, consensus was reached in 4/5 (80%) of the main principles and 8/10 (80%) for consensus statements. The final list of 5 main principles and 10 consensus statements identify key indications specific to the use of brodalumab in the treatment of moderate-to-severe psoriasis in Italy. These statements aid dermatologists in the management of patients with moderate-to-severe psoriasis.
Collapse
Affiliation(s)
- Maria Concetta Fargnoli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
- Dermatology Unit, Ospedale San Salvatore, 67100 L'Aquila, Italy
| | - Federico Bardazzi
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico S. Orsola Malpighi, 40126 Bologna, Italy
| | - Luca Bianchi
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
- Dermatology Unit, Azienda Ospedaliera Universitaria "Policlinico Tor Vergata", 00133 Rome, Italy
| | - Paolo Dapavo
- Dermatology Clinic, Department of Medical Sciences, University of Turin, 10124 Turin, Italy
| | - Gabriella Fabbrocini
- Section of Dermatology, Department of Clinical, Medicine and Surgery, University of Naples Federico II, 80138 Naples, Italy
| | - Paolo Gisondi
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Giuseppe Micali
- Department of Dermatology, University of Catania, 95123 Catania, Italy
| | - Anna Maria Offidani
- Department of Clinical and Molecular Sciences, Dermatology Unit, Polytechnic Marche University, 60121 Ancona, Italy
| | - Giovanni Pellacani
- Dermatology Clinic, Department of Clinical Internal, Anesthesiologic and Cardiovascular Sciences, Sapienza Medical School, Sapienza University of Rome, 00185 Rome, Italy
| | - Nevena Skroza
- Dermatology Unit "D. Innocenzi", Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome-Polo Pontino, 04100 Latina, Italy
| | | | - Martina Burlando
- Clinica Dermatologica, DissaL, Ospedale Policlinico San Martino-IRCCS, 16132 Genova, Italy
| | - Anna Campanati
- Department of Clinical and Molecular Sciences, Dermatology Clinic, Polytechnic Marche University, 60121 Ancona, Italy
| | - Carlo Giovanni Carrera
- Dermatology Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Andrea Chiricozzi
- Dermatologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- UOC di Dermatologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
| | - Andrea Conti
- Dermatologic Unit, Department of Surgery, Infermi Hospital, AUSL Romagna, 47923 Rimini, Italy
| | - Clara De Simone
- Dermatologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- UOC di Dermatologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
| | - Vito Di Lernia
- Dermatology Unit, Arcispedale S. Maria Nuova, Azienda USL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy
| | - Enzo Errichetti
- Institute of Dermatology, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), University of Udine, 33100 Udine, Italy
| | - Marco Galluzzo
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
- Dermatology Unit, Azienda Ospedaliera Universitaria "Policlinico Tor Vergata", 00133 Rome, Italy
| | - Claudio Guarneri
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98122 Messina, Italy
| | - Claudia Lasagni
- Clinica Dermatologica, Dipartimento delle Medicine Specialistiche AOU Policlinico di Modena, 41121 Modena, Italy
| | - Serena Lembo
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", University of Salerno, 84084 Fisciano, Italy
| | - Francesco Loconsole
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, 70121 Bari, Italy
- Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, 70124 Bari, Italy
| | - Matteo Megna
- Section of Dermatology, Department of Clinical, Medicine and Surgery, University of Naples Federico II, 80138 Naples, Italy
| | | | - Francesca Prignano
- Department of Health Sciences, Section of Dermatology, University of Florence, 50125 Florence, Italy
| | - Antonio Giovanni Richetta
- Unit of Dermatology, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, 00185 Rome, Italy
| | - Emanuele Trovato
- Section of Dermatology, Department of Medical, Surgical and Neurological Science, S. Maria alle Scotte Hospital, University of Siena, 53100 Siena, Italy
| | - Marina Venturini
- Dermatology Department, University of Brescia, 25121 Brescia, Italy
| | - Ketty Peris
- Dermatologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- UOC di Dermatologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
| | | |
Collapse
|
35
|
Li X, Wang W, Ding X. Pan-cancer investigation of psoriasis-related BUB1B gene: genetical alteration and oncogenic immunology. Sci Rep 2023; 13:6058. [PMID: 37055476 PMCID: PMC10102166 DOI: 10.1038/s41598-023-33174-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/08/2023] [Indexed: 04/15/2023] Open
Abstract
Unknown factors contribute to psoriasis' hyperproliferative, chronic, inflammatory, and arthritic features. Psoriasis patients have been linked to an increased risk of cancer, though the underlying genetics remain unknown. Since our prior research indicated that BUB1B contributes to the development of psoriasis, we designed and carried out this investigation using bioinformatics analysis. Using the TCGA database, we investigated the oncogenic function of BUB1B in 33 tumor types. To sum up, our work sheds light on BUB1B's function in pan-cancer from various perspectives, including its pertinent signaling pathways, mutation locations, and connection to immune cell infiltration. BUB1B was shown to have a non-negligible role in pan-cancer, which is connected to immunology, cancer stemness, and genetic alterations in a variety of cancer types. BUB1B is highly expressed in a variety of cancers and may serve as a prognostic marker. This study is anticipated to offer molecular details on the elevated cancer risk that psoriasis sufferers experience.
Collapse
Affiliation(s)
- Xiaobin Li
- Department of Orthopedic Surgery, Linping Campus, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenwen Wang
- Department of Dermatology and Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiaoxia Ding
- Center for Plastic and Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
36
|
Sorokin AV, Arnardottir H, Svirydava M, Ng Q, Baumer Y, Berg A, Pantoja CJ, Florida E, Teague HL, Yang ZH, Dagur PK, Powell-Wiley TM, Yu ZX, Playford MP, Remaley AT, Mehta NN. Comparison of the dietary omega-3 fatty acids impact on murine psoriasis-like skin inflammation and associated lipid dysfunction. J Nutr Biochem 2023; 117:109348. [PMID: 37044136 DOI: 10.1016/j.jnutbio.2023.109348] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023]
Abstract
Persistent skin inflammation and impaired resolution are the main contributors to psoriasis and associated cardiometabolic complications. Omega-3 polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are known to exert beneficial effects on inflammatory response and lipid function. However, a specific role of omega-3 PUFAs in psoriasis and accompanied pathologies are still a matter of debate. Here, we carried out a direct comparison between EPA and DHA 12 weeks diet intervention treatment of psoriasis-like skin inflammation in the K14-Rac1V12 mouse model. By utilizing sensitive techniques, we targeted EPA- and DHA-derived specialized pro-resolving lipid mediators and identified tightly connected signaling pathways by RNA sequencing. Treatment with experimental diets significantly decreased circulating pro-inflammatory cytokines and bioactive lipid mediators, altered psoriasis macrophage phenotypes and genes of lipid oxidation. The superficial role of these changes was related to DHA treatment and included increased levels of resolvin D5, protectin DX and maresin 2 in the skin. EPA treated mice had less pronounced effects but demonstrated a decreased skin accumulation of prostaglandin E2 and thromboxane B2. These results indicate that modulating psoriasis skin inflammation with the omega-3 PUFAs may have clinical significance and DHA treatment might be considered over EPA in this specific disease.
Collapse
Affiliation(s)
- Alexander V Sorokin
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Hildur Arnardottir
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institute, Sweden
| | - Maryia Svirydava
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Qimin Ng
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander Berg
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carla J Pantoja
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth Florida
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heather L Teague
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhi-Hong Yang
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pradeep K Dagur
- Flow Cytometry Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tiffany M Powell-Wiley
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zu-Xi Yu
- Pathology Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martin P Playford
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
37
|
Wang J, Liu Y, Zhang Y, Wang S, Kang S, Mi N, Li R, Zou Y. Identification immune response genes in psoriasis after treatment with secukinumab. BMC Med Genomics 2023; 16:77. [PMID: 37029373 PMCID: PMC10082531 DOI: 10.1186/s12920-023-01507-w] [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: 08/31/2022] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Secukinumab is a fully human IgG1κ MoAb that selectively binds to IL-17A with high affinity, and it has been proven effective for the treatment of psoriasis. However, the immune response pathways and mechanisms during the treatment are still masked. Therefore, the current study was designed to investigate the potential immune response genes via bioinformatics approaches. METHODS Gene expression data of severe plaque-type psoriasis was retrieved from the GEO database. Quantification of immune infiltration by ssGSEA and identification of differentially infiltrated immune cells were conducted to validate the treatment effect of secukinumab. After data processing, differentially expressed genes were identified between the treatment and untreated group. TC-seq was employed to analyze the trend of gene expression and clustering analysis. IL-17 therapeutic immune response genes were selected by taking the intersection of the genes inside the key cluster set and the MAD3-PSO geneset. Based on these therapeutic response genes, protein-protein interaction networks were built for key hub gene selection. These hub genes would work as potential immune response genes, and be validated via an external dataset. RESULTS Enrichment scores calculated by ssGSEA illustrated that the immune infiltration level of T cells had a strong difference before and after medication, which validated the treatment effect of Secukinumab. 1525 genes that have significantly different expression patterns before and after treatment were extracted for further analysis, and the enrichment result shows that these genes have the function related to epidermal development, differentiation, and keratinocytes differentiation. After overlapping candidate genes with MAD3-PSO gene set, 695 genes were defined as anti-IL7A treatment immune response genes, which were mainly enriched in receptor signaling and IL-17 signaling pathways. Hub gene were pinpointed from the PPI network constructed by anti-IL7A treatment immune response genes, their expression pattern fits TC-seq gene expression pattern. CONCLUSION Our study revealed the potential anti-IL7A treatment immune response genes, and the central hub genes, which may act critical roles in Secukinumab, induced immune response. This would open up a novel and effective avenue for the treatment of psoriasis.
Collapse
Affiliation(s)
- Jing Wang
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Yufang Liu
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Yuxin Zhang
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Shiyan Wang
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Shaomei Kang
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Ningyu Mi
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Ruxin Li
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Yulin Zou
- Department of Dermatology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
- Department of Dermatology, Jinzhou Medical University Graduate Training Base, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
| |
Collapse
|
38
|
Zapata-Salazar NA, Kubelis-Lopez DE, Salinas-Santander MA, Sanchez-Dominguez CN, Xolalpa-Rosales AC, Gomez-Galindo ME, Ocampo-Candiani J. Association of rs4711998 of IL-17A, rs2275913 of IL-17A and rs763780 IL-17F gene polymorphisms with non-segmental vitiligo in a Mexican population. Arch Dermatol Res 2023; 315:447-454. [PMID: 35960353 DOI: 10.1007/s00403-022-02382-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/27/2022] [Accepted: 08/02/2022] [Indexed: 12/27/2022]
Abstract
Vitiligo is the most common depigmenting disease characterized by achromic macules due to selective loss of melanocytes. The pathogenesis remains poorly elucidated, and multiple hypotheses exist regarding its pathogenesis. Evidence suggests that stress on melanocytes can result in activation of the immune system, and involvement of both activated cluster of differentiation (CD8+) cytotoxic and CD4+ T cells in the dysfunction, depigmentation, and apoptosis of melanocytes. Recent studies show that the interleukin 17 (IL-17) axis plays a central role in the pathogenesis of the disease. IL-17 is an important regulatory effector cytokine in this pathway. The aim of this study was to evaluate the association of IL-17A rs4711998 (-832A/G), IL-17A rs2275913 (-197G/A), and IL-17F rs763780 (7488A/G) with vitiligo in a Northeastern Mexican population. This was a case-control study and included 116 patients with vitiligo and 116 control subjects. Genotype characterization of IL-17A rs4711998 (-832A/G), IL-17A rs2275913 (-197G/A), and IL-17F rs763780 (7488A/G) was performed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. A p ≤ 0.05 was considered significant. It was observed that the combination of the genotypes GG/GA for IL-17F rs763780 (7488A/G) was associated with an increased risk for the development of vitiligo (OR 2.0943, 95% Cl 1.2375-3.5445, p = 0.0056). Regarding IL-17A rs4711998 (-832A/G) and IL-17A rs2275913 (-197G/A) genotyping, no association with vitiligo development was found. In conclusion, the SNP rs763780 in the IL-17F gene appears to be a risk factor for vitiligo development in this Mexican population and it may be useful in future studies, especially for the development of new therapies.
Collapse
Affiliation(s)
- Natalia Aranza Zapata-Salazar
- Department of Dermatology, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon (UANL), Av. Madero and Gonzalitos S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | - David Emmanuel Kubelis-Lopez
- Department of Dermatology, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon (UANL), Av. Madero and Gonzalitos S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico
| | | | - Celia Nohemi Sanchez-Dominguez
- Department of Biochemistry and Molecular Medicine, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon (UANL), Monterrey, Nuevo Leon, Mexico
| | - Ana Cecilia Xolalpa-Rosales
- Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon (UANL), Monterrey, Nuevo Leon, Mexico
| | - Marely Eugenia Gomez-Galindo
- Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon (UANL), Monterrey, Nuevo Leon, Mexico
| | - Jorge Ocampo-Candiani
- Department of Dermatology, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autonoma de Nuevo Leon (UANL), Av. Madero and Gonzalitos S/N, Mitras Centro, 64460, Monterrey, Nuevo Leon, Mexico.
| |
Collapse
|
39
|
Rompoti N, Politou M, Stefanaki I, Vavouli C, Papoutsaki M, Neofotistou A, Rigopoulos D, Stratigos A, Nicolaidou E. Brodalumab in plaque psoriasis: Real-world data on effectiveness, safety and clinical predictive factors of initial response and drug survival over a period of 104 weeks. J Eur Acad Dermatol Venereol 2023; 37:689-697. [PMID: 36562663 DOI: 10.1111/jdv.18825] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Brodalumab, a fully human IgG2k antibody blocking the receptor of IL17, is characterized by a rapid onset of action with high skin clearance rates in clinical trials. Since setting PASI90/100 or absolute PASI ≤ 3 as treatment goals have become attainable, evaluating the effectiveness and safety profile of biologic agents, such as brodalumab, in a real-world setting is essential. OBJECTIVE The aim of this study was to evaluate the effectiveness and safety profile of brodalumab over a period of 104 weeks in everyday practice. Clinical predictive factors of initial (week 12/16) response to treatment and long-term drug survival were also investigated. METHODS In this monocentric, retrospective study, PASI90/100 and absolute PASI ≤ 1/3 were assessed in 91 patients with moderate-to-severe skin psoriasis under brodalumab at weeks 12/16, 24, 52 and 104 of treatment. At week 12/16, patients with an absolute PASI ≤ 3 were defined as 'initial responders' and ≤1 as 'super-responders'. Clinical parameters, such as age, gender, BMI, comorbidities and previous systemic treatment, were assessed in order to predict 'super-responders'. Drug survival and its prognostic factors were also evaluated. RESULTS PASI90/100 has reached 81.1/66.0% in week 12/16. This response rate increased at week 104, where 87.1/80.7% had PASI90/100 and 84.9% had absolute PASI ≤ 1. The presence of >3 comorbidities, prior treatment with >2 systemic agents and obesity tended to be negative predictive factors of 'super-response'. Previous exposure to IL17 inhibitors had no impact on both PASI < 1 and PASI < 3 initial response. One- and two-year drug survival probability was 87.6% and 77.32%, respectively. 'Initial responders' and anti-IL17 drug-naïve patients had better drug survival. Drug discontinuation occurred in 24.2%, mostly due to secondary failure, and arthralgia was the most common adverse event that led to discontinuation. CONCLUSIONS Our study confirms the high effectiveness and good safety profile of brodalumab in the real-world setting.
Collapse
Affiliation(s)
- Natalia Rompoti
- Department of Dermatology-Venereology, Faculty of Medicine, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases, Athens, Greece
| | - Maria Politou
- Department of Dermatology-Venereology, Faculty of Medicine, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases, Athens, Greece
| | - Irene Stefanaki
- Department of Dermatology-Venereology, Faculty of Medicine, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases, Athens, Greece
| | - Charitomeni Vavouli
- Department of Dermatology-Venereology, Faculty of Medicine, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases, Athens, Greece
| | - Marina Papoutsaki
- Department of Dermatology-Venereology, Faculty of Medicine, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases, Athens, Greece
| | - Afroditi Neofotistou
- Department of Dermatology-Venereology, Faculty of Medicine, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases, Athens, Greece
| | - Dimitrios Rigopoulos
- Department of Dermatology-Venereology, Faculty of Medicine, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases, Athens, Greece
| | - Alexander Stratigos
- Department of Dermatology-Venereology, Faculty of Medicine, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases, Athens, Greece
| | - Electra Nicolaidou
- Department of Dermatology-Venereology, Faculty of Medicine, National and Kapodistrian University of Athens, 'A. Sygros' Hospital for Skin and Venereal Diseases, Athens, Greece
| |
Collapse
|
40
|
Lee SB, Kang JH, Sim EJ, Jung YR, Kim JH, Hillman PF, Nam SJ, Kang TB. Cornus officinalis Seed Extract Inhibits AIM2-Inflammasome Activation and Attenuates Imiquimod-Induced Psoriasis-like Skin Inflammation. Int J Mol Sci 2023; 24:ijms24065653. [PMID: 36982727 PMCID: PMC10051512 DOI: 10.3390/ijms24065653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/07/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
The AIM2 inflammasome is an innate immune system component that defends against cytosolic bacteria and DNA viruses, but its aberrant activation can lead to the progression of various inflammatory diseases, including psoriasis. However, there have been few reports of specific inhibitors of AIM2 inflammasome activation. In this study, we aimed to investigate the inhibitory activity of ethanolic extracts of seeds of Cornus officinalis (CO), a herb and food plant used in traditional medicine, on AIM2-inflammasome activation. We found that CO inhibited the release of IL-1β induced by dsDNA in both BMDMs and HaCaT cells, but that it showed no effect on the release of IL-1β induced by NLRP3 inflammasome triggers, such as nigericin and silica, or the NLRC4 inflammasome trigger flagellin. Furthermore, we demonstrated that CO inhibited the cleavage of caspase-1, an inflammasome activation marker, and an upstream event, the translocation and speck formation of ASC. In addition, further experiments and mechanistic investigations revealed that CO can inhibit AIM2 speck formation induced by dsDNA in AIM2-overexpressing HEK293T cells. To verify the correlation in vivo, we investigated the efficacy of CO in an imiquimod (IMQ)-induced psoriasis model, which has reported associations with the AIM2 inflammasome. We found that topical application of CO alleviated psoriasis-like symptoms, such as erythema, scaling, and epidermal thickening, in a dose-dependent manner. Moreover, CO also significantly decreased IMQ-induced expression of AIM2 inflammasome components, including AIM2, ASC, and caspase-1, and led to the elevation of serum IL-17A. In conclusion, our results suggest that CO may be a valuable candidate for the discovery of AIM2 inhibitors and the regulation of AIM2-related diseases.
Collapse
Affiliation(s)
- Se-Bin Lee
- BK21 Project Team, Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Ju-Hui Kang
- BK21 Project Team, Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Eun-Jung Sim
- BK21 Project Team, Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Ye-Rin Jung
- BK21 Project Team, Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
| | - Jeong-Hyeon Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Prima F. Hillman
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sang-Jip Nam
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Tae-Bong Kang
- BK21 Project Team, Department of Applied Life Science, Graduate School, Konkuk University, Chungju 27478, Republic of Korea
- Department of Biotechnology, Research Institute of Inflammatory Diseases, Research Institute (RIBHS), College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea
- Correspondence: ; Tel.: +82-043-840-3904
| |
Collapse
|
41
|
Zhou Y, Li N, Fan X, Xu M, Wang B. Intranasal streptococcal infection exacerbates psoriasis-like dermatitis via the induction of skin tissue-resident memory T cells. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166629. [PMID: 36563916 DOI: 10.1016/j.bbadis.2022.166629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
Recurrent streptococcal tonsillitis exacerbates psoriasis. Studies have indicated that T cells responding to streptococcal antigens in the skin are involved in the pathogenesis of the disease. However, a direct link between streptococcal tonsillitis and psoriasis has not been evidenced. In the present study, the impact of intranasal (i.n.) streptococcal infection on psoriasis was investigated using the imiquimod (IMQ) psoriasis mouse model. The results showed that repeated i.n. infection with group A Streptococcus (GAS) induced a robust and persistent Th17 response in the nasal-associated lymphoid tissue (NALT) and exacerbated IMQ-mediated psoriatic skin lesions. ELISpot and flow cytometry analyses revealed that GAS-reactive tissue-resident memory T cells (TRM) were present in the skin of GAS-infected mice and produced IL-17/IL-23 axis cytokines in response to IMQ, compared to mice uninfected with GAS. In addition, i.n. infection with Streptococcus pneumoniae (Sp), a pathogen not associated with the development of psoriasis, also induced a persistent Th17 response in NALT but did not exacerbate IMQ-induced psoriatic inflammation nor elicited Sp-specific T cells in the skin. The results provide in vivo evidence that GAS-associated psoriasis is dependent on the skin GAS-specific TRM cells induced by GAS nasopharyngeal infection and can be later activated by environmental triggers, leading to psoriatic inflammation. Reducing the reservoir of Th17 cells, which are source of skin TRM cells, may constitute a promising treatment for psoriasis.
Collapse
Affiliation(s)
- Ya Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Ning Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Xin Fan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Meiyi Xu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China; Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics and Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Beinan Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
42
|
Springall R, Ortega-Springall MF, Guerrero-Ponce AE, Vega-Memije ME, Amezcua-Guerra LM. Interleukin-17 and Tumor Necrosis Factor Show a Functional Hierarchy to Regulate the Production of Matrix Metalloproteases by Monocytes from Patients with Psoriasis. J Interferon Cytokine Res 2023; 43:140-146. [PMID: 36939813 DOI: 10.1089/jir.2022.0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Interleukin-17 (IL-17) and tumor necrosis factor (TNF) regulate tissue remodeling through matrix metalloproteinases (MMPs). It is not yet clear whether these cytokines have a functional hierarchy in psoriasis. Serum levels of TNF (1,403 versus 1,058 pg/mL), IL-17 (1,528 versus 820 pg/mL), MMP-1 (1,999 versus 1,039 pg/mL), and MMP-9 (1,950 versus 1,561 pg/mL) were higher in psoriasis subjects (n = 60) than in control subjects (n = 60). Tissue inhibitor of MMPs (TIMP-1; 1,374 versus 1,218 pg/mL) was lower in psoriasis subjects. Serum IL-17 was correlated with MMP-2 (rs = 0.40) and TIMP-1 (rs = -0.26) levels. Unstimulated production of MMP-1, MMP-2, and MMP-9 by monocytes was higher in psoriasis subjects, whereas TIMP-1 production was lower. TNF stimulation increased all MMPs, whereas TIMP-1 production was unchanged. IL-17 stimulation increased all MMPs, whereas TIMP-1 production was decreased in psoriasis subjects. MMP-9 production was higher in monocytes stimulated with IL-17 compared with TNF. TIMP-1 production was decreased more by IL-17 than by TNF, but only in psoriasis cells. MMP-1/TIMP-1, MMP-2/TIMP-1, and MMP-9/TIMP-1 ratios were higher after IL-17 stimulation (compared with TNF stimulation) in psoriasis subjects; this occurred in controls only for the MMP-2/TIMP-1 ratio. IL-17 has a greater ability than TNF to dysregulate the MMPs/TIMP-1 balance, supporting IL-17 blockade as first-line treatment in cutaneous psoriasis.
Collapse
Affiliation(s)
- Rashidi Springall
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | | | | | | - Luis M Amezcua-Guerra
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
- Department of Health Care, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| |
Collapse
|
43
|
Zhang L, Ma X, Shi R, Zhang L, Zhao R, Duan R, Qin Y, Gao S, Li X, Duan J, Li J. Allicin ameliorates imiquimod-induced psoriasis-like skin inflammation via disturbing the interaction of keratinocytes with IL-17A. Br J Pharmacol 2023; 180:628-646. [PMID: 36355777 DOI: 10.1111/bph.15983] [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: 05/14/2022] [Revised: 09/13/2022] [Accepted: 10/20/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Psoriasis is an inflammatory skin disease of chronic recurrence mediated by the interaction between IL-17 and keratinocytes, which sustains a vicious circle of inflammation. Safe and effective natural medicine is a potential strategy for the clinical treatment of psoriasis. Given its prominent anti-proliferative and anti-inflammatory properties, we investigated the actions of allicin in improving psoriasis. EXPERIMENTAL APPROACH Pharmacodynamic studies were carried out in mice after topical administration of allicin against psoriasis-like lesions induced by imiquimod. Skin sensitization tests were evaluated on guinea pigs. Toxicological studies and skin irritation tests were assessed by consecutive topical allicin alone on the skin of rabbits. RNA-seq probed transcriptomic changes following allicin. Western blot explored the actions of allicin on the interaction between IL-17A and keratinocytes. Changes in inflammatory factor expression were analysed by qPCR and immunohistochemistry. KEY RESULTS Allicin significantly improved the epidermal structure by inhibiting the excessive proliferation and reduced apoptosis of keratinocytes. Furthermore, allicin reduced the secretion of inflammatory cytokines (IL-17A/F, IL-22, IL-12, and IL-20), chemokines (CXCL2, CXCL5, and CCL20), and anti-bacterial peptides (S100a8/9). Mechanistically, allicin directly inhibited the IL-17-induced TRAF6/MAPK/NF-κB and STAT3/NF-κB signalling cascades in keratinocytes, thus breaking the positive inflammatory feedback and alleviating imiquimod-induced psoriasis-like dermatitis in mice. Importantly, topical administration of allicin did not cause skin allergy, and the safety and adaptability of long-term application were verified. CONCLUSIONS AND IMPLICATIONS Interfering with IL-17 signalling in keratinocytes with allicin is a promising strategy for treating psoriasis, given its safety and effectiveness.
Collapse
Affiliation(s)
- Lu Zhang
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Xuehong Ma
- College of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Rongmei Shi
- College of Pharmacy, Xinjiang Medical University, Urumqi, China.,Key Laboratory of Garlic Medicinal Research in Xinjiang, Urumqi, China
| | - Libo Zhang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ruolin Zhao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ran Duan
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yuanyuan Qin
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Sijia Gao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xinxia Li
- College of Pharmacy, Xinjiang Medical University, Urumqi, China.,Key Laboratory of Garlic Medicinal Research in Xinjiang, Urumqi, China
| | - Jingjing Duan
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jianguang Li
- Xinjiang University of Science and Technology, Korla, China
| |
Collapse
|
44
|
Roy T, Banang-Mbeumi S, Boateng ST, Ruiz EM, Chamcheu RCN, Kang L, King JA, Walker AL, Nagalo BM, Kousoulas KG, Esnault S, Huang S, Chamcheu JC. Dual targeting of mTOR/IL-17A and autophagy by fisetin alleviates psoriasis-like skin inflammation. Front Immunol 2023; 13:1075804. [PMID: 36741386 PMCID: PMC9889994 DOI: 10.3389/fimmu.2022.1075804] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
Psoriasis is a chronic autoimmune inflammatory skin disorder characterized by epidermal hyperplasia and aberrant immune response. In addition to aberrant cytokine production, psoriasis is associated with activation of the Akt/mTOR pathway. mTOR/S6K1 regulates T-lymphocyte activation and migration, keratinocytes proliferation and is upregulated in psoriatic lesions. Several drugs that target Th1/Th17 cytokines or their receptors have been approved for treating psoriasis in humans with variable results necessitating improved therapies. Fisetin, a natural dietary polyphenol with anti-oxidant and anti-proliferative properties, covalently binds mTOR/S6K1. The effects of fisetin on psoriasis and its underlying mechanisms have not been clearly defined. Here, we evaluated the immunomodulatory effects of fisetin on Th1/Th17-cytokine-activated adult human epidermal keratinocytes (HEKa) and anti-CD3/CD28-stimulated inflammatory CD4+ T cells and compared these activities with those of rapamycin (an mTOR inhibitor). Transcriptomic analysis of HEKa revealed 12,713 differentially expressed genes (DEGs) in the fisetin-treated group compared to 7,374 DEGs in the rapamycin-treated group, both individually compared to a cytokine treated group. Gene ontology analysis revealed enriched functional groups related to PI3K/Akt/mTOR signaling pathways, psoriasis, and epidermal development. Using in silico molecular modeling, we observed a high binding affinity of fisetin to IL-17A. In vitro, fisetin significantly inhibited mTOR activity, increased the expression of autophagy markers LC3A/B and Atg5 in HEKa cells and suppressed the secretion of IL-17A by activated CD4+ T lymphocytes or T lymphocytes co-cultured with HEKa. Topical administration of fisetin in an imiquimod (IMQ)-induced mouse psoriasis model exhibited a better effect than rapamycin in reducing psoriasis-like inflammation and Akt/mTOR phosphorylation and promoting keratinocyte differentiation and autophagy in mice skin lesions. Fisetin also significantly inhibited T-lymphocytes and F4/80+ macrophage infiltration into skin. We conclude that fisetin potently inhibits IL-17A and the Akt/mTOR pathway and promotes keratinocyte differentiation and autophagy to alleviate IMQ-induced psoriasis-like disease in mice. Altogether, our findings suggest fisetin as a potential treatment for psoriasis and possibly other inflammatory skin diseases.
Collapse
Affiliation(s)
- Tithi Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States
| | - Sergette Banang-Mbeumi
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States
- School of Nursing and Allied Health Sciences, Louisiana Delta Community College, Monroe, LA, United States
| | - Samuel T. Boateng
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States
| | - Emmanuelle M. Ruiz
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Roxane-Cherille N. Chamcheu
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States
| | - Lin Kang
- Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, LA, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Judy A. King
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Anthony L. Walker
- School of Clinical Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States
| | - Bolni Marius Nagalo
- Department of Pathology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, United States
- The Winthrop P. Rockefeller Cancer Institute, UAMS, Little Rock, AR, United States
| | - Konstantin G. Kousoulas
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Stephane Esnault
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, Madison, WI, United States
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Department of Hematology and Oncology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Jean Christopher Chamcheu
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA, United States
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
45
|
Balato A, Scala E, Eyerich K, Brembilla NC, Chiricozzi A, Sabat R, Ghoreschi K. Management of Infections in Psoriatic Patients Treated with Systemic Therapies: A Lesson from the Immunopathogenesis of Psoriasis. Dermatol Pract Concept 2023; 13:dpc.1301a16. [PMID: 36892377 PMCID: PMC9946081 DOI: 10.5826/dpc.1301a16] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Modern treatments continue to be developed based on identifying targets within the innate and adaptive immune pathways associated with psoriasis. Whilst there is a sound biologic rationale for increased risk of infection following treatment with immunomodulators, the clinical evidence is confounded by these agents being used in patients affected with several comorbidities. In an era characterized by an ever greater and growing risk of infections, it is necessary to always be updated on this risk. In this mini-review, we will discuss recent updates in psoriasis immunopathogenesis as a rationale for systemic therapy, outline the risk of infections linked to the disease itself and systemic therapy as well, and provide an overview of the prevention and management of infections.
Collapse
Affiliation(s)
- Anna Balato
- Dermatology Unit, University of Campania, Naples, Italy
| | - Emanuele Scala
- Division of Dermatology and Venereology, Department of Medicine Solna, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Dermatology and Venereology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kilian Eyerich
- Division of Dermatology and Venereology, Department of Medicine Solna, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Dermatology and Venereology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Dermatology and Venereology, Unit of Dermatology, Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Robert Sabat
- Interdisciplinary Group of Molecular Immunopathology, Dermatology/Medical Immunology, Charité-Universitätsmedizin, Berlin, Germany.,Psoriasis Research and Treatment Center, Department of Dermatology and Allergy and Institute of Medical Immunology, Charité-Universitätsmedizin, Berlin, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin, Berlin, Germany
| |
Collapse
|
46
|
Veras FP, Publio GA, Melo BM, Prado DS, Norbiato T, Cecilio NT, Hiroki C, Damasceno LEA, Jung R, Toller-Kawahisa JE, Martins TV, Assunção SF, Lima D, Alves MG, Vieira GV, Tavares LA, Alves-Rezende ALR, Karbach SH, Nakaya HI, Cunha TM, Souza CS, Cunha FQ, Sales KU, Waisman A, Alves-Filho JC. Pyruvate kinase M2 mediates IL-17 signaling in keratinocytes driving psoriatic skin inflammation. Cell Rep 2022; 41:111897. [PMID: 36577385 DOI: 10.1016/j.celrep.2022.111897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 11/17/2022] [Accepted: 12/08/2022] [Indexed: 12/28/2022] Open
Abstract
Psoriasis is an inflammatory skin disease characterized by keratinocyte proliferation and inflammatory cell infiltration induced by IL-17. However, the molecular mechanism through which IL-17 signaling in keratinocytes triggers skin inflammation remains not fully understood. Pyruvate kinase M2 (PKM2), a glycolytic enzyme, has been shown to have non-metabolic functions. Here, we report that PKM2 mediates IL-17A signaling in keratinocytes triggering skin psoriatic inflammation. We find high expression of PKM2 in the epidermis of psoriatic patients and mice undergoing psoriasis models. Specific depletion of PKM2 in keratinocytes attenuates the development of experimental psoriasis by reducing the production of pro-inflammatory mediators. Mechanistically, PKM2 forms a complex with Act1 and TRAF6 regulating NF-κB transcriptional signaling downstream of the IL-17 receptor. As IL-17 also induces PKM2 expression in keratinocytes, our findings reveal a sustained signaling circuit critical for the psoriasis-driving effects of IL-17A, suggesting that PKM2 is a potential therapeutic target for psoriasis.
Collapse
Affiliation(s)
- Flávio P Veras
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University of Mainz, Mainz, Germany.
| | - Gabriel A Publio
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruno M Melo
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Douglas S Prado
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Thainá Norbiato
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Nerry T Cecilio
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Carlos Hiroki
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luis Eduardo A Damasceno
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rebecca Jung
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University of Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg, University of Mainz, Mainz, Germany
| | - Juliana E Toller-Kawahisa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Timna V Martins
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Stella F Assunção
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Diogenes Lima
- Department of Clinical and Toxicological Analyses of the School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcia G Alves
- Department of Cell Biology, Ribeirão Preto Medical School University of São Paulo, Ribeirão Preto, Brazil
| | - Gabriel V Vieira
- Department of Cell Biology, Ribeirão Preto Medical School University of São Paulo, Ribeirão Preto, Brazil
| | - Lucas A Tavares
- Department of Cell Biology, Ribeirão Preto Medical School University of São Paulo, Ribeirão Preto, Brazil
| | - Ana L R Alves-Rezende
- Division of Dermatology, Internal Medicine Department, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Susanne H Karbach
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University of Mainz, Mainz, Germany; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg, University of Mainz, Mainz, Germany; Center for Cardiology, Cardiology I, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Helder I Nakaya
- Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Department of Clinical and Toxicological Analyses of the School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Cacilda S Souza
- Division of Dermatology, Internal Medicine Department, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Katiuchia U Sales
- Department of Cell Biology, Ribeirão Preto Medical School University of São Paulo, Ribeirão Preto, Brazil
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University of Mainz, Mainz, Germany
| | - José C Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil; Center of Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
47
|
Yu J, Zhao Q, Wang X, Zhou H, Hu J, Gu L, Hu Y, Zeng F, Zhao F, Yue C, Zhou P, Li G, Li Y, Wu W, Zhou Y, Li J. Pathogenesis, multi-omics research, and clinical treatment of psoriasis. J Autoimmun 2022; 133:102916. [PMID: 36209691 DOI: 10.1016/j.jaut.2022.102916] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/07/2022]
Abstract
Psoriasis is a common inflammatory skin disease involving interactions between keratinocytes and immune cells that significantly affects the quality of life. It is characterized by hyperproliferation and abnormal differentiation of keratinocytes and excessive infiltration of immune cells in the dermis and epidermis. The immune mechanism underlying this disease has been elucidated in the past few years. Research shows that psoriasis is regulated by the complex interactions among immune cells, such as keratinocytes, dendritic cells, T lymphocytes, neutrophils, macrophages, natural killer cells, mast cells, and other immune cells. An increasing number of signaling pathways have been found to be involved in the pathogenesis of psoriasis, which has prompted the search for new treatment targets. In the past decades, studies on the pathogenesis of psoriasis have focused on the development of targeted and highly effective therapies. In this review, we have discussed the relationship between various types of immune cells and psoriasis and summarized the major signaling pathways involved in the pathogenesis of psoriasis, including the PI3K/AKT/mTOR, JAK-STAT, JNK, and WNT pathways. In addition, we have discussed the results of the latest omics research on psoriasis and the epigenetics of the disease, which provide insights regarding its pathogenesis and therapeutic prospects; we have also summarized its treatment strategies and observations of clinical trials. In this paper, the various aspects of psoriasis are described in detail, and the limitations of the current treatment methods are emphasized. It is necessary to improve and innovate treatment methods from the molecular level of pathogenesis, and further provide new ideas for the treatment and research of psoriasis.
Collapse
Affiliation(s)
- Jiadong Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Qixiang Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Xiaoyan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Hong Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Jing Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Linna Gu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Yawen Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Fanlian Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Fulei Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Chengcheng Yue
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Pei Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Guolin Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Ya Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Wenling Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Yifan Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
48
|
Wang Q, Shi Q, Lu J, Wang Z, Hou J. Causal relationships between inflammatory factors and multiple myeloma: A bidirectional Mendelian randomization study. Int J Cancer 2022; 151:1750-1759. [PMID: 35841389 DOI: 10.1002/ijc.34214] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/21/2022] [Accepted: 07/05/2022] [Indexed: 11/07/2022]
Abstract
Changes in serum inflammatory factors occur throughout the onset and multiple myeloma (MM) progression, the feedback loops make it harder to distinguish between causes and effects. In the present study, we performed a bidirectional summary-level Mendelian randomization (MR) analysis to elucidate the causal relationships of C-reactive protein (CRP) and inflammatory regulators with MM. Summary-level data of genetic variants associated with inflammation were extracted from two genome-wide association studies (GWASs) on CRP and human cytokines, while data on MM was from large meta-analyses of GWASs among 372 617 UK Biobank participants. The inverse-variance weighted (IVW) method was used as the primary MR analysis and MR-Egger, weighted median, and MR-pleiotropy residual sum and outlier (MR-PRESSO) were used as the sensitivity analyses. Our results suggested that higher levels of monocyte-specific chemokine-3 (IVW estimate odds ratio [ORIVW ] per SD genetic cytokines change: 1.24; 95% confidence interval [CI]: 1.03-1.49; P = .02), vascular endothelial growth factor (1.14, 1.03-1.27; P = .02), interleukin-10 (1.33, 1.01-1.75; P = .04) and interleukin-7 (1.24, 1.03-1.48; P = .02) were associated with increased risk of MM, while lower levels of tumor necrosis factor-β (0.84, 0.74-0.92; P < .001) was strongly associated with an increased risk of MM. And conversely, genetically predicted MM was related to increased levels of interleukin-17 (IVW estimate β: 0.051, 95% CI: 0.018-0.085; P = 2.7 × 10-3 ). Besides, we observed no such significant associations for other inflammatory factors in our study. Overall, our study provides genetic evidence on the relationships of CRP and systemic inflammatory regulators with MM. Targeted interventions of specific inflammatory factors may have implications to alleviate MM cancer risk.
Collapse
Affiliation(s)
- Qiangsheng Wang
- Department of Hematology, Ningbo Hangzhou Bay Hospital, Ningbo, Zhejiang, China
| | - Qiqin Shi
- Department of Ophthalmology, Ningbo Hangzhou Bay Hospital, Ningbo, Zhejiang, China
| | - Jiawen Lu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhenqian Wang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jian Hou
- Department of Hematology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
49
|
Nassar AA, Bakr NM, Elyousefi EHI, Elkholy BM, Fawzy MM. Serum immunoglobulin E and Interleukin-17 levels in patients with chronic plaque psoriasis: A case-control study. J Cosmet Dermatol 2022; 21:6377-6384. [PMID: 35957511 DOI: 10.1111/jocd.15299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 07/18/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Psoriasis is a skin disorder mainly mediated by T helper (Th)-1 and Th-17 cells. Recently, high serum immunoglobulin (Ig)-E levels were detected in psoriatic patients. The etiopathogenesis of IgE overproduction in psoriatic patients is still unknown, but IL-17 has been suggested to be responsible for this abnormality. OBJECTIVE To compare levels of IgE and IL-17 in the sera of patients with chronic plaque psoriasis (CPP) to healthy subjects. METHODS This study included 40 patients with CPP and 40 age- and sex-matched healthy individuals. Serum IgE and IL-17 concentrations were measured using the enzyme-linked immunosorbent assay (ELISA) technique. RESULTS Levels of IgE and IL-17 were significantly higher in the sera of psoriatic patients than in controls (p = 0.001 and 0.024, respectively). Psoriatic patients with abnormally high serum IgE levels had higher serum IL-17 levels than those with normal serum IgE levels, but the difference was statistically insignificant (p = 0.080). Furthermore, no significant correlation was found between serum IgE and IL-17 concentrations in psoriatic patients (p = 0.385). CONCLUSIONS It is possible that IgE and IL-17 interact in psoriasis pathogenesis; however, this was not evident in the current study, possibly due to the small sample size. Therefore, other potential causes of elevated IgE levels in psoriatic patients should be investigated. Moreover, the interaction between IgE and IL-17 should be investigated in patients with other clinical variants of psoriasis.
Collapse
Affiliation(s)
- Amany A Nassar
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Noha M Bakr
- Biochemistry Department, National Research Centre, Giza, Egypt
| | - Enas H I Elyousefi
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Basma M Elkholy
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Manal M Fawzy
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
50
|
Demirel Öğüt N, Koç Yıldırım S, Erbağcı E, Hapa FA. Ixekizumab treatment in patients with moderate-to-severe plaque psoriasis in a real-world clinical setting. J Cosmet Dermatol 2022; 21:6215-6224. [PMID: 35801372 DOI: 10.1111/jocd.15217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND The efficacy and safety reports of ixekizumab for moderate-to-severe plaque psoriasis may vary between clinical trials and real-world studies. AIM To analyze the real-world data of ixekizumab therapy to evaluate its efficacy and safety and highlight the factors influencing the treatment response in the real-world scenario. PATIENTS/METHODS Data of 82 adult patients with moderate-to-severe chronic plaque psoriasis are included in this study. Psoriasis area severity index (PASI) 75/90/100 responses at 4, 16, 24, and 48 weeks were analyzed retrospectively from patient charts by examining demographic and clinical characteristics of the patients, especially their previous biologic experience, obesity, and involvement of hard-to-treat areas. RESULTS PASI75, PASI90, and PASI100 responses were achieved in 92.4%, 86.1%, and 26.6% patients at week 16 and maintained till week 48 in 92.3%, 86.5%, and 17.3% patients. PASI90 responses in obese patients were significantly lower than non-obese patients at week 4 (33.3% vs. 69.6%, p = 0.042), but this difference was minimized by week 16 (82.4% vs. 90%, p = 0.405). PASI90 responses in biologic-naive patients were significantly higher than biologic-experienced patients at week 16 (p = 0.015). Involvement of hard-to-treat areas was negatively associated with PASI90 responses at week 16 (OR: 1591805.842; 95% CI: 1.223-2071404486740.201; p = 0.047). CONCLUSION Ixekizumab provides an effective and safe biologic treatment option to patients with moderate-to-severe plaque psoriasis. Obesity, though it affects the early treatment response (till week 4), does not upset the overall treatment response beyond week 16. Previous biologic exposure and involvement of hard-to-treat areas are important prognostic factors for achieving high PASI responses in psoriatic patients.
Collapse
Affiliation(s)
- Neslihan Demirel Öğüt
- Department of Dermatology and Venereology, Uşak Training and Research Hospital, Uşak, Turkey
| | - Sema Koç Yıldırım
- Department of Dermatology and Venereology, Uşak Training and Research Hospital, Uşak, Turkey
| | - Ece Erbağcı
- Department of Dermatology and Venereology, Uşak Training and Research Hospital, Uşak, Turkey
| | - Fatma Aslı Hapa
- Department of Dermatology and Venereology, İzmir Democracy University, İzmir, Turkey
| |
Collapse
|