151
|
Mahmoud AM. Meta-analysis and GRADE assessment of randomized controlled trials on the efficacy and safety of bimekizumab in psoriatic arthritis patients. Curr Med Res Opin 2023; 39:1031-1043. [PMID: 37345857 DOI: 10.1080/03007995.2023.2228613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023]
Abstract
OBJECTIVE A persistent immune-mediated inflammatory disorder called psoriatic arthritis affects about 25% of persons with psoriasis. Bimekizumab, a humanized monoclonal IgG1 antibody, is a novel therapeutic approach that inhibits homodimers and heterodimers of IL-17A and IL-17F by binding to comparable locations in these molecules. Bimekizumab was the subject of a meta-analysis to assess its efficacy and safety in psoriatic arthritis patients. METHODS All randomized clinical trials were looked up on PubMed, Scopus, and Web of Science. The Systematic Review Accelerator tool was used to screen them, and RevMan was used to analyze them. The Mean Difference (MD) and 95% Confidence Interval (CI) were used to examine continuous data, whereas the Risk Ratio (RR) and 95% CI were used to evaluate dichotomous data. RESULTS A total of 1364 participants from 4 trials were included in this meta-analysis. The number of participants who met the American College of Rheumatology 50 threshold was significantly higher in the bimekizumab group compared to the placebo group [RR = 4.94, 95% CI (3.73, 6.55), p < .00001]. Psoriasis Area and Severity Index 100 was achieved by significantly more people in the bimekizumab group than in the placebo group [RR = 11.45, 95% CI (6.67, 19.67), p < .00001]. There was no significant difference between the bimekizumab group and the placebo group in terms of treatment-emergent adverse events [RR = 1.08, 95% CI (0.97, 1.21), p = .15]. CONCLUSION In comparison to a placebo, bimekizumab treatment significantly improved joint and skin efficacy outcomes. Also, its safety results were acceptable.
Collapse
|
152
|
Wei G, Shi Z, Wu X, Hwang ST. The Emerging Potential of Bile Acids as a Modulator of Psoriatic Inflammation. JOURNAL OF PSORIASIS AND PSORIATIC ARTHRITIS 2023; 8:118-123. [PMID: 39296312 PMCID: PMC11361517 DOI: 10.1177/24755303231177965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Background Bile acids (BAs) are cholesterol-based amphipathic surfactants that are most widely known for their contributions to lipid metabolism, but more recently have been increasingly recognized as a key signaling molecule in inflammatory diseases as well as, potentially, psoriatic disease. Objective This brief review reviews relevant literature in order to briefly describe the synthesis of bile acids and their subsequent metabolism and to analyze recent animal and human data that supports anti-inflammatory activity of some BAs in psoriasiform dermatitis. Methods Pubmed and other public sources were used to survey the literature relevant to the topic of bile acids and their potential use in psoriasis. Conclusion There is clinical and preclinical evidence to support a potential role for BA Supplementation (or modulation BA metabolism and signaling) in the treatment of psoriasis.
Collapse
Affiliation(s)
- Grace Wei
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Zhenrui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuesong Wu
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Samuel T Hwang
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
153
|
Staniszewska M, Kiełbowski K, Rusińska K, Bakinowska E, Gromowska E, Pawlik A. Targeting cyclin-dependent kinases in rheumatoid arthritis and psoriasis - a review of current evidence. Expert Opin Ther Targets 2023; 27:1097-1113. [PMID: 37982244 DOI: 10.1080/14728222.2023.2285784] [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: 08/04/2023] [Accepted: 11/16/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic inflammatory disease associated with synovial proliferation and bone erosion, which leads to the structural and functional impairment of the joints. Immune cells, together with synoviocytes, induce a pro-inflammatory environment and novel treatment agents target inflammatory cytokines. Psoriasis is a chronic immune-mediated skin disease, and several cytokines are considered as typical mediators in the progression of the disease, including IL-23, IL-22, and IL-17, among others. AREA COVERED In this review, we try to evaluate whether cyclin-dependent kinases (CDK), enzymes that regulate cell cycle and transcription of various genes, could become novel therapeutic targets in RA and psoriasis. We present the main results of in vitro and in vivo studies, as well as scarce clinical reports. EXPERT OPINION CDK inhibitors seem promising for treating RA and psoriasis because of their multidirectional effects. CDK inhibitors may affect not only the process of osteoclastogenesis, thereby reducing joint destruction in RA, but also the process of apoptosis of neutrophils and macrophages responsible for the development of inflammation in both RA and psoriasis. However, assessing the efficacy of these drugs in clinical practice requires multi-center, long-term clinical trials evaluating the effectiveness and safety of CDK-blocking therapy in RA and psoriasis.
Collapse
Affiliation(s)
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Klaudia Rusińska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Ewa Gromowska
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
154
|
Rusiñol L, Carmona-Rocha E, Puig L. Psoriasis: a focus on upcoming oral formulations. Expert Opin Investig Drugs 2023; 32:583-600. [PMID: 37507233 DOI: 10.1080/13543784.2023.2242767] [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: 06/11/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 07/30/2023]
Abstract
INTRODUCTION Targeted therapies have greatly improved the quality of life of patients with psoriasis. Despite the extensive list of treatments available, multiple new drugs are being developed, especially oral therapies with potential advantages as regards comfort of administration. However, the efficacy and safety of these new oral therapies need to be improved to match those of novel biologics. AREAS COVERED We provide a narrative review of the oral therapies for psoriasis that are currently under development, from Jak inhibitors to oral IL-17 and IL-23 inhibitors, among others. A literature search was performed for articles published from 1 January 2020, to 6 June 2023. EXPERT OPINION The approval of deucravacitinib, the first Jak inhibitor for the treatment of moderate-to-severe plaque psoriasis, heralds a bright therapeutic future with multiple new oral formulations. A great number of oral treatments with singular mechanism of action, like A3AR agonists, HSP90 inhibitors, ROCK-2 inhibitors, oral TNF inhibitors, oral IL-23 inhibitors, oral IL-17 inhibitors, PD4 inhibitors (orismilast) and several Tyk2 inhibitors, are currently being evaluated in clinical trials and could be suitable for approval in the future. Growing variation in treatment modes of administration will allow dermatologists to better integrate patient preferences in the therapeutic decision process.
Collapse
Affiliation(s)
- Lluís Rusiñol
- Dermatology Department IIB Sant Pau, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - Elena Carmona-Rocha
- Dermatology Department IIB Sant Pau, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - Lluís Puig
- Dermatology Department IIB Sant Pau, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| |
Collapse
|
155
|
Fu D, Zheng S, Li J, Hu H, Wang Q, Fu X, Li M, Yan D, Yang Z, Tian Z, Song X. Anti-interleukin 33 treatment alleviates psoriatic dermatitis in mice induced imiquimod. Int Immunopharmacol 2023; 121:110480. [PMID: 37343370 DOI: 10.1016/j.intimp.2023.110480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/23/2023]
Abstract
Interleukin-33(IL-33), is constitutively expressed in the epithelial cells of the skin. It has been reported that IL-33 contributed to the severity of the disease in psoriasis-like mouse models. In the current study, we evaluated the effect of anti-IL-33 antibody (Ab) in imiquimod-induced psoriatic dermatitis in mice. Our observations showed that anti-IL-33 Ab ameliorated the erythema, scaling, epidermal thickness and spleen index. Additionally, we found anti-IL-33 Ab significantly decreased the expression of psoriasis-related cytokines. Moreover, anti-IL-33 Ab significantly reduced Ki-67 positive cells, CD3+CD4+T cells, and CD3+CD8+T cells in the skin lesions. Furthermore, anti-IL-33 Ab treatment down-regulated the expression of phosphorylation of STAT3 and IL-33 in model mouse. These results indicated that the anti-IL-33 Ab alleviated the seriousness of skin lesions, inhibited the activation of the STAT3, lymphocyte infiltration and the secretion of pro-inflammatory cytokines in imiquimod-induced psoriatic dermatitis in mice, suggesting IL-33 may be a therapeutic target for the treatment of psoriasis.
Collapse
Affiliation(s)
- Dandan Fu
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Shuting Zheng
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Jialin Li
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Hua Hu
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Qingqing Wang
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Xiuyu Fu
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China
| | - Min Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Dong Yan
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Zishan Yang
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453000, Henan, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, China
| | - Zhongwei Tian
- Department of Dermatology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan, China.
| | - Xiangfeng Song
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453000, Henan, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, 453000, Henan, China.
| |
Collapse
|
156
|
Sánchez-Rodríguez G, Puig L. Pathogenic Role of IL-17 and Therapeutic Targeting of IL-17F in Psoriatic Arthritis and Spondyloarthropathies. Int J Mol Sci 2023; 24:10305. [PMID: 37373452 DOI: 10.3390/ijms241210305] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
The interleukin 17 (IL-17) family, a subset of cytokines consisting of IL-17A-F, plays crucial roles in host defence against microbial organisms and the development of inflammatory diseases, including psoriasis (PsO), axial spondyloarthritis (axSpA), and psoriatic arthritis (PsA). IL-17A is the signature cytokine produced by T helper 17 (Th17) cells and is considered the most biologically active form. The pathogenetic involvement of IL-17A in these conditions has been confirmed, and its blockade with biological agents has provided a highly effective therapeutical approach. IL-17F is also overexpressed in the skin and synovial tissues of patients with these diseases, and recent studies suggest its involvement in promoting inflammation and tissue damage in axSpA and PsA. The simultaneous targeting of IL-17A and IL-17F by dual inhibitors and bispecific antibodies may improve the management of Pso, PsA, and axSpA, as demonstrated in the pivotal studies of dual specific antibodies such as bimekizumab. The present review focuses on the role of IL-17F and its therapeutic blockade in axSpA and PsA.
Collapse
Affiliation(s)
- Guillermo Sánchez-Rodríguez
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Carrer de Sant Quintí, 89, 08041 Barcelona, Spain
| | - Lluís Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Carrer de Sant Quintí, 89, 08041 Barcelona, Spain
| |
Collapse
|
157
|
Gyllemark P, Sjöwall J, Forsberg P, Ernerudh J, Henningsson AJ. Intrathecal Th17-driven inflammation is associated with prolonged post-treatment convalescence for patients with Lyme neuroborreliosis. Sci Rep 2023; 13:9722. [PMID: 37322136 PMCID: PMC10272195 DOI: 10.1038/s41598-023-36709-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: 02/27/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023] Open
Abstract
Lyme neuroborreliosis (LNB) is associated with increased levels of pro-inflammatory cytokines and chemokines in the cerebrospinal fluid (CSF). Residual symptoms after antibiotic treatment can have deleterious effects on patients and knowledge regarding the pathogenesis linked to prolonged recovery is lacking. In this prospective follow-up study, we investigated the B cell-associated and T helper (Th) cell-associated immune responses in well-characterized patients with LNB and controls. The aims were to assess the kinetics of selected cytokines and chemokines involved in the inflammatory response and to identify potential prognostic markers. We investigated 13 patients with LNB according to a standardized clinical protocol before antibiotic treatment and after 1, 6 and 12 months of follow-up. CSF and blood samples were obtained at baseline and after 1 month. As controls, we used CSF samples from 37 patients who received spinal anesthesia during orthopedic surgery. The CSF samples were analyzed for CXCL10 (Th1-related), CCL22 (Th2-related) and IL-17A, CXCL1 and CCL20 (Th17-related), as well as for the B cell-related cytokines of a proliferation-inducing ligand (APRIL), B cell-activating factor (BAFF) and CXCL13. The CSF levels of all the cytokines and chemokines, with the exception of APRIL, were significantly higher at baseline in patients with LNB compared with controls. All the cytokines and chemokines, except for IL-17A were significantly reduced at 1-month follow-up. Patients with quick recovery (< 1 month, n = 3) had significantly lower levels of CCL20 at baseline and lower levels of IL-17A at 1-month follow-up. Patients with time of recovery > 6 months (n = 7) had significantly higher levels of IL-17A at the one-month follow-up. No other cytokines or chemokines were associated with prolonged recovery. Dominating residual symptoms were fatigue, myalgia, radiculitis and/or arthralgia. In this prospective follow-up study of patients with LNB, we found significantly lower levels of CCL20 in those who recovered rapidly, and increased levels of IL-17A in patients with delayed recovery post-treatment. Our findings indicate persistent Th17-driven inflammation in the CSF, possibly contributing to a longer convalescence, and suggest IL-17A and CCL20 as potential biomarker candidates for patients with LNB.
Collapse
Affiliation(s)
- Paula Gyllemark
- Department of Infectious Diseases, Region Jönköping County, 551 85, Jönköping, Sweden.
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| | - Johanna Sjöwall
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Infectious Diseases, Region Östergötland County, Linköping/Norrköping, Sweden
| | - Pia Forsberg
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jan Ernerudh
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anna J Henningsson
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- National Reference Laboratory for Borrelia and Other Tick-Borne Bacteria, Division of Clinical Microbiology, Laboratory Medicine, Region Jönköping County, Jönköping, Sweden
| |
Collapse
|
158
|
Frustaci A, Galea N, Dominici L, Verardo R, Alfarano M, Scialla R, Richetta AG. Interleukin-17A-Correlated Myocarditis in Patients with Psoriasis: Cardiac Recovery following Secukinumab Administration. J Clin Med 2023; 12:4010. [PMID: 37373705 PMCID: PMC10299195 DOI: 10.3390/jcm12124010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
(1) Background: Psoriasis (PS) is a common immune-mediated disease of the skin with possible extension to joints, aorta and eye. Myocardial inflammation has rarely been suggested. (2) Aims: Report of PS-related myocarditis. (3) Methods and Results: One hundred consecutive patients with PS were screened for cardiac involvement. Among them, five male patients (aged 56 ± 9.5 years) with a moderate-severe form of PS showed dilated cardiomyopathy (LVEF < 35%) with normal coronary arteries and valves. They underwent a left-ventricular endomyocardial biopsy for evaluation of myocardial substrate. Endomyocardial samples were processed for histology and immunohistochemistry, including myocardial expression of Toll-Like Receptor 4 (TLR4) and interleukin-17A (IL-17A), which play a major role in PS pathogenesis. Real-time PCRs were carried out for cardiotropic viruses, and Western blot analysis was conducted for myocardial expression of IL-17A. Patients' sera were tested for anti-heart autoantibodies. Active lymphocytic myocarditis was revealed in all five patients, characterized by an absence of viral genomes with PCR, positive anti-heart autoantibodies, overexpression of TLR-4 and enhancement of IL-17-A during western blot analysis, showing a 2.48-fold increase in psoriatic myocarditis compared with no psoriatic myocarditis and a six-fold increase compared to myocardial controls. Treatment included combination of prednisone (1 mg/kg daily for 4 weeks, tapered to 0.33 mg/kg) and azathioprine (2 mg/kg, daily) in 3 pts or secukinumab (SK, 150 mg/weekly for 4 weeks followed by 150 mg/monthly) in 2 pts for 6 months. LVEDD and LVEF improved in the first 3 pts (-14% and + 118%, respectively), while they completely recovered (LVEF > 50%) in the last 2 pts on SK. (4) Conclusions: IL-17A-related myocarditis can occur in up to 5% of patients with PS. It manifests as progressive dilated cardiomyopathy. It may completely recover following SK administration.
Collapse
Affiliation(s)
| | - Nicola Galea
- Department of Clinical, Internal, Anesthesiologist and Cardiovascular Sciences, Sapienza University, 00185 Rome, Italy; (N.G.); (L.D.); (M.A.); (A.G.R.)
| | - Lorenzo Dominici
- Department of Clinical, Internal, Anesthesiologist and Cardiovascular Sciences, Sapienza University, 00185 Rome, Italy; (N.G.); (L.D.); (M.A.); (A.G.R.)
| | - Romina Verardo
- Cellular and Molecular Cardiology Lab, IRCCS L. Spallanzani, 00149 Rome, Italy; (R.V.); (R.S.)
| | - Maria Alfarano
- Department of Clinical, Internal, Anesthesiologist and Cardiovascular Sciences, Sapienza University, 00185 Rome, Italy; (N.G.); (L.D.); (M.A.); (A.G.R.)
| | - Rossella Scialla
- Cellular and Molecular Cardiology Lab, IRCCS L. Spallanzani, 00149 Rome, Italy; (R.V.); (R.S.)
| | - Antonio Giovanni Richetta
- Department of Clinical, Internal, Anesthesiologist and Cardiovascular Sciences, Sapienza University, 00185 Rome, Italy; (N.G.); (L.D.); (M.A.); (A.G.R.)
| |
Collapse
|
159
|
Qu Y, Li D, Xiong H, Shi D. Transcriptional regulation on effector T cells in the pathogenesis of psoriasis. Eur J Med Res 2023; 28:182. [PMID: 37270497 PMCID: PMC10239166 DOI: 10.1186/s40001-023-01144-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/15/2023] [Indexed: 06/05/2023] Open
Abstract
Psoriasis is one of the most common inflammatory diseases, characterized by scaly erythematous plaques on the skin. The accumulated evidence on immunopathology of psoriasis suggests that inflammatory reaction is primarily mediated by T helper (Th) cells. The differentiation of Th cells plays important roles in psoriatic progression and it is regulated by transcription factors such as T-bet, GATA3, RORγt, and FOXP3, which can convert naïve CD4+ T cells, respectively, into Th1, Th2, Th17 and Treg subsets. Through the activation of the JAK/STAT and Notch signaling pathways, together with their downstream effector molecules including TNF-α, IFN-γ, IL-17, TGF-β, these subsets of Th cells are then deeply involved in the pathogenesis of psoriasis. As a result, keratinocytes are abnormally proliferated and abundant inflammatory immune cells are infiltrated in psoriatic lesions. We hypothesize that modulation of the expression of transcription factors for each Th subset could be a new therapeutic target for psoriasis. In this review, we will focus on the recent literature concerning the transcriptional regulation of Th cells in psoriasis.
Collapse
Affiliation(s)
- Yuying Qu
- College of Clinical Medicine, Jining Medical University, Jining, 272067, Shandong, China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, USA
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, 272067, Shandong, China.
| | - Dongmei Shi
- Department of Dermatology, Jining No. 1 People's Hospital, Jining, 272067, Shandong, China.
| |
Collapse
|
160
|
Solá P, Mereu E, Bonjoch J, Casado-Peláez M, Prats N, Aguilera M, Reina O, Blanco E, Esteller M, Di Croce L, Heyn H, Solanas G, Benitah SA. Targeting lymphoid-derived IL-17 signaling to delay skin aging. NATURE AGING 2023; 3:688-704. [PMID: 37291218 PMCID: PMC10275755 DOI: 10.1038/s43587-023-00431-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/02/2023] [Indexed: 06/10/2023]
Abstract
Skin aging is characterized by structural and functional changes that contribute to age-associated frailty. This probably depends on synergy between alterations in the local niche and stem cell-intrinsic changes, underscored by proinflammatory microenvironments that drive pleotropic changes. The nature of these age-associated inflammatory cues, or how they affect tissue aging, is unknown. Based on single-cell RNA sequencing of the dermal compartment of mouse skin, we show a skew towards an IL-17-expressing phenotype of T helper cells, γδ T cells and innate lymphoid cells in aged skin. Importantly, in vivo blockade of IL-17 signaling during aging reduces the proinflammatory state of the skin, delaying the appearance of age-related traits. Mechanistically, aberrant IL-17 signals through NF-κB in epidermal cells to impair homeostatic functions while promoting an inflammatory state. Our results indicate that aged skin shows signs of chronic inflammation and that increased IL-17 signaling could be targeted to prevent age-associated skin ailments.
Collapse
Affiliation(s)
- Paloma Solá
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Júlia Bonjoch
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Neus Prats
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mònica Aguilera
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Oscar Reina
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Enrique Blanco
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Manel Esteller
- Josep Carreras Leukemia Research Institute, Badalona, Spain
- ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| | - Luciano Di Croce
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
- ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| | - Holger Heyn
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Guiomar Solanas
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, Barcelona, Spain.
- ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, Spain.
| |
Collapse
|
161
|
Williams KL, Enslow R, Suresh S, Beaton C, Hodge M, Brooks AE. Using the Microbiome as a Regenerative Medicine Strategy for Autoimmune Diseases. Biomedicines 2023; 11:1582. [PMID: 37371676 DOI: 10.3390/biomedicines11061582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Autoimmune (AI) diseases, which present in a multitude of systemic manifestations, have been connected to many underlying factors. These factors include the environment, genetics, individual microbiomes, and diet. An individual's gut microbiota is an integral aspect of human functioning, as it is intimately integrated into the metabolic, mechanical, immunological, and neurologic pathways of the body. The microbiota dynamically changes throughout our lifetimes and is individually unique. While the gut microbiome is ever-adaptive, gut dysbiosis can exert a significant influence on physical and mental health. Gut dysbiosis is a common factor in various AI, and diets with elevated fat and sugar content have been linked to gut microbiome alterations, contributing to increased systemic inflammation. Additionally, multiple AI's have increased levels of certain inflammatory markers such as TNF-a, IL-6, and IL-17 that have been shown to contribute to arthropathy and are also linked to increased levels of gut dysbiosis. While chronic inflammation has been shown to affect many physiologic systems, this review explores the connection between gut microbiota, bone metabolism, and the skeletal and joint destruction associated with various AI, including psoriatic arthritis, systemic lupus erythematosus, irritable bowel disease, and rheumatoid arthritis. This review aims to define the mechanisms of microbiome crosstalk between the cells of bone and cartilage, as well as to investigate the potential bidirectional connections between AI, bony and cartilaginous tissue, and the gut microbiome. By doing this, the review also introduces the concept of altering an individual's specific gut microbiota as a form of regenerative medicine and potential tailored therapy for joint destruction seen in AI. We hope to show multiple, specific ways to target the microbiome through diet changes, rebalancing microbial diversity, or decreasing specific microbes associated with increased gut permeability, leading to reduced systemic inflammation contributing to joint pathology. Additionally, we plan to show that diet alterations can promote beneficial changes in the gut microbiota, supporting the body's own endogenous processes to decrease inflammation and increase healing. This concept of microbial alteration falls under the definition of regenerative medicine and should be included accordingly. By implementing microbial alterations in regenerative medicine, this current study could lend increasing support to the current research on the associations of the gut microbiota, bone metabolism, and AI-related musculoskeletal pathology.
Collapse
Affiliation(s)
- Kaitlin L Williams
- College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80112, USA
| | - Ryan Enslow
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA
| | - Shreyas Suresh
- College of Osteopathic Medicine, Rocky Vista University, Ivins, UT 84738, USA
| | - Camille Beaton
- College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80112, USA
| | - Mitchell Hodge
- College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80112, USA
| | - Amanda E Brooks
- Department of Research and Scholarly Activity, Rocky Vista University, Ivins, UT 84738, USA
- Department of Research and Scholarly Activity, Rocky Vista University, Parker, CO 80112, USA
| |
Collapse
|
162
|
Niu M, Yuan J, Yan M, Yang G, Yan Z, Yang X. Discovery of CLEC2B as a diagnostic biomarker and screening of celastrol as a candidate drug for psoriatic arthritis through bioinformatics analysis. J Orthop Surg Res 2023; 18:390. [PMID: 37246213 DOI: 10.1186/s13018-023-03843-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/08/2023] [Indexed: 05/30/2023] Open
Abstract
BACKGROUND Psoriatic arthritis (PSA) is a chronic, immune-mediated inflammatory joint disease that is liked to mortality due to cardiovascular disease. Diagnostic markers and effective therapeutic options for PSA remain limited due to the lack of understanding of the pathogenesis. We aimed to identify potential diagnostic markers and screen the therapeutic compounds for PSA based on bioinformatics analysis. METHODS Differentially expressed genes (DEGs) of PSA were identified from the GSE61281 dataset. WGCNA was used to identify PSA-related modules and prognostic biomarkers. Clinical samples were collected to validate the expression of the diagnostic gene. These DEGs were subjected to the CMap database for the identification of therapeutic candidates for PSA. Potential pathways and targets for drug candidates to treat PSA were predicted using Network Pharmacology. Molecular docking techniques were used to validate key targets. RESULTS CLEC2B was identified as a diagnostic marker for PSA patients (AUC > 0.8) and was significantly upregulated in blood samples. In addition, celastrol was identified as a candidate drug for PSA. Subsequently, the network pharmacology approach identified four core targets (IL6, TNF, GAPDH, and AKT1) of celastrol and revealed that celastrol could treat PSA by modulating inflammatory-related pathways. Finally, molecular docking demonstrated stable binding of celastrol to four core targets in the treatment of PSA. Animal experiments indicated celastrol alleviated inflammatory response in the mannan-induced PSA. CONCLUSION CLEC2B was a diagnostic marker for PSA patients. Celastrol was identified as a potential therapeutic drug for PSA via regulating immunity and inflammation.
Collapse
Affiliation(s)
- Min Niu
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jingman Yuan
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Meixi Yan
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Ge Yang
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Ziyi Yan
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xichao Yang
- Department of Rheumatology Immunology and Endocrinology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
163
|
Vereertbrugghen A, Pizzano M, Sabbione F, Keitelman IA, Shiromizu CM, Aguilar DV, Fuentes F, de Paiva CS, Giordano M, Trevani A, Galletti JG. An ocular Th1 immune response promotes corneal nerve damage independently of the development of corneal epitheliopathy. J Neuroinflammation 2023; 20:120. [PMID: 37217914 PMCID: PMC10201717 DOI: 10.1186/s12974-023-02800-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023] Open
Abstract
Proper sight is not possible without a smooth, transparent cornea, which is highly exposed to environmental threats. The abundant corneal nerves are interspersed with epithelial cells in the anterior corneal surface and are instrumental to corneal integrity and immunoregulation. Conversely, corneal neuropathy is commonly observed in some immune-mediated corneal disorders but not in others, and its pathogenesis is poorly understood. Here we hypothesized that the type of adaptive immune response may influence the development of corneal neuropathy. To test this, we first immunized OT-II mice with different adjuvants that favor T helper (Th)1 or Th2 responses. Both Th1-skewed mice (measured by interferon-γ production) and Th2-skewed (measured by interleukin-4 production) developed comparable ocular surface inflammation and conjunctival CD4+ T cell recruitment but no appreciable corneal epithelial changes upon repeated local antigenic challenge. Th1-skewed mice showed decreased corneal mechanical sensitivity and altered corneal nerve morphology (signs of corneal neuropathy) upon antigenic challenge. However, Th2-skewed mice also developed milder corneal neuropathy immediately after immunization and independently of ocular challenge, suggestive of adjuvant-induced neurotoxicity. All these findings were confirmed in wild-type mice. To circumvent unwanted neurotoxicity, CD4+ T cells from immunized mice were adoptively transferred to T cell-deficient mice. In this setup, only Th1-transferred mice developed corneal neuropathy upon antigenic challenge. To further delineate the contribution of each profile, CD4+ T cells were polarized in vitro to either Th1, Th2, or Th17 cells and transferred to T cell-deficient mice. Upon local antigenic challenge, all groups had commensurate conjunctival CD4+ T cell recruitment and macroscopic ocular inflammation. However, none of the groups developed corneal epithelial changes and only Th1-transferred mice showed signs of corneal neuropathy. Altogether, the data show that corneal nerves, as opposed to corneal epithelial cells, are sensitive to immune-driven damage mediated by Th1 CD4+ T cells in the absence of other pathogenic factors. These findings have potential therapeutic implications for ocular surface disorders.
Collapse
Affiliation(s)
- Alexia Vereertbrugghen
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Manuela Pizzano
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Florencia Sabbione
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Irene Angelica Keitelman
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Carolina Maiumi Shiromizu
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Douglas Vera Aguilar
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Federico Fuentes
- Confocal Microscopy Unit, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Cintia S de Paiva
- Department of Ophthalmology, Ocular Surface Center, Cullen Eye Institute, Baylor College of Medicine, Houston, TX, USA
| | - Mirta Giordano
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Analía Trevani
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina
| | - Jeremías G Galletti
- Innate Immunity Laboratory, Institute of Experimental Medicine (CONICET/National Academy of Medicine of Buenos Aires), Buenos Aires, Argentina.
| |
Collapse
|
164
|
Wang Z, Hu Y, Wang X, Chen Y, Wu D, Ji H, Yu C, Fang J, Pan C, Wang L, Wang S, Guo Y, Lu Y, Wu D, Ren F, Zhu H, Shi Y. Comparative Analysis of the Therapeutic Effects of Fresh and Cryopreserved Human Umbilical Cord Derived Mesenchymal Stem Cells in the Treatment of Psoriasis. Stem Cell Rev Rep 2023:10.1007/s12015-023-10556-8. [PMID: 37199874 DOI: 10.1007/s12015-023-10556-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2023] [Indexed: 05/19/2023]
Abstract
Psoriasis, an inflammatory autoimmune skin disease, is characterized by scaly white or erythematous plaques, which severely influence patients' quality of life and social activities. Mesenchymal stem cells derived from the human umbilical cord (UCMSCs) represent a promising therapeutic approach for psoriasis because of its unique superiority in ethical agreeableness, abundant source, high proliferation capacity, and immunosuppression. Although cryopreservation provided multiple benefits to the cell therapy, it also greatly compromised clinical benefits of MSCs due to impaired cell functions. The current study aims to evaluate the therapeutic efficacy of cryopreserved UCMSCs in a mouse model of psoriasis as well as in patients with psoriasis. Our results showed that cryopreserved and fresh UCMSCs have comparable effects on the suppression of psoriasis-like symptoms such as thickening, erythema, and scaling, and serum IL-17 A secretion in mice model of psoriasis. Moreover, psoriatic patients injected with cryopreserved UCMSCs had a significant improvement in the Psoriasis Area and Severity Index (PASI), Physician Global Assessment (PGA), and Patient Global Assessments (PtGAs) scores compared to baseline values. Mechanically, cryopreserved UCMSCs markedly inhibit the proliferation of PHA-activated PBMCs, type 1 T helper (Th1) and type 17 T helper (Th17) cell differentiation and secretion of inflammatory cytokines including IFN-γ, TNF-a and IL-17 A in PBMCs stimulated by anti-CD3/CD28 beads. Taken together, these data indicated that cryopreserved UCMSCs exhibited great beneficial effect on psoriasis. Thus, cryopreserved UCMSCs can be systemically administered as ''off-the-shelf'' cell product for psoriasis therapy. Trial Registration ChiCTR1800019509. Registered on November 15, 2018-Retrospectively registered, http://www.chictr.org.cn/ .
Collapse
Affiliation(s)
- Zhifeng Wang
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China.
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China.
| | - Yifan Hu
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, 200443, China
| | - Xiaoyu Wang
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Youdong Chen
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, 200443, China
| | - Danfeng Wu
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Houli Ji
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Cuicui Yu
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Jingmeng Fang
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Chunrong Pan
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Lianjian Wang
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Shouxin Wang
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Yinhong Guo
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Yi Lu
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Di Wu
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Fangfang Ren
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China
| | - Hao Zhu
- Sinoneural Cell Engineering Group Holdings Co., Ltd, No. 1188, Lianhang Road, Shanghai, 201100, China.
| | - Yuling Shi
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China.
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, 200443, China.
| |
Collapse
|
165
|
Xu H, Wang W, Liu X, Huang W, Zhu C, Xu Y, Yang H, Bai J, Geng D. Targeting strategies for bone diseases: signaling pathways and clinical studies. Signal Transduct Target Ther 2023; 8:202. [PMID: 37198232 DOI: 10.1038/s41392-023-01467-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/02/2023] [Accepted: 04/19/2023] [Indexed: 05/19/2023] Open
Abstract
Since the proposal of Paul Ehrlich's magic bullet concept over 100 years ago, tremendous advances have occurred in targeted therapy. From the initial selective antibody, antitoxin to targeted drug delivery that emerged in the past decades, more precise therapeutic efficacy is realized in specific pathological sites of clinical diseases. As a highly pyknotic mineralized tissue with lessened blood flow, bone is characterized by a complex remodeling and homeostatic regulation mechanism, which makes drug therapy for skeletal diseases more challenging than other tissues. Bone-targeted therapy has been considered a promising therapeutic approach for handling such drawbacks. With the deepening understanding of bone biology, improvements in some established bone-targeted drugs and novel therapeutic targets for drugs and deliveries have emerged on the horizon. In this review, we provide a panoramic summary of recent advances in therapeutic strategies based on bone targeting. We highlight targeting strategies based on bone structure and remodeling biology. For bone-targeted therapeutic agents, in addition to improvements of the classic denosumab, romosozumab, and PTH1R ligands, potential regulation of the remodeling process targeting other key membrane expressions, cellular crosstalk, and gene expression, of all bone cells has been exploited. For bone-targeted drug delivery, different delivery strategies targeting bone matrix, bone marrow, and specific bone cells are summarized with a comparison between different targeting ligands. Ultimately, this review will summarize recent advances in the clinical translation of bone-targeted therapies and provide a perspective on the challenges for the application of bone-targeted therapy in the clinic and future trends in this area.
Collapse
Affiliation(s)
- Hao Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wentao Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Xin Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Chen Zhu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, Anhui, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, P. R. China.
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
166
|
Krajewski PK, Szepietowski JC. Secukinumab for the treatment of moderate-to-severe plaque psoriasis in pediatric patients aged six years and older. Expert Rev Clin Immunol 2023:1-6. [PMID: 37191153 DOI: 10.1080/1744666x.2023.2214365] [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: 05/17/2023]
Abstract
INTRODUCTION Psoriasis (PSO) is a chronic, recurrent, inflammatory skin disorder that significantly influences patients' quality of life. The prevalence of PSO among children varies depending on the age and ranges from 0.12% at one year to 2% at 18 years. Due to systemic inflammation, PSO is associated with obesity, hypertension, inflammatory bowel disease, and cardiovascular incidences.Secukinumab is a fully human monoclonal antibody selectively targeting IL-17A. The drug has been approved for children aged 6-18 with moderate to severe plaque psoriasis. Nevertheless, only a few reports thoroughly describe its efficacy and safety in the pediatric population. AREAS COVERED This review aims to describe the efficacy and safety of secukinumab for the treatment of moderate-to-severe plaque PSO in pediatric patients aged six years and older. EXPERT OPINION Although there are only two clinical trials assessing the efficacy and safety of secukinumab in the pediatric population with plaque PSO, this therapy seems to be a significant advancement in managing those patients. Both dose regimens (depending on the weight, low-dose, 75/75/150 mg, and high-dose 75/150/300 mg) of secukinumab were more effective in achieving significant clinical improvement (PASI 75/90 and IGA response) than placebo or active-comparator. Both regimens showed good safety and tolerability profiles, with mostly mild AEs and no SAEs observed.
Collapse
Affiliation(s)
- Piotr K Krajewski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wrocław, Poland
| | - Jacek C Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
167
|
Kavanaugh A, Baraliakos X, Gao S, Chen W, Sweet K, Chakravarty SD, Song Q, Shawi M, Rahman P. Genetic and Molecular Distinctions Between Axial Psoriatic Arthritis and Radiographic Axial Spondyloarthritis: Post Hoc Analyses from Four Phase 3 Clinical Trials. Adv Ther 2023; 40:2439-2456. [PMID: 36995469 PMCID: PMC10129963 DOI: 10.1007/s12325-023-02475-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/20/2023] [Indexed: 03/31/2023]
Abstract
INTRODUCTION Emerging evidence suggests psoriatic arthritis (PsA) with axial involvement (axPsA) and radiographic axial spondyloarthritis (r-axSpA) may possibly represent distinct disorders, with some differing clinical manifestations, genetic associations, and radiographic findings. Moreover, axPsA and r-axSpA may respond differently to therapies: guselkumab (interleukin [IL]-23p19 subunit inhibitor [i]) and ustekinumab (IL-12/23p40i) demonstrated improvements in axial symptoms in patients with PsA; however, neither risankizumab (IL-23p19i) nor ustekinumab demonstrated efficacy versus placebo in patients with r-axSpA. Current analyses aim to further understand potential molecular distinctions between axPsA and r-axSpA and examine the pharmacodynamic effects of guselkumab in patients with axPsA and those with PsA without axial involvement (non-axPsA). METHODS Post hoc analyses utilized biomarker data from blood and serum samples collected from a subset of participants in phase 3 studies of ustekinumab in r-axSpA and guselkumab in PsA (DISCOVER-1 and DISCOVER-2). Participants with axPsA were identified by investigator-verified sacroiliitis (imaging-confirmed) and axial symptoms. HLA mapping, serum cytokine analysis, and whole-blood RNA sequencing were conducted. RESULTS Relative to r-axSpA, patients with axPsA had a lower prevalence of HLA-B27, HLA-C01, and HLA-C02 alleles and a higher prevalence of HLA-B13, HLA-B38, HLA-B57, HLA-C06, and HLA-C12 alleles. Compared with r-axSpA, patients with axPsA had elevated baseline levels of serum IL-17A and IL-17F cytokines, enrichment of IL-17 and IL-10 pathway-associated genes, and neutrophil gene markers. Across axPsA and non-axPsA cohorts, reductions in cytokine levels and normalization of pathway-associated gene expression with guselkumab treatment were comparable. CONCLUSION The differences in HLA genetic associations, serum cytokines, and enrichment scores support the concept that axPsA and r-axSpA may be distinct disorders. The comparable pharmacodynamic effects of guselkumab on cytokine levels and pathway-associated genes observed in patients with axPsA and non-axPsA are consistent with demonstrated clinical improvements across PsA cohorts. These findings contribute to the understanding of potential genetic and molecular distinctions between axPsA and r-axSpA. TRIAL REGISTRATION ClinicalTrials.gov identifiers, NCT03162796, NCT0315828, NCT02437162, and NCT02438787.
Collapse
Affiliation(s)
- Arthur Kavanaugh
- University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | | | - Sheng Gao
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Warner Chen
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Kristen Sweet
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Soumya D Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Qingxuan Song
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - May Shawi
- Immunology Global Medical Affairs, Janssen Pharmaceutical Companies of Johnson & Johnson, Horsham, PA, USA
| | - Proton Rahman
- Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
168
|
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
|
169
|
Monasterio C, Decker A, Schauer F, Büttner N, Schmidt A, Schmitt-Gräff A, Kreisel W. [Esophageal Lichen Planus - An Underdiagnosed Disease]. Laryngorhinootologie 2023; 102:272-281. [PMID: 37040750 DOI: 10.1055/a-1861-7187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
An involvement of the esophagus in patients with lichen planus was described for the first time in 1982. Ever since, it has been seen as a rarity. However, studies over the last 10 years have shown a higher prevalence than expected. It may even be supposed that esophageal lichen planus (ELP) is more common than eosinophilic esophagitis. ELP mostly affects middleaged women. The principal symptom is dysphagia. Endoscopically, ELP is characterized by denudation and tearing of the mucosa, trachealization and hyperkeratosis and esophageal stenosis may occur in patients with long courses of the disease. Histologic findings including mucosal detachment, T-lymphocytic infiltrate, intraepithelial apoptosis (civatte bodies) and dyskeratosis are crucial. Direct immunofluorescence shows fibrinogen deposits along the basement membrane zone. So far, there is no well-established therapy but a treatment with topic steroids is effective in 2/3 of the patients. Common therapy of lichen planus of the skin seems to be ineffective for treatment of ELP. Symptomatic esophageal stenosis should be endoscopically dilated. ELP joins the group of "new" immunologic diseases of the esophagus.
Collapse
Affiliation(s)
- Carmen Monasterio
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, 79106 Freiburg, Germany
| | - Annegrit Decker
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, 79106 Freiburg, Germany
| | - Franziska Schauer
- Klinik für Dermatologie und Venerologie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Nico Büttner
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, 79106 Freiburg, Germany
| | - Arthur Schmidt
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, 79106 Freiburg, Germany
| | - Annette Schmitt-Gräff
- Medizinische Fakultät, Universitätsklinikum Freiburg, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- Institut für Dermatohistologie, Pathologie und Molekularpathologie, Dr. Helmut Laaff, Freiburg, Germany
| | - Wolfgang Kreisel
- Klinik für Innere Medizin II, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
170
|
Yadav K, Singh D, Singh MR, Minz S, Princely Ebenezer Gnanakani S, Sucheta, Yadav R, Vora L, Sahu KK, Bagchi A, Singh Chauhan N, Pradhan M. Preclinical study models of psoriasis: State-of-the-art techniques for testing pharmaceutical products in animal and nonanimal models. Int Immunopharmacol 2023; 117:109945. [PMID: 36871534 DOI: 10.1016/j.intimp.2023.109945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/18/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023]
Abstract
Local and systemic treatments exist for psoriasis, but none can do more than control its symptoms because of its numerous unknown mechanisms. The lack of validated testing models or a defined psoriatic phenotypic profile hinders antipsoriatic drug development. Despite their intricacy, immune-mediated diseases have no improved and precise treatment. The treatment actions may now be predicted for psoriasis and other chronic hyperproliferative skin illnesses using animal models. Their findings confirmed that a psoriasis animal model could mimic a few disease conditions. However, their ethical approval concerns and inability to resemble human psoriasis rightly offer to look for more alternatives. Hence, in this article, we have reported various cutting-edge techniques for the preclinical testing of pharmaceutical products for the treatment of psoriasis.
Collapse
Affiliation(s)
- Krishna Yadav
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492010, India; Raipur Institute of Pharmaceutical Education and Research, Sarona, Raipur, Chhattisgarh 492010, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492010, India
| | - Manju Rawat Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492010, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | | | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | - Renu Yadav
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | - Lalitkumar Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Anindya Bagchi
- Tumor Initiation & Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road La Jolla, CA 92037, USA
| | - Nagendra Singh Chauhan
- Drugs Testing Laboratory Avam Anusandhan Kendra (AYUSH), Government Ayurvedic College, Raipur, India
| | | |
Collapse
|
171
|
Monasterio C, Decker A, Schauer F, Büttner N, Schmidt A, Schmitt-Gräff A, Kreisel W. Der Lichen planus des Ösophagus – Eine unterschätzte Erkrankung. AKTUELLE DERMATOLOGIE 2023. [DOI: 10.1055/a-1753-7120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
ZusammenfassungEine Beteiligung des Ösophagus bei der Hauterkrankung Lichen planus wurde erstmals 1982 beschrieben und fast 30 Jahre lang als eine Rarität angesehen. Untersuchungen der letzten 10 Jahre aber zeigen, dass diese Erkrankung weniger selten ist als angenommen. Es ist sogar anzunehmen, dass der ösophageale Lichen planus (Esophageal Lichen planus, ELP) häufiger ist als die Eosinophile Ösophagitis (EoE). Die Ösophagusbeteiligung betrifft meist Frauen im mittleren Alter. Das Hauptsymptom ist eine Dysphagie. Endoskopisch erkennt man in der Speiseröhre eine charakteristische Schleimhautablösung, eine Trachealisierung, und gelegentlich Hyperkeratosen und bei langem Bestehen auch Stenosen. Wegweisend ist die Histologie mit einer subepithelialen Ablösung sowie einem bandförmigen Infiltrat aus T-Lymphozyten, dem Nachweis von apoptotischen Keratinozyten (Civatte Bodies) und Dyskeratosen. Die direkte Immunfluoreszenz zeigt Fibrinogen-Ablagerungen entlang der Basalmembran. Eine etablierte Therapie gibt es bisher nicht. Die Behandlung mit topischen Steroiden ist in ⅔ der Fälle wirksam. Eine Therapie wie beim klassischen Lichen planus scheint unwirksam zu sein. Bei symptomatischen Stenosen kann eine Dilatation indiziert sein. Der ELP reiht sich in die Gruppe der „neuen“ immunologisch vermittelten Erkrankungen des Ösophagus ein.
Collapse
Affiliation(s)
- Carmen Monasterio
- Klinik für Innere Medizin II, Gastroenterologie, Hepatologie, Endokrinologie und Infektiologie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg
| | - Annegrit Decker
- Klinik für Innere Medizin II, Gastroenterologie, Hepatologie, Endokrinologie und Infektiologie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg
| | - Franziska Schauer
- Klinik für Dermatologie und Venerologie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg
| | - Nico Büttner
- Klinik für Innere Medizin II, Gastroenterologie, Hepatologie, Endokrinologie und Infektiologie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg
| | - Arthur Schmidt
- Klinik für Innere Medizin II, Gastroenterologie, Hepatologie, Endokrinologie und Infektiologie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg
| | - Annette Schmitt-Gräff
- Medizinische Fakultät, Universitätsklinikum Freiburg, Albert-Ludwigs-Universität Freiburg
- Institut für Dermatohistologie, Pathologie und Molekularpathologie, Dr. Helmut Laaff, Freiburg
| | - Wolfgang Kreisel
- Klinik für Innere Medizin II, Gastroenterologie, Hepatologie, Endokrinologie und Infektiologie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg
| |
Collapse
|
172
|
Chen X, Zhang Y, Ding Q, He Y, Li H. Role of IL-17A in different stages of ischemic stroke. Int Immunopharmacol 2023; 117:109926. [PMID: 37012860 DOI: 10.1016/j.intimp.2023.109926] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/01/2023] [Accepted: 02/18/2023] [Indexed: 03/16/2023]
Abstract
Interleukin-17A (IL-17A) plays an important role in the progression of ischemic stroke. IL-17A mediates the endothelial inflammatory response, promotes water and sodium retention, and changes the electrophysiological structure of the atrium, accelerating the progression of ischemic stroke risk factors such as atherosclerotic plaques, hypertension, and atrial fibrillation. In the acute phase of ischemic stroke, IL-17A mediates neuronal injury through neutrophil chemotaxis to the site of injury, the induction of neuronal apoptosis, and activation of the calpain-TRPC-6 (transient receptor potential channel-6) pathway. During ischemic stroke recovery, IL-17A, which is mainly derived from reactive astrocytes, promotes and maintains the survival of neural precursor cells (NPCs) in the subventricular zone (SVZ), neuronal differentiation, and synapse formation and participates in the repair of neurological function. Therapies targeting IL-17A-associated inflammatory signaling pathways can reduce the risk of ischemic stroke and neuronal damage and are a new therapeutic strategy for ischemic stroke and its risk factors. In this paper, we will briefly discuss the pathophysiological role of IL-17A in ischemic stroke risk factors, acute and chronic inflammatory responses, and the potential therapeutic value of targeting IL-17A.
Collapse
Affiliation(s)
- Xiuping Chen
- Department of Rehabilitation, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yi Zhang
- Department of General Medicine, Jiangkou Town Center Hospital, Ganxian 341100, China
| | - Qian Ding
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Disease, Tianjin 300052, China
| | - Yanru He
- Medical Insurance Department, Mingya Insurance Brokers Co., Ltd., Beijing 100020, China
| | - Hui Li
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
173
|
Skougaard M, Ditlev SB, Søndergaard MF, Kristensen LE. Cytokine Signatures in Psoriatic Arthritis Patients Indicate Different Phenotypic Traits Comparing Responders and Non-Responders of IL-17A and TNFα Inhibitors. Int J Mol Sci 2023; 24:ijms24076343. [PMID: 37047315 PMCID: PMC10093817 DOI: 10.3390/ijms24076343] [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: 02/23/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
This study aimed to explore the dynamic interactions between 32 cytokines and biomarkers in Psoriatic Arthritis (PsA) patients to compare cytokine signatures of treatment responders and non-responders. Biomarkers were measured before and after four months of treatment in 39 PsA patients initiating either Tumor Necrosis Factor alpha inhibitor (TNFi) or Interleukin-17A inhibitor (IL-17Ai). Response to treatment was defined by the composite measure, Disease Activity in Psoriatic Arthritis (DAPSA). A two-component principal component analysis (PCA) was implemented to describe cytokine signatures comparing DAPSA50 responders and non-responders. The cytokine signature of TNFi responders was driven by the correlated cytokines interferon γ (IFNγ) and IL-6, additionally associated with IL-12/IL-23p40, TNFα, and CRP, while the cytokine signature of TNFi non-responders was driven by the correlated cytokines IL-15, IL-8, and IFNγ. IL-17Ai responders were characterized by contributions of strongly correlated Th17 inflammatory cytokines, IL-17A, IL-12/IL-23p40, IL-22 to the cytokine signature, whereas IL-17A and IL-12/IL-23p40 did not demonstrate significant contribution in IL-17Ai non-responders. Based on PCA results it was possible to differentiate DAPSA50 responders and non-responders to treatment, endorsing additional examination of cytokine interaction models in PsA patients and supporting further PsA patient immune stratification to improve individualized treatment of PsA patients.
Collapse
Affiliation(s)
- Marie Skougaard
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000 Frederiksberg, Denmark
- Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
- Correspondence:
| | - Sisse Bolm Ditlev
- Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
| | - Magnus Friis Søndergaard
- Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark
| | - Lars Erik Kristensen
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000 Frederiksberg, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| |
Collapse
|
174
|
van der Meer RG, Spoorenberg A, Brouwer E, Doornbos-van der Meer B, Boots AMH, Arends S, Abdulahad WH. Mucosal-associated invariant T cells in patients with axial spondyloarthritis. Front Immunol 2023; 14:1128270. [PMID: 36969157 PMCID: PMC10038212 DOI: 10.3389/fimmu.2023.1128270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
BackgroundSeveral studies implicate Th17-cells and its cytokine (IL-17) in disease pathogenesis of spondyloarthritis (SpA), with available evidence supporting a pathogenic role of CD8+ T-cells. However, data on the involvement of CD8+ mucosal-associated invariant T-cells (MAIT) and their phenotypic characterization and inflammatory function including IL-17 and Granzyme A production in a homogenous population of SpA-patients with primarily axial disease (axSpA) are lacking.ObjectivesQuantify and characterize the phenotype and function of circulating CD8+MAIT-cells in axSpA-patients with primarily axial disease.MethodsBlood samples were obtained from 41 axSpA-patients and 30 age- and sex-matched healthy controls (HC). Numbers and percentages of MAIT-cells (defined as CD3+CD8+CD161highTCRVα7.2+) were determined, and production of IL-17 and Granzyme A (GrzA) by MAIT-cells were examined by flow cytometry upon in vitro stimulation. Serum IgG specific for CMV was measured by ELISA.ResultsNo significant differences in numbers and percentages of circulating MAIT-cells were found between axSpA-patients and HCr zijn meer resultaten de centrale memory CD8 T cellen. cellen van patirculating MAIT cells.. Further phenotypic analysis revealed a significant decrease in numbers of central memory MAIT-cells of axSpA-patients compared to HC. The decrease in central memory MAIT-cells in axSpA patients was not attributed to an alteration in CD8 T-cell numbers, but correlated inversely with serum CMV-IgG titers. Production of IL-17 by MAIT-cells was comparable between axSpA-patients and HC, whereas a significant decrease in the production of GrzA by MAIT-cells from axSpA-patients was observed.ConclusionsThe decrease in cytotoxic capability of circulating MAIT-cells in axSpA-patients might implicate that these cell types migrate to the inflamed tissue and therefore associate with the axial disease pathogenesis.
Collapse
Affiliation(s)
- Rienk Gerben van der Meer
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- *Correspondence: Rienk Gerben van der Meer,
| | - Anneke Spoorenberg
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Berber Doornbos-van der Meer
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Annemieke M. H. Boots
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Suzanne Arends
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Wayel H. Abdulahad
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
175
|
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
|
176
|
Webb C. Extracorporeal photopheresis in conditions of autoimmunity. Transfus Apher Sci 2023; 62:103678. [PMID: 36870909 DOI: 10.1016/j.transci.2023.103678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Affiliation(s)
- Christopher Webb
- Pathology (Transfusion Medicine), Associate Medical Director, Apheresis, UT Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
177
|
Parab S, Doshi G. The Experimental Animal Models in Psoriasis Research: A Comprehensive Review. Int Immunopharmacol 2023; 117:109897. [PMID: 36822099 DOI: 10.1016/j.intimp.2023.109897] [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: 12/08/2022] [Revised: 01/31/2023] [Accepted: 02/11/2023] [Indexed: 02/23/2023]
Abstract
Psoriasis is an autoimmune, chronic, inflammatory skin condition mediated by T cells. It differs from other inflammatory conditions by causing significant alterations in epidermal cell proliferation and differentiation that are both complicated and prominent. The lack of an appropriate animal model has significantly hindered studies into the pathogenic mechanisms of psoriasis since animals other than humans typically do not exhibit the complex phenotypic features of human psoriasis. A variety of methods, including spontaneous mutations, drug-induced mutations, genetically engineered animals, xenotransplantation models, and immunological reconstitution approaches, have all been employed to study specific characteristics in the pathogenesis of psoriasis. Although some of these approaches have been used for more than 50 years and far more models have been introduced recently, they have surprisingly not yet undergone detailed validation. Despite their limitations, these models have shown a connection between keratinocyte hyperplasia, vascular hyperplasia, and a cell-mediated immune response in the skin. The xenotransplantation of diseased or unaffected human skin onto immune-compromised recipients has also significantly aided psoriasis research. This technique has been used in a variety of ways to investigate the function of T lymphocytes and other cells, including preclinical therapeutic studies. The design of pertinent in vivo and in vitro psoriasis models is currently of utmost concern and a crucial step toward its cure. This article outlines the general approach in the development of psoriasis-related animal models, aspects of some specific models, along with their strengths and limitations.
Collapse
Affiliation(s)
- Siddhi Parab
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India
| | - Gaurav Doshi
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India.
| |
Collapse
|
178
|
Brunner SM, Ramspacher A, Rieser C, Leitner J, Heil H, Ablinger M, Tevini J, Wimmer M, Koller A, Piñón Hofbauer J, Felder TK, Bauer JW, Kofler B, Lang R, Wally V. Topical Diacerein Decreases Skin and Splenic CD11c + Dendritic Cells in Psoriasis. Int J Mol Sci 2023; 24:ijms24054324. [PMID: 36901755 PMCID: PMC10001455 DOI: 10.3390/ijms24054324] [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: 12/11/2022] [Revised: 02/08/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Psoriasis is an inflammatory skin disease characterized by increased neo-vascularization, keratinocyte hyperproliferation, a pro-inflammatory cytokine milieu and immune cell infiltration. Diacerein is an anti-inflammatory drug, modulating immune cell functions, including expression and production of cytokines, in different inflammatory conditions. Therefore, we hypothesized that topical diacerein has beneficial effects on the course of psoriasis. The current study aimed to evaluate the effect of topical diacerein on imiquimod (IMQ)-induced psoriasis in C57BL/6 mice. Topical diacerein was observed to be safe without any adverse side effects in healthy or psoriatic animals. Our results demonstrated that diacerein significantly alleviated the psoriasiform-like skin inflammation over a 7-day period. Furthermore, diacerein significantly diminished the psoriasis-associated splenomegaly, indicating a systemic effect of the drug. Remarkably, we observed significantly reduced infiltration of CD11c+ dendritic cells (DCs) into the skin and spleen of psoriatic mice with diacerein treatment. As CD11c+ DCs play a pivotal role in psoriasis pathology, we consider diacerein to be a promising novel therapeutic candidate for psoriasis.
Collapse
Affiliation(s)
- Susanne M. Brunner
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
- Correspondence: ; Tel.: +43-5-7255-57283
| | - Andrea Ramspacher
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Caroline Rieser
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Julia Leitner
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Hannah Heil
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Michael Ablinger
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Julia Tevini
- Department of Laboratory Medicine, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Monika Wimmer
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Andreas Koller
- Research Program for Experimental Dermatology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Josefina Piñón Hofbauer
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Thomas K. Felder
- Department of Laboratory Medicine, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Johann W. Bauer
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Roland Lang
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | - Verena Wally
- EB House Austria, Research Program for Molecular Therapy of Genodermatoses, Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| |
Collapse
|
179
|
Narbutt J, Niedźwiedź M, Lesiak A, Ceryn J, Skibińska M. Secukinumab for the Treatment of Psoriasis in Pediatrics: Patient Selection and Acceptability. Patient Prefer Adherence 2023; 17:421-431. [PMID: 36815128 PMCID: PMC9940655 DOI: 10.2147/ppa.s350753] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Psoriasis (PsO) is a chronic, systemic, immune-mediated inflammatory skin disease affecting 1% to 5% population worldwide. In one-third of patients, the first symptoms of PsO manifest in childhood, with a mean age of nine years. Psoriasis in children under 16 years of age constitutes 4% of dermatological problems in this age group. Chronic inflammation of the skin observed in PsO is associated with a development of potentially serious comorbidities, including psoriatic arthritis, hypertension, metabolic syndrome, cardiovascular diseases, inflammatory bowel disease, depression and anxiety. It is reported that among children with psoriasis between 5 and 16 years of age health-related quality of life is reduced by 30.5%. Early diagnosis and effective treatment are crucial in pediatric psoriatic patients to avoid future complications and stigmatization. Treatment for psoriasis consists of a range of topical medications, phototherapy and non-biologic and biologic systemic therapies. Approved biologics for PsO in pediatric patients include etanercept, adalimumab, ustekinumab, ixekizumab and secukinumab. Secukinumab, a recombinant, fully human monoclonal antibody targeting IL-17A, was approved by the EMA (2020) and FDA (2021) in pediatric patients above 6 years of age for the treatment of moderate to severe plaque psoriasis who are candidates for systemic therapy. This review discusses the selection and acceptability of secukinumab in children with psoriasis.
Collapse
Affiliation(s)
- Joanna Narbutt
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, Lodz, Poland
| | - Michał Niedźwiedź
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, Lodz, Poland
| | - Aleksandra Lesiak
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, Lodz, Poland
| | - Justyna Ceryn
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, Lodz, Poland
- International Doctoral School of the Medical University of Lodz, Lodz, Poland
| | - Małgorzata Skibińska
- Department of Dermatology, Pediatric Dermatology and Dermatological Oncology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
180
|
Li M, Lay K, Zimmer A, Technau-Hafsi K, Wong J, Reimer-Taschenbrecker A, Rohr J, Abdalla E, Fischer J, Reversade B, Has C. A homozygous p.Leu813Pro gain-of-function NLRP1 variant causes phenotypes of different severity in two siblings. Br J Dermatol 2023; 188:259-267. [PMID: 36763876 DOI: 10.1093/bjd/ljac039] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/03/2022] [Accepted: 09/24/2022] [Indexed: 01/22/2023]
Abstract
BACKGROUND A trio exome sequencing study identified a previously unreported NLRP1 gene variant resulting in a p.Leu813Pro substitution of the LRR (leucine-rich repeats) domain of the NLRP1 protein (NACHT, LRR and PYD domains-containing protein 1). This homozygous mutation was shared by two sisters with different clinical presentation: the younger sister had generalized inflammatory nodules with keratotic plugs, clinically resembling multiple keratoacanthomas, while the older had manifestations of familial keratosis lichenoides chronica. OBJECTIVES To analyse the consequences of this NLRP1 variant in two siblings with a different clinical spectrum of severity. METHODS To demonstrate the pathogenicity, p.Leu813Pro was recombinantly expressed, and its effect on inflammasome assembly was assessed. Exome sequencing and RNA-Seq were performed to identify factors with potentially modifying effects on the severity of the skin manifestation between each sibling. RESULTS The variant p.Leu813Pro triggered activation of the NLRP1 inflammasome leading to ASC (apoptosis-associated speck-like protein containing a CARD) speck formation and interleukin (IL)-1β release. The more severely affected sister had several additional genomic variants associated with atopy and psoriasis that were not present in her sibling. IL-5 and IL-17 emerged as dominant cytokines driving prominent inflammation in the skin of the severely affected sibling. CONCLUSIONS To the best of our knowledge, this is the first report of a NLRP1 variant that leads to a different clinical spectrum of severity within the same sibship. IL-5 and IL-17 were the main cytokines expressed in the inflammatory lesions of the severely affected patient and might be regarded as disease modifying factors, and therefore may be considered as therapeutic targets.
Collapse
Affiliation(s)
| | - Kenneth Lay
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | | | | | - Jasmine Wong
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | | | - Jan Rohr
- Department of Paediatrics, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Ebtesam Abdalla
- Department of Human Genetics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | - Bruno Reversade
- Laboratory of Human Genetics & Therapeutics, Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | | |
Collapse
|
181
|
Singh Gautam A, Kumar Singh R. Therapeutic potential of targeting IL-17 and its receptor signaling in neuroinflammation. Drug Discov Today 2023; 28:103517. [PMID: 36736763 DOI: 10.1016/j.drudis.2023.103517] [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: 08/27/2022] [Revised: 12/26/2022] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
T helper 17 cells are thought to significantly contribute to the neuroinflammation process during neurogenerative diseases via their signature cytokine, interleukin (IL)-17. Recently, an emerging key role of IL-17 and its receptors has been documented in inflammatory and autoimmune diseases. The clinical studies conducted on patients with neurodegenerative disease have also shown an increase in IL-17 levels in serum as well as cerebrospinal fluid samples. Therapeutic targeting of either IL-17 receptors or direct IL-17 neutralizing antibodies has shown a promising preclinical and clinical proof of concept for treating chronic autoimmune neurodegenerative diseases such as multiple sclerosis. Thus, IL-17 and its receptors have a central role in regulation of neuroinflammation and can be considered as one of the major therapeutic targets in chronic neuroinflammatory diseases.
Collapse
Affiliation(s)
- Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Transit Campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Transit Campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India.
| |
Collapse
|
182
|
Phytocannabinoids in the Pharmacotherapy of Psoriasis. Molecules 2023; 28:molecules28031192. [PMID: 36770858 PMCID: PMC9920113 DOI: 10.3390/molecules28031192] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
Phytocannabinoids are naturally occurring compounds, the main source of which is Cannabis sativa L. Through direct action or interaction with G protein-coupled receptors, they affect ROS and pro-inflammatory cytokines levels and modify the effectiveness of transcription factor responsible for the biosynthesis of antioxidants which lead to oxidative stress and its consequences. Due to the modification of the redox balance and inflammation, phytocannabinoids are used in the treatment of various diseases, including autoimmune dermatoses, such as atopic dermatitis and psoriasis. Psoriasis is one of the most common dermatoses, and one of unknown etiology. A disturbed redox balance with a shift towards the oxidation leads to oxidative stress, resulting in oxidative modifications, mainly of lipids and proteins, and prolonged activation of immune cells and increased generation of pro-inflammatory cytokines, resulting in chronic inflammation. Given the biological activity of phytocannabinoids, they have become the focus of research as components of pharmacotherapy for psoriasis. Beneficial effects were shown by various representatives of phytocannabinoids, but the effect of cannabidiol (CBD) on skin cells (in vitro and ex vivo) and on blood cells from patients with psoriasis vulgaris and psoriatic arthritis has been most often evaluated in recent years.
Collapse
|
183
|
Zhu W, Lévy-Leduc C, Ternès N. Identification of prognostic and predictive biomarkers in high-dimensional data with PPLasso. BMC Bioinformatics 2023; 24:25. [PMID: 36690931 PMCID: PMC9869528 DOI: 10.1186/s12859-023-05143-0] [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: 07/30/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
In clinical trials, identification of prognostic and predictive biomarkers has became essential to precision medicine. Prognostic biomarkers can be useful for the prevention of the occurrence of the disease, and predictive biomarkers can be used to identify patients with potential benefit from the treatment. Previous researches were mainly focused on clinical characteristics, and the use of genomic data in such an area is hardly studied. A new method is required to simultaneously select prognostic and predictive biomarkers in high dimensional genomic data where biomarkers are highly correlated. We propose a novel approach called PPLasso, that integrates prognostic and predictive effects into one statistical model. PPLasso also takes into account the correlations between biomarkers that can alter the biomarker selection accuracy. Our method consists in transforming the design matrix to remove the correlations between the biomarkers before applying the generalized Lasso. In a comprehensive numerical evaluation, we show that PPLasso outperforms the traditional Lasso and other extensions on both prognostic and predictive biomarker identification in various scenarios. Finally, our method is applied to publicly available transcriptomic and proteomic data.
Collapse
Affiliation(s)
- Wencan Zhu
- Université Paris-Saclay, AgroParisTech, INRAE, UMR MIA Paris-Saclay, 91120, Palaiseau, France.
- Biostatistics and Programming Department, Sanofi R&D, 91380, Chilly Mazarin, France.
| | - Céline Lévy-Leduc
- Université Paris-Saclay, AgroParisTech, INRAE, UMR MIA Paris-Saclay, 91120, Palaiseau, France
| | - Nils Ternès
- Biostatistics and Programming Department, Sanofi R&D, 91380, Chilly Mazarin, France
| |
Collapse
|
184
|
Poryadin GV, Zakhvatov AN, Parshina AY. Pathogenetic aspects of the development of psoriatic arthritis in people with generalized chronic periodontitis. BULLETIN OF SIBERIAN MEDICINE 2023. [DOI: 10.20538/1682-0363-2022-4-183-192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The pathogenetic mechanisms of progression of chronic periodontitis and psoriatic arthritis have common components in immune and inflammatory responses.The pathogenesis of chronic periodontitis involves interaction of microbial and immunological components. As a chronic immune-mediated inflammatory disease and a consequence of an infectious trigger that originally affects gingival soft tissue, periodontitis is typically characterized by periodontal destruction and damage to adjacent connective tissues. Neutrophils contribute to the development of periodontitis and participate in its progression by recruiting T helper 17 cells and stimulating synthesis of the receptor activator of the nuclear factor kappa-β ligand (RANKL), contributing to bone resorption.Macrophages as producers of proinflammatory cytokines (interleukin (IL)-1β, IL-6, IL-22, IL-23, tumor necrosis factor (TNF)), free radicals, and matrix metalloproteinases contribute to the chronic course of the disease. Tissue destruction results in generation of reactive oxygen species by neutrophils, which, against the background of a decrease in the antioxidant potential, leads to development of oxidative stress. These processes together lead to tooth mobility, formation of periodontal pockets, and bone resorption.The key factors in the formation of psoriatic arthritis against the background of periodontitis are overproduction of proinflammatory cytokines in target tissues (skin, joints, gingival microflora) and development of an excessive systemic immune response to the microbiota inhabiting the epithelial and periodontal tissues. A statistically confirmed correlation of the progression of periodontal destruction with the presence of psoriatic arthritis proves the significance of the effects of inflammation as a background for the progression of a comorbidity. Increased IL-17 synthesis plays a crucial role in the development of immune responses of pathological bone remodeling and bone resorption in periodontitis and psoriatic arthritis.
Collapse
Affiliation(s)
| | - A. N. Zakhvatov
- Medical institute, National Research Ogarev Mordovia State University
| | - A. Yu. Parshina
- Medical institute, National Research Ogarev Mordovia State University
| |
Collapse
|
185
|
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
|
186
|
Asahina A, Okubo Y, Morita A, Tada Y, Igarashi A, Langley RG, Deherder D, Matano M, Vanvoorden V, Wang M, Ohtsuki M, Nakagawa H. Bimekizumab Efficacy and Safety in Japanese Patients with Plaque Psoriasis in BE VIVID: A Phase 3, Ustekinumab and Placebo-Controlled Study. Dermatol Ther (Heidelb) 2023; 13:751-768. [PMID: 36648594 PMCID: PMC9984664 DOI: 10.1007/s13555-022-00883-y] [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: 09/29/2022] [Accepted: 12/21/2022] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION Bimekizumab treatment resulted in improved clinical outcomes in patients with moderate-to-severe plaque psoriasis in BE VIVID, a 52-week, phase 3, randomized, ustekinumab and placebo-controlled study. We present data from the BE VIVID Japan patient subpopulation. METHODS Globally, patients were randomized to receive bimekizumab 320 mg every 4 weeks (Q4W), ustekinumab (45/90 mg weight-based at baseline and week 4, then every 12 weeks), or placebo (Q4W through week 16, then bimekizumab 320 mg Q4W). Efficacy endpoints included week 16 Psoriasis Area and Severity Index (PASI) 90 and Investigator's Global Assessment (IGA) 0/1, and other outcomes [PASI 100, PASI 75, IGA 0, Dermatology Life Quality Index (DLQI) 0/1, absolute PASI, scalp IGA, Psoriasis Symptoms and Impacts Measure (P-SIM) responses]. Safety analyses were conducted. RESULTS There were 108 Japanese randomized patients (bimekizumab: 62; ustekinumab: 29; placebo: 17). At week 16, bimekizumab-treated patients had a higher clinical response versus ustekinumab and placebo (PASI 90: 85.5% versus 51.7% and 5.9%; IGA 0/1: 82.3% versus 48.3% and 0.0%). Over 52 weeks, improved clinical response was maintained with bimekizumab, including patients switching from placebo at week 16. Overall, the safety profile in Japanese patients was consistent with that observed in the global population. CONCLUSION Bimekizumab resulted in improved clinical response versus ustekinumab and placebo, and was well-tolerated in Japanese patients. TRIAL REGISTRATION NCT03370133.
Collapse
Affiliation(s)
- Akihiko Asahina
- grid.411898.d0000 0001 0661 2073Department of Dermatology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yukari Okubo
- grid.410793.80000 0001 0663 3325Department of Dermatology, Tokyo Medical University, Tokyo, Japan
| | - Akimichi Morita
- grid.260433.00000 0001 0728 1069Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yayoi Tada
- grid.264706.10000 0000 9239 9995Department of Dermatology, Teikyo University School of Medicine, Tokyo, Japan
| | - Atsuyuki Igarashi
- grid.414992.3Department of Dermatology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Richard G. Langley
- grid.55602.340000 0004 1936 8200Division of Clinical Dermatology and Cutaneous Science, Department of Medicine, Dalhousie University, Halifax, NS Canada
| | - Delphine Deherder
- grid.421932.f0000 0004 0605 7243UCB Pharma, Braine L’alleud, Belgium
| | - Mizuho Matano
- UCB Pharma, UCB Japan Co., Ltd, 8-17-1 Nishi-Shinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.
| | - Veerle Vanvoorden
- grid.421932.f0000 0004 0605 7243UCB Pharma, Braine L’alleud, Belgium
| | | | - Mamitaro Ohtsuki
- grid.410804.90000000123090000Department of Dermatology, Jichi Medical University, Tochigi, Japan
| | - Hidemi Nakagawa
- grid.411898.d0000 0001 0661 2073Department of Dermatology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
187
|
Gangadaran P, Madhyastha H, Madhyastha R, Rajendran RL, Nakajima Y, Watanabe N, Velikkakath AKG, Hong CM, Gopi RV, Muthukalianan GK, Valsala Gopalakrishnan A, Jeyaraman M, Ahn BC. The emerging role of exosomes in innate immunity, diagnosis and therapy. Front Immunol 2023; 13:1085057. [PMID: 36726968 PMCID: PMC9885214 DOI: 10.3389/fimmu.2022.1085057] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/16/2022] [Indexed: 01/17/2023] Open
Abstract
Exosomes, which are nano-sized transport bio-vehicles, play a pivotal role in maintaining homeostasis by exchanging genetic or metabolic information between different cells. Exosomes can also play a vital role in transferring virulent factors between the host and parasite, thereby regulating host gene expression and the immune interphase. The association of inflammation with disease development and the potential of exosomes to enhance or mitigate inflammatory pathways support the notion that exosomes have the potential to alter the course of a disease. Clinical trials exploring the role of exosomes in cancer, osteoporosis, and renal, neurological, and pulmonary disorders are currently underway. Notably, the information available on the signatory efficacy of exosomes in immune-related disorders remains elusive and sporadic. In this review, we discuss immune cell-derived exosomes and their application in immunotherapy, including those against autoimmune connective tissue diseases. Further, we have elucidated our views on the major issues in immune-related pathophysiological processes. Therefore, the information presented in this review highlights the role of exosomes as promising strategies and clinical tools for immune regulation.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Radha Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Yuichi Nakajima
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Nozomi Watanabe
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Anoop Kumar G. Velikkakath
- Center for System Biology and Molecular Medicine, Yenepoya Research center, Yenepoya (Deemed to be University), Mangaluru, Karnataka, India
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Rahul Velikkakath Gopi
- Department of Tissue Engineering and Regeneration Technologies, Sree Chitra Thirunal Institute of Medical Sciences and Technology, Thiruvananthapuram, India
| | | | | | - Madhan Jeyaraman
- Department of Orthopaedics, Faculty of Medicine, Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu, India
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
188
|
Zhou AY, Taylor BE, Barber KG, Lee CA, Taylor ZRR, Howell SJ, Taylor PR. Anti-IL17A Halts the Onset of Diabetic Retinopathy in Type I and II Diabetic Mice. Int J Mol Sci 2023; 24:ijms24021347. [PMID: 36674854 PMCID: PMC9860974 DOI: 10.3390/ijms24021347] [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: 12/09/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
There are ~463 million diabetics worldwide, and more than half have diabetic retinopathy. Yet, treatments are still lacking for non-proliferative diabetic retinopathy. We and others previously provided evidence that Interleukin-17A (IL-17A) plays a pivotal role in non-proliferative diabetic retinopathy. However, all murine studies used Type I diabetes models. Hence, it was the aim of this study to determine if IL-17A induces non-proliferative diabetic retinopathy in Type II diabetic mice, as identified for Type I diabetes. While examining the efficacy of anti-IL-17A as a potential therapeutic in a short-term Type I and a long-term Type II diabetes model; using different routes of administration of anti-IL-17A treatments. Retinal inflammation was significantly decreased (p < 0.05) after Type I-diabetic mice received 1 intravitreal injection, and Type II-diabetic mice received seven intraperitoneal injections of anti-IL-17A. Further, vascular tight junction protein Zonula Occludens-1 (ZO-1) was significantly decreased in both Type I and II diabetic mice, which was significantly increased when mice received anti-IL-17A injections (p < 0.05). Similarly, tight junction protein Occludin degradation was halted in Type II diabetic mice that received anti-IL-17A treatments. Finally, retinal capillary degeneration was halted 6 months after diabetes was confirmed in Type II-diabetic mice that received weekly intraperitoneal injections of anti-IL-17A. These findings provide evidence that IL-17A plays a pivotal role in non-proliferative diabetic retinopathy in Type II diabetic mice, and suggests that anti-IL-17A could be a good therapeutic candidate for non-proliferative diabetic retinopathy.
Collapse
Affiliation(s)
- Amy Y. Zhou
- Department of Ophthalmology and Visual Science, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brooklyn E. Taylor
- Department of Ophthalmology and Visual Science, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | - Chieh A. Lee
- Department of Ophthalmology and Visual Science, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Zakary R. R. Taylor
- Department of Ophthalmology and Visual Science, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Scott J. Howell
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Patricia R. Taylor
- Department of Ophthalmology and Visual Science, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
- Correspondence:
| |
Collapse
|
189
|
Laurindo LF, de Carvalho GM, de Oliveira Zanuso B, Figueira ME, Direito R, de Alvares Goulart R, Buglio DS, Barbalho SM. Curcumin-Based Nanomedicines in the Treatment of Inflammatory and Immunomodulated Diseases: An Evidence-Based Comprehensive Review. Pharmaceutics 2023; 15:pharmaceutics15010229. [PMID: 36678859 PMCID: PMC9861982 DOI: 10.3390/pharmaceutics15010229] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Curcumin (CUR) is a polyphenol extracted from the rhizome of Curcuma longa that possesses potent anti-inflammatory and antioxidant potential. Despite CUR's numerous beneficial effects on human health, it has limitations, such as poor absorption. Nano-based drug delivery systems have recently been applied to improve CUR's solubility and bioavailability and potentialize its health effects. This review investigated the effects of different CUR-based nanomedicines on inflammatory and immunomodulated diseases. PUBMED, EMBASE, COCHRANE, and GOOGLE SCHOLAR databases were searched, and the Scale for Assessment of Narrative Review Articles (SANRA) was used for quality assessment and PRISMA guidelines. Overall, 66 studies were included comprising atherosclerosis, rheumatoid arthritis (RA), Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), Huntington's disease (HD), inflammatory bowel diseases (IBD), psoriasis, liver fibrosis, epilepsy, and COVID-19. The available scientific studies show that there are many known nanoformulations with curcumin. They can be found in nanosuspensions, nanoparticles, nanoemulsions, solid lipid particles, nanocapsules, nanospheres, and liposomes. These formulations can improve CUR bioavailability and can effectively be used as adjuvants in several inflammatory and immune-mediated diseases such as atheroma plaque formation, RA, dementia, AD, PD, MS, IBD, psoriasis, epilepsy, COVID-19, and can be used as potent anti-fibrotic adjuvants in fibrotic liver disease.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Gabriel Magno de Carvalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Bárbara de Oliveira Zanuso
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Maria Eduardo Figueira
- Laboratory of Systems Integration Pharmacology, Clinical & Regulatory Science, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical & Regulatory Science, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Ricardo de Alvares Goulart
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Daiene Santos Buglio
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília, São Paulo 17500-000, Brazil
- Correspondence: ; Tel.: +55-14-99655-3190
| |
Collapse
|
190
|
Psoriasis improvements and inflammatory biomarker normalization with secukinumab: the randomized ObePso-S study. J Dermatol Sci 2023; 109:12-21. [PMID: 36690571 DOI: 10.1016/j.jdermsci.2023.01.003] [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: 07/11/2022] [Revised: 12/07/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
BACKGROUND The IL-17A inhibitor secukinumab has demonstrated consistent efficacy and safety in patients with moderate-to-severe plaque psoriasis, with normalization of molecular and histopathologic psoriasis markers. OBJECTIVE To investigate treatment effects of secukinumab on clinical signs and psoriatic inflammation markers over 52 weeks in patients with psoriasis. METHODS In the ObePso-S study (NCT03055494), patients with psoriasis were randomized 2:1 to receive secukinumab 300 mg (n = 54) or placebo (n = 28), stratified by body weight (<90 or ≥90 kg), for 52 weeks. At Week 12, patients receiving placebo were switched to secukinumab. Psoriasis Area and Severity Index improvement of 90% (PASI90) and Investigator's Global Assessment modified 2011 0/1 responses were assessed at Weeks 12 and 52. Immunohistochemistry for keratin 16 (K16) and gene expression profiles were evaluated in lesional and non-lesional skin biopsies collected at baseline, Week 12, and Week 52. RESULTS Of patients receiving secukinumab, 55.8% and 59.6% achieved PASI90 at Weeks 12 and 52, respectively. K16 was absent in 93.1% of Week 12 PASI90 responders and 93.6% of Week 52 PASI90 responders, which mirrored the down-regulated expression of psoriatic inflammation. Week 52 PASI90 non-responders experienced regression of clinical and inflammatory marker responses toward baseline levels. Lower control of inflammatory gene expression at Week 12 was associated with suboptimal clinical responses at Week 52. CONCLUSION Sustained clinical responses with secukinumab were associated with rapid and sustained normalization of K16 and inflammatory gene expression in most patients. Molecular anti-inflammatory effects of secukinumab at Week 12 were associated with clinical responses at Week 52.
Collapse
|
191
|
Merola JF, Landewé R, McInnes IB, Mease PJ, Ritchlin CT, Tanaka Y, Asahina A, Behrens F, Gladman DD, Gossec L, Gottlieb AB, Thaçi D, Warren RB, Ink B, Assudani D, Bajracharya R, Shende V, Coarse J, Coates LC. Bimekizumab in patients with active psoriatic arthritis and previous inadequate response or intolerance to tumour necrosis factor-α inhibitors: a randomised, double-blind, placebo-controlled, phase 3 trial (BE COMPLETE). Lancet 2023; 401:38-48. [PMID: 36495881 DOI: 10.1016/s0140-6736(22)02303-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/21/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Bimekizumab is a monoclonal IgG1 antibody that selectively inhibits interleukin (IL)-17F and IL-17A. This study compared the efficacy and safety of bimekizumab with placebo over 16 weeks in patients with active psoriatic arthritis and previous inadequate response or intolerance to tumour necrosis factor-α (TNFα) inhibitors. METHODS BE COMPLETE was a phase 3, multicentre, randomised, double-blind, placebo-controlled trial conducted across 92 sites (including hospitals, clinics, and research centres) in 11 countries (Australia, Canada, Czech Republic, Germany, Hungary, Italy, Japan, Poland, Russia, the UK, and the USA). Eligible patients were aged 18 years or older with adult-onset psoriatic arthritis (meeting the Classification Criteria for Psoriatic Arthritis for at least 6 months before screening) with a history of inadequate response or intolerance to treatment with one or two TNFα inhibitors for either psoriatic arthritis or psoriasis. We stratified patients with active psoriatic arthritis by region and previous TNFα inhibitor use. Patients were randomly assigned (2:1) to receive subcutaneous bimekizumab 160 mg every 4 weeks or placebo by an interactive-voice and web-response system on the basis of a predetermined randomisation schedule. The primary endpoint was the proportion of patients with 50% or greater improvement in American College of Rheumatology criteria (ACR50) at week 16 (non-responder imputation). Efficacy analyses were done in the randomised population. The safety analysis set comprised patients who received one or more doses of study treatment. This trial was registered at ClinicalTrials.gov, NCT03896581, and is completed. FINDINGS Between March 28, 2019, and Feb 14, 2022, 556 patients were screened and 400 patients were randomly assigned to bimekizumab 160 mg every 4 weeks (n=267) or placebo (n=133). The primary and all hierarchical secondary endpoints were met at week 16. 116 (43%) of 267 patients receiving bimekizumab reached ACR50, compared with nine (7%) of 133 patients receiving placebo (adjusted odds ratio [OR] 11·1 [95% CI 5·4-23·0], p<0·0001). 121 (69%) of 176 patients with psoriasis affecting at least 3% body surface area at baseline who received bimekizumab reached 90% or greater improvement in the Psoriasis Area and Severity Index (PASI90), compared with six (7%) of 88 patients who received placebo (adjusted OR 30·2 [12·4-73·9], p<0·0001). Treatment-emergent adverse events up to week 16 were reported in 108 (40%) of 267 patients receiving bimekizumab and 44 (33%) of 132 patients receiving placebo. There were no new safety signals and no deaths. INTERPRETATION Bimekizumab treatment led to superior improvements in joint and skin efficacy outcomes at week 16 compared with placebo in patients with psoriatic arthritis and inadequate response or intolerance to TNFα inhibitors. The safety profile of bimekizumab was consistent with previous phase 3 studies in patients with plaque psoriasis, and studies of IL-17A inhibitors. FUNDING UCB Pharma.
Collapse
Affiliation(s)
- Joseph F Merola
- Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Robert Landewé
- Amsterdam Rheumatology and Clinical Immunology Center, Amsterdam, Netherlands; Zuyderland MC, Heerlen, Netherlands
| | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| | - Philip J Mease
- Swedish Medical Center and Providence St Joseph Health and University of Washington, Seattle, WA, USA
| | | | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Akihiko Asahina
- Department of Dermatology, The Jikei University School of Medicine, Tokyo, Japan
| | - Frank Behrens
- Division of Rheumatology, University Hospital and Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Goethe University, Frankfurt am Main, Germany
| | - Dafna D Gladman
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, University of Toronto, ON, Canada
| | - Laure Gossec
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France; Rheumatology Department, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Alice B Gottlieb
- Department of Dermatology, The Icahn School of Medicine at Mt Sinai, New York, NY, USA
| | - Diamant Thaçi
- Institute and Comprehensive Center for Inflammation Medicine, University of Lübeck, Lübeck, Germany
| | - Richard B Warren
- Dermatology Centre, Salford Royal NHS Foundation Trust, Manchester NIHR Biomedical Research Centre, University of Manchester, Manchester, UK
| | | | | | | | | | | | - Laura C Coates
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Diseases University of Oxford, Oxford, UK; Oxford Biomedical Research Centre, Oxford University Hospitals NHS Trust, Oxford, UK
| |
Collapse
|
192
|
Wang Y, Li S, Bai J, Cai X, Tang S, Lin P, Sun Q, Qiao J, Fang H. Bimekizumab for the treatment of moderate-to-severe plaque psoriasis: a meta-analysis of randomized clinical trials. Ther Adv Chronic Dis 2023; 14:20406223231163110. [PMID: 37051072 PMCID: PMC10084576 DOI: 10.1177/20406223231163110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/24/2023] [Indexed: 04/07/2023] Open
Abstract
Background: Bimekizumab is a monoclonal IgG1 antibody that selectively inhibits both interleukin (IL)-17A and IL-17F, and is a promising drug for patients with moderate-to-severe plaque psoriasis. Objectives: This study aimed to assess the efficacy and safety of bimekizumab in treating patients with psoriasis and to determine the optimal maintenance dosing schedules of bimekizumab. Methods and design: Eligible trials were identified from PubMed, Cochrane Controlled Register of Trials, Embase, ClinicalTrials.gov, and Chinese medical databases. Only double-blind, randomized, active comparator, or placebo-controlled trials of bimekizumab treatment on patients with psoriasis were included in this study. Results: Five studies were identified, which included 2473 patients with moderate-to-severe plaque psoriasis. The results indicated that bimekizumab had better efficacy than placebo or active comparator for Psoriasis Area and Severity Index (PASI) 90 [risk ratio (RR) = 29.29, 1.52; 95% confidence interval (CI) = 10.30–83.30, 1.06–2.19], PASI 100 (RR = 59.87, 2.06; 95% CI = 15.06–237.99, 1.12–3.79), and Investigator’s Global Assessment scores of 0 or 1 (IGA 0/1) (RR = 21.55, 1.36; 95% CI = 9.25–50.19, 1.02–1.81). Faster onset of clinically meaningful responses was observed with bimekizumab compared with both active comparators (RR = 2.59; 95% CI = 1.32–5.10) and placebo (RR = 40.46; 95% CI = 13.19–124.13), with PASI 75 response observed at week 4 after one dose. Subgroup analysis showed no significant difference in the reduction of PASI scores between 320 mg q4w dosage and q8w dosage (RR = 1.00; 95% CI = 0.96–1.03). Rates of patients with adverse events (AEs) were comparable in the bimekizumab and active comparator groups (RR = 1.13; 95% CI = 1.01–1.26), and oral candidiasis was one of the most common treatment-emergent AEs. Conclusion: The results of this meta-analysis suggest that bimekizumab is more efficacious and has a rapid onset of action than active comparators and placebo in the treatment of moderate-to-severe plaque psoriasis. After 16 weeks of initial maintenance treatment, both bimekizumab maintenance dosing schedules (320 mg every 4 and 8 weeks) had similar efficacy.
Collapse
Affiliation(s)
- Yuqian Wang
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Sheng Li
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Juan Bai
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Xiaoxuan Cai
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Shunli Tang
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Peiyi Lin
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Qingmiao Sun
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Jianjun Qiao
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou 310000, People’s Republic of China
| | - Hong Fang
- Department of Dermatology, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou 310000, People’s Republic of China
| |
Collapse
|
193
|
Psoriatic arthritis: review of potential biomarkers predicting response to TNF inhibitors. Inflammopharmacology 2023; 31:77-87. [PMID: 36508130 PMCID: PMC9957889 DOI: 10.1007/s10787-022-01092-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/18/2022] [Indexed: 12/14/2022]
Abstract
Psoriatic arthritis (PsA) is a chronic and painful inflammatory immune-mediated disease. It affects up to 40% of people with psoriasis and it is associated with several comorbidities such as obesity, diabetes, metabolic syndrome, and hypertension. PsA is difficult to diagnose because of its diverse symptoms, namely axial and peripheral arthritis, enthesitis, dactylitis, skin changes, and nail dystrophy. Different drugs exist to treat the inflammation and pain. When patients do not respond to conventional drugs, they are treated with biologic drugs. Tumour necrosis factor inhibitors (TNFi's) are commonly given as the first biologic drug; beside being expensive, they also lack efficacy in 50% of patients. A biomarker predicting individual patient's response to TNFi would help treating them earlier with an appropriate biologic drug. This study aimed to review the literature to identify potential biomarkers that should be investigated for their predictive ability. Several such biomarkers were identified, namely transmembrane TNFα (tmTNF), human serum albumin (HSA) and its half-life receptor, the neonatal Fc receptor (FcRn) which is also involved in IgG lifespan; calprotectin, high mobility group protein B1 (HMGB1) and advanced glycation end products (AGEs) whose overexpression lead to excessive production of pro-inflammatory cytokines; lymphotoxin α (LTα) which induces inflammation by binding to TNF receptor (TNFR); and T helper 17 (Th17) cells which induce inflammation by IL-17A secretion.
Collapse
|
194
|
Xiang C, Li H, Tang W. Targeting CSF-1R represents an effective strategy in modulating inflammatory diseases. Pharmacol Res 2023; 187:106566. [PMID: 36423789 DOI: 10.1016/j.phrs.2022.106566] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Colony-stimulating factor-1 receptor (CSF-1R), also known as FMS kinase, is a type I single transmembrane protein mainly expressed in myeloid cells, such as monocytes, macrophages, glial cells, and osteoclasts. The endogenous ligands, colony-stimulating factor-1 (CSF-1) and Interleukin-34 (IL-34), activate CSF-1R and downstream signaling pathways including PI3K-AKT, JAK-STATs, and MAPKs, and modulate the proliferation, differentiation, migration, and activation of target immune cells. Over the past decades, the promising therapeutic potential of CSF-1R signaling inhibition has been widely studied for decreasing immune suppression and escape in tumors, owing to depletion and reprogramming of tumor-associated macrophages. In addition, the excessive activation of CSF-1R in inflammatory diseases is consecutively uncovered in recent years, which may result in inflammation in bone, kidney, lung, liver and central nervous system. Agents against CSF-1R signaling have been increasingly investigated in preclinical or clinical studies for inflammatory diseases treatment. However, the pathological mechanism of CSF-1R in inflammation is indistinct and whether CSF-1R signaling can be identified as biomarkers remains controversial. With the background information aforementioned, this review focus on the dialectical roles of CSF-1R and its ligands in regulating innate immune cells and highlights various therapeutic implications of blocking CSF-1R signaling in inflammatory diseases.
Collapse
Affiliation(s)
- Caigui Xiang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heng Li
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Wei Tang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
195
|
Park J, Son MJ, Ho CC, Lee SH, Kim Y, An J, Lee SK. Transcriptional inhibition of STAT1 functions in the nucleus alleviates Th1 and Th17 cell-mediated inflammatory diseases. Front Immunol 2022; 13:1054472. [PMID: 36591260 PMCID: PMC9800178 DOI: 10.3389/fimmu.2022.1054472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022] Open
Abstract
T helper 1 cells (Th1 cells) and T helper 17 cells (Th17 cells) play pivotal roles in the pathogenesis of various autoimmune diseases, including psoriasis and inflammatory bowel disease (IBD). Signal transducer and activator of transcription 1 (STAT1) regulates the Th1 and Th17 cell lineage commitment at an early stage and maintains their immunological functions in vitro and in vivo. The previous strategies to block STAT1 functions to treat autoimmune diseases inhibit Th1 cell activity but simultaneously cause hyper-activation of Th17 cells. Herein, to modulate the functions of pathogenic Th1 and Th17 cells without genetic modification in normal physiological conditions, we generated the nucleus-deliverable form of the transcription modulation domain of STAT1 (ndSTAT1-TMD), which can be transduced into the nucleus of the target cells in a dose- and time-dependent manner without affecting the cell viability and T cell activation signaling events. ndSTAT1-TMD significantly blocked the differentiation of naïve CD4+ T cells into Th1 or Th17 cells via competitive inhibition of endogenous STAT1-mediated transcription, which did not influence Th2 and Treg cell differentiation. When the gene expression profile of Th1 or Th17 cells after ndSTAT1-TMD treatment was analyzed by mRNA sequencing, the expression of the genes involved in the differentiation capacity and the immunological functions of Th1 or Th17 cells were substantially reduced. The therapeutic potential of ndSTAT1-TMD was tested in the animal model of psoriasis and colitis, whose pathogenesis is mainly contributed by Th1 or/and Th17 cells. The symptoms and progression of psoriasis and colitis were significantly alleviated by ndSTAT1-TMD treatment, comparable to anti-IL-17A antibody treatment. In conclusion, our study demonstrates that ndSTAT1-TMD can be a new therapeutic reagent for Th1/17 cell-mediated autoimmune diseases by modulating the functions of pathogenic Th1 and Th17 cells together.
Collapse
Affiliation(s)
- Jiyoon Park
- Department of Biotechnology, Yonsei University of Life Science and Biotechnology, Seoul, South Korea
| | - Min-Ji Son
- Department of Biotechnology, Yonsei University of Life Science and Biotechnology, Seoul, South Korea
| | - Chun-Chang Ho
- Department of Biotechnology, Yonsei University of Life Science and Biotechnology, Seoul, South Korea
| | - Su-Hyeon Lee
- Department of Biotechnology, Yonsei University of Life Science and Biotechnology, Seoul, South Korea
| | - Yuna Kim
- Department of Biotechnology, Yonsei University of Life Science and Biotechnology, Seoul, South Korea
| | - Jaekyeung An
- Department of Biotechnology, Yonsei University of Life Science and Biotechnology, Seoul, South Korea
| | - Sang-Kyou Lee
- Department of Biotechnology, Yonsei University of Life Science and Biotechnology, Seoul, South Korea
- Good T Cells, Inc., Seoul, South Korea
| |
Collapse
|
196
|
Hutton J, Mease P, Jadon D. Horizon scan: State-of-the-art therapeutics for psoriatic arthritis. Best Pract Res Clin Rheumatol 2022; 36:101809. [PMID: 36567224 DOI: 10.1016/j.berh.2022.101809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Psoriatic arthritis (PsA) is a common immune-mediated inflammatory disease (IMID) that can present with a heterogenous clinical phenotype. The advent of advanced therapies has substantially improved patient outcomes, but many patients still have suboptimal or unsustained response, resulting in morbidity, structural damage and functional impairment. There remains a need for better therapeutic options and precision medicine approaches to improve outcomes for patients with PsA. This review synthesises recently approved the state-of-the-art therapeutics for PsA, including inhibitors of IL-23, Janus kinase (JAK), tyrosine kinase 2 (TYK2) and dual-target IL-17A/F. The evidence base for emerging therapeutics, including MK-2 inhibitors, nano-IL-17 inhibitors, nanobodies and other dual-target therapies for PsA is also reviewed. Potential future therapeutic strategies and unmet research needs are discussed.
Collapse
Affiliation(s)
- Joseph Hutton
- Department of Rheumatology, Cambridge University Hospitals NHSFT, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Philip Mease
- Seattle Rheumatology Associates, 601 Broadway, Suite 600, Seattle, WA 98122, USA.
| | - Deepak Jadon
- Rheumatology Research Unit, Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
197
|
Esquivel-García R, Seker A, Abu-Lail NI, García-Pérez M, Ochoa-Zarzosa A, García-Pérez ME. Ethanolic extract, solvent fractions, and bio-oils from Urtica subincisa: Chemical composition, toxicity, and anti-IL-17 activity on HaCaT keratinocytes. J Herb Med 2022. [DOI: 10.1016/j.hermed.2022.100599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
198
|
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
|
199
|
Sun L, Yu G, Jiang H, Shi K, Huang D, Bao H, Huang Y, Che L, Chen M. Experimental study on the anti-inflammatory effect of Mongolian medicine Sendeng Decoction on mice with psoriasis. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
200
|
Drakos A, Vender R. A Review of the Clinical Trial Landscape in Psoriasis: An Update for Clinicians. Dermatol Ther (Heidelb) 2022; 12:2715-2730. [PMID: 36319883 PMCID: PMC9674811 DOI: 10.1007/s13555-022-00840-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
As our understanding of the pathogenesis of psoriasis has evolved over the past two decades, so has the number of treatment options. The introduction of biologic agents targeting specific cytokines in the interleukin (IL)-23/IL-17 pathway has proven successful in promoting skin clearance among patients. However, their use is often limited owing to cost, parenteral administration, and possible reduced efficacy over time. Topical therapies have also seen limited advancement, with agents such as corticosteroids and vitamin D derivatives remaining the mainstay of treatment, despite side effects limiting their long-term use. New therapeutic agents are needed to improve disease management for patients. In this review, we summarize pipeline and recently approved therapies undergoing clinical trials for psoriasis during a 12-month search period (30 June 2021 to 30 June 2022) using ClinicalTrials.gov. New-generation biologics and oral small molecules in phase II or III development were included, and pivotal data identified through various search modalities (PubMed, conference presentations, etc.) evaluating each drug candidate will be discussed. Topical therapies will also be discussed in line with recent US Food and Drug Administration approvals. As new therapies continue to enter the treatment landscape, long-term data and comparative trials will be needed to better understand their place among existing therapeutic agents.
Collapse
Affiliation(s)
| | - Ronald Vender
- Dermatrials Research Inc., Venderm Innovations in Psoriasis, Hamilton, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|