51
|
Lin L, Huang Z, Jianchi M, Guo Z, Shi Z, Tang Z, Guo Q, Xiong H. Artesunate alleviates psoriasis-like dermatitis by reducing interleukin-23 expression in tumor necrosis factor-alpha-induced HaCaT cells. Clin Exp Pharmacol Physiol 2023; 50:903-913. [PMID: 37635387 DOI: 10.1111/1440-1681.13816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 06/29/2023] [Accepted: 08/06/2023] [Indexed: 08/29/2023]
Abstract
Artesunate (ART), an antimalarial drug with a multifunctional immunomodulatory effect, reduces psoriasis disease. ART can alleviate psoriasis-like dermatitis in mice but has no effect on proinflammatory cytokines in the blood. Thus, we hypothesized that the skin might be the target tissue of ART during the treatment of psoriasis. The interleukin (IL)-23/IL-17 axis has a key role in the pathogenesis of psoriasis. However, whether and how ART manipulates the IL-23 signal during psoriasis is unknown. This study found that IL-23 is highly expressed in the epidermis of psoriasis lesions and positively correlated with histological neutrophil infiltration and clinical psoriasis area and severity index (PASI) scores. Furthermore, ART inhibits the migration and cell cycle, as well as tumor necrosis factor-alpha (TNF-α)-induced IL-23 expression in HaCaT cells in a dose-dependent manner, probably through interference with the nuclear factor kappa B (NF-κB) signalling pathway. Animal experiments in imiquimod (IMQ)-induced psoriasis-like mice model also suggested that ART dose-dependently reduces IL-23 in the epidermis and ameliorates neutrophil infiltration. These findings thus provide further molecular evidence supporting ART as a promising drug for psoriasis in clinic.
Collapse
Affiliation(s)
- Lixian Lin
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhongzhou Huang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ma Jianchi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhixuan Guo
- Department of Dermatology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhenrui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zengqi Tang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing Guo
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Xiong
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
52
|
Zhang C, Tang B, Zheng X, Luo Q, Bi Y, Deng H, Yu J, Lu Y, Han L, Chen H, Lu C. Analysis of the potential pyroptosis mechanism in psoriasis and experimental validation of NLRP3 in vitro and in vivo. Int Immunopharmacol 2023; 124:110811. [PMID: 37647679 DOI: 10.1016/j.intimp.2023.110811] [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: 02/12/2023] [Revised: 08/07/2023] [Accepted: 08/15/2023] [Indexed: 09/01/2023]
Abstract
Pyroptosis provides new perspectives on the mechanisms underlying psoriasis and the development of new treatment strategies. Here, we aimed to identify pyroptosis-related genes (PRGs) involved in the pathogenesis and progression of psoriasis. Based on the inclusion/exclusion criteria, three gene datasets were downloaded from the Gene Expression Omnibus (GEO) database. Differential gene expression, weighted gene co-expression network analysis (WGCNA), and functional enrichment analyses were performed to identify candidate PRGs for psoriasis. Least absolute shrinkage and selection operator (LASSO) regression was used to identify hub genes, and receiver operating characteristic (ROC) curves were used to determine the clinical value of the hub genes. Imiquimod-inducedpsoriasis-like mice and lipopolysaccharide (LPS)-induced RAW 264.7 cells were employed to verify the pro-inflammatory factors that may drive changes in pyroptosis. In total, 159 skin samples were analysed, and a total of 21 common targets were obtained by crossing PRGs with all the differentially expressed genes (DEGs) in different disease states. 11 genes were identified via LASSO screening. Similarly, the last six PRGs biomarkers and the green module genes were screened. All hub genes with an area under the ROC curve > 0.5 were intersected, and NLRP3 was identified. NLRP3 expression was elevated in imiquimod-induced psoriatic lesions in mice and LPS-stimulated RAW 264.7 cells. The mice exhibited reduced psoriasis area and severity index scores, hyperproliferation, and inflammation after treatment with MCC950 (a specific inhibitor of NLRP3). MCC950 decreased IL-1β, IL-6, and TNF-α mRNA expression, and NLRP3 and p-p65 protein levels in LPS-stimulated RAW 264.7 cells. Our study indicates that NLRP3 may be a promising therapeutic target for the treatment of psoriasis.
Collapse
Affiliation(s)
- Chen Zhang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of clinical pharmacy, Guangzhou First People's Hospital, Guangzhou, China
| | - Bin Tang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China; Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine Dermatology, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuwei Zheng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qianqian Luo
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Bi
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hao Deng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China; Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine Dermatology, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjie Yu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China; Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine Dermatology, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue Lu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Ling Han
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China; Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine Dermatology, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haiming Chen
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China; Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine Dermatology, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Chuanjian Lu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China; Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine Dermatology, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
53
|
Son SE, Im DS. Therapeutic effects of candesartan in inflammatory skin disorders by suppressing Th17 differentiation. Int Immunopharmacol 2023; 124:110995. [PMID: 37801970 DOI: 10.1016/j.intimp.2023.110995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/15/2023] [Accepted: 09/22/2023] [Indexed: 10/08/2023]
Abstract
As angiotensin II is associated with inflammation, type I angiotensin II receptor blockers (ARBs) exibit anti-inflammatory effects in patients with hypertension as well as inflammatory disease animal models including arthritis models. The present study aimed to investigate whether ARBs exert anti-inflammatory effects in vivo in skin disorders. We tested effects of ARBs on 1-chloro-2,4-dinitrobenzene(CDNB)-induced atopic dermatitis-like and imiquimod-induced psoriasis-like skin models. CDNB-induced atopic dermatitis-like skin lesions were suppressed by administration of candesartan or telmisartan. The suppressive effect of telmisartan was blocked by the presence of GW9662, a selective PPARγ inhibitor, but not that of candesartan. Both ARBs suppressed increases in pro-inflammatory cytokine (IL-4, IL-13, IFN-γ, and IL-17A) levels, and GW9662 inhibited telmisartan-induced suppression but not candesartan. Candesartan significantly inhibited in vitro differentiation of naïve T cells into Th17 cells to a greater extent than telmisartan. In the imiquimod-induced psoriasis model, whose primary etiology is activation of IL-23/IL-17 axis, candesartan significantly suppressed psoriasis-like skin lesions and Th17 cell populations in both lymph nodes and spleens to a greater extent than telmisartan. Overall, certain ARBs may have anti-inflammatory effects in skin diseases. Candesartan may have therapeutic implications in inflammatory skin disorders by suppressing Th17 differentiation, while telmisartan might have therapeutic potential by activating PPARγ.
Collapse
Affiliation(s)
- So-Eun Son
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dong-Soon Im
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
54
|
Ke Y, Li BZ, Nguyen K, Wang D, Wang S, Young CD, Wang XJ. IL-22RA2 Is a SMAD7 Target Mediating the Alleviation of Dermatitis and Psoriatic Phenotypes in Mice. J Invest Dermatol 2023; 143:2243-2254.e10. [PMID: 37211203 PMCID: PMC11127768 DOI: 10.1016/j.jid.2023.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/23/2023]
Abstract
Long-term management of inflammatory skin diseases is challenging because of side effects from repeated use of systemic treatments or topical corticosteroids. This study sought to identify the mechanisms and developmental therapeutics for these diseases using genetic models and pharmacological approaches. We found that mice overexpressing SMAD7 in keratinocytes but not mice overexpressing the N-terminal domain of SMAD7 (i.e., N-SMAD7) were resistant to imiquimod-induced T helper 1/17- and T helper 2-type inflammation. We generated a Tat-PYC-SMAD7 (truncated SMAD7 protein encompassing C-terminal SMAD7 and PY motif fused with cell-penetrating Tat peptide). Topically applied Tat-PYC-SMAD7 to inflamed skin entered cells upon contact and attenuated imiquimod-, 2,4-dinitrofluorobenzene-, and tape-stripping-induced inflammation. RNA-sequencing analyses of mouse skin exposed to these insults showed that in addition to inhibiting TGFβ/NF-κB, SMAD7 blunted IL-22/signal transducer and activator of transcription 3 activation and associated pathogenesis, which is due to SMAD7 transcriptionally upregulating IL-22 antagonist IL-22RA2. Mechanistically, SMAD7 facilitated nuclear translocation and DNA binding of C/EBPβ to IL22RA2 promoter for IL22RA2 transactivation. Consistent with the observations in mice mentioned earlier, transcript levels of IL22RA2 were increased in human atopic dermatitis and psoriasis lesions with clinical remission. Our study identified the anti-inflammation functional domain of SMAD7 and suggests the mechanism and feasibility for developing SMAD7-based biologics as a topical therapy for skin inflammatory disorders.
Collapse
Affiliation(s)
- Yao Ke
- Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA; Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California, USA
| | - Ben-Zheng Li
- Department of Physiology and Biophysics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Khoa Nguyen
- Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Donna Wang
- Allander Biotechnologies, Aurora, Colorado, USA
| | - Suyan Wang
- Allander Biotechnologies, Aurora, Colorado, USA
| | - Christian D Young
- Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA; Allander Biotechnologies, Aurora, Colorado, USA.
| | - Xiao-Jing Wang
- Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA; Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California, USA; Allander Biotechnologies, Aurora, Colorado, USA.
| |
Collapse
|
55
|
Ricci AA, Dapavo P, Mastorino L, Roccuzzo G, Wolff S, Ribero S, Cassoni P, Senetta R, Quaglino P. Exploring Psoriasis Inflammatory Microenvironment by NanoString Technologies. J Clin Med 2023; 12:6820. [PMID: 37959285 PMCID: PMC10650153 DOI: 10.3390/jcm12216820] [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: 09/26/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease whose molecular mechanisms and microenvironment are poorly understood. We performed gene expression analysis through the nCounter® PanCancer Immune Profiling Panel (NanoString Technologies, Seattle, WA, USA) on 22 FFPE punch biopsies from 19 psoriasis-affected patients. A subset of five cases was analyzed before (T0) and after 6 months (T6) of treatment with dimethyl fumarate (DMF) to address immune microenvironment changes. Molecular comparisons according to biopsy site and age of onset showed a different distribution of innate immune cells (mast cells, macrophages, NK cells, and DC cells) and pathways (complement regulation and transporter functions). The analysis according to PASI (Psoriasis Area and Severity Index) led to non-significant results, suggesting no link between molecular expression profile and clinical amount of skin disease. In DMF-treated patients, we observed a strong immunomodulatory effect after treatment: A subversion of exhausted CD8 T cells, NK CD56dim cells, Tregs, neutrophils, CD45+ cells, T cells, B cells, and macrophages was reported between the two analyzed time-points, as well as the reduction in pro-inflammatory pathways and molecules, including cytotoxicity, pathogen defense, antigen processing, adhesion, cell cycle, chemokines, cytokines, and interleukins. The inflammatory psoriatic microenvironment can be modulated using DMF with encouraging results, achieving an immune-tolerant and non-inflammatory condition through the regulation of both innate and adaptive immunity.
Collapse
Affiliation(s)
- Alessia Andrea Ricci
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (A.A.R.); (P.C.)
| | - Paolo Dapavo
- Department of Medical Sciences, Section of Dermatology, University of Turin, 10126 Turin, Italy; (P.D.); (L.M.); (G.R.); (S.W.); (P.Q.)
| | - Luca Mastorino
- Department of Medical Sciences, Section of Dermatology, University of Turin, 10126 Turin, Italy; (P.D.); (L.M.); (G.R.); (S.W.); (P.Q.)
| | - Gabriele Roccuzzo
- Department of Medical Sciences, Section of Dermatology, University of Turin, 10126 Turin, Italy; (P.D.); (L.M.); (G.R.); (S.W.); (P.Q.)
| | - Samanta Wolff
- Department of Medical Sciences, Section of Dermatology, University of Turin, 10126 Turin, Italy; (P.D.); (L.M.); (G.R.); (S.W.); (P.Q.)
| | - Simone Ribero
- Department of Medical Sciences, Section of Dermatology, University of Turin, 10126 Turin, Italy; (P.D.); (L.M.); (G.R.); (S.W.); (P.Q.)
| | - Paola Cassoni
- Pathology Unit, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (A.A.R.); (P.C.)
| | - Rebecca Senetta
- Pathology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy;
| | - Pietro Quaglino
- Department of Medical Sciences, Section of Dermatology, University of Turin, 10126 Turin, Italy; (P.D.); (L.M.); (G.R.); (S.W.); (P.Q.)
| |
Collapse
|
56
|
Zhang P, Su Y, Li S, Chen H, Wu R, Wu H. The roles of T cells in psoriasis. Front Immunol 2023; 14:1081256. [PMID: 37942312 PMCID: PMC10628572 DOI: 10.3389/fimmu.2023.1081256] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 09/29/2023] [Indexed: 11/10/2023] Open
Abstract
Psoriasis is a recurring inflammatory skin condition characterized by scaly, red patches on the skin. It affects approximately 3% of the US population and is associated with histological changes such as epidermal hyperplasia, increased blood vessel proliferation, and infiltration of leukocytes into the skin's dermis. T cells, which are classified into various subtypes, have been found to play significant roles in immune-mediated diseases, particularly psoriasis. This paper provides a review of the different T lymphocyte subtypes and their functions in psoriasis, as well as an overview of targeted therapies for treating psoriasis.
Collapse
Affiliation(s)
| | | | | | | | - Ruifang Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
57
|
Zeng F, Lu H, Wu M, Dai C, Li J, Wang J, Hu G. Topical application of TOPK inhibitor OTS514 suppresses psoriatic progression by inducing keratinocytes cell cycle arrest and apoptosis. Exp Dermatol 2023; 32:1823-1833. [PMID: 37578092 DOI: 10.1111/exd.14909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/13/2023] [Accepted: 07/29/2023] [Indexed: 08/15/2023]
Abstract
T-LAK cell-oriented protein kinase (TOPK) potently promotes malignant proliferation of tumour cells and is considered as a maker of tumour progression. Psoriasis is a common inflammatory skin disease characterized by abnormal proliferation of keratinocytes. However, the role of TOPK in psoriasis has not been well elucidated. This study aims to investigate the expression and role of TOPK in psoriasis, and the role of TOPK inhibitor in psoriasis attenuation. Gene Expression Omnibus datasets derived from psoriasis patients and psoriatic model mice were screened for analysis. Skin specimens from psoriasis patients were collected for TOPK immunohistochemical staining to investigate the expression and localization of TOPK. Next, psoriatic mice model was established to further confirm TOPK expression pattern. Then, TOPK inhibitor was applied to investigate the role of TOPK in psoriasis progression. Finally, cell proliferation assay, apoptosis assay and cell cycle analysis were performed to investigate the potential mechanism involved. Our study showed that TOPK was upregulated in the lesions of both psoriasis patients and psoriatic model mice, and TOPK levels were positively associated with psoriasis progression. TOPK was upregulated in psoriatic lesions and expressed predominantly by epidermal keratinocytes. In addition, TOPK levels in epidermal keratinocytes were positively correlated with epidermal hyperplasia. Furthermore, topical application of TOPK inhibitor OTS514 obviously alleviated disease severity and epidermal hyperplasia. Mechanismly, inhibiting TOPK induces G2/M phase arrest and apoptosis of keratinocytes, thereby attenuating epidermal hyperplasia and disease progression. Collectively, this study identifies that upregulation of TOPK in keratinocytes promotes psoriatic progression, and inhibiting TOPK attenuates epidermal hyperplasia and psoriatic progression.
Collapse
Affiliation(s)
- Fanfan Zeng
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Lu
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengjun Wu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chan Dai
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianyu Li
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinbiao Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoyun Hu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
58
|
Teng YS, Yu S. Molecular Mechanisms of Cutaneous Immune-Related Adverse Events (irAEs) Induced by Immune Checkpoint Inhibitors. Curr Oncol 2023; 30:6805-6819. [PMID: 37504358 PMCID: PMC10378098 DOI: 10.3390/curroncol30070498] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/29/2023] Open
Abstract
Over the past few decades, immune checkpoint inhibitors (ICIs) have emerged as promising therapeutic options for the treatment of various cancers. These novel treatments effectively target key mediators of immune checkpoint pathways. Currently, ICIs primarily consist of monoclonal antibodies that specifically block cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death 1 (PD-1), programmed cell death-ligand 1 (PD-L1), and lymphocyte activation gene 3 protein (LAG-3). Despite the notable efficacy of ICIs in cancer treatment, they can also trigger immune-related adverse events (irAEs), which present as autoimmune-like or inflammatory conditions. IrAEs have the potential to affect multiple organ systems, with cutaneous toxicities being the most commonly observed. Although cutaneous irAEs are typically of low-grade severity and can usually be managed effectively, there are cases where severe irAEs can become life-threatening. Therefore, early recognition and a comprehensive understanding of the mechanisms underlying cutaneous irAEs are crucial for improving clinical outcomes in cancer patients. However, the precise pathogenesis of cutaneous irAEs remains unclear. This review focuses on the skin manifestations induced by ICIs, the prognosis related to cutaneous irAEs, and the exploration of potential mechanisms involved in cutaneous irAEs.
Collapse
Affiliation(s)
- Yi-Shan Teng
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Sebastian Yu
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Dermatology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| |
Collapse
|
59
|
Singh VK, Sahoo D, Agrahari K, Khan A, Mukhopadhyay P, Chanda D, Yadav NP. Anti-inflammatory, anti-proliferative and anti-psoriatic potential of apigenin in RAW264.7 cells, HaCaT cells and psoriasis like dermatitis in BALB/c mice. Life Sci 2023:121909. [PMID: 37414141 DOI: 10.1016/j.lfs.2023.121909] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/30/2023] [Accepted: 07/02/2023] [Indexed: 07/08/2023]
Abstract
AIMS Psoriasis is an immune-mediated skin disease characterized by keratinocytes hyperproliferation, abnormal differentiation and inflammation. Therefore, this study aimed to investigate in-vitro and in-vivo anti-inflammatory and anti-proliferative activity to evaluate anti-psoriatic potential of apigenin. MAIN METHODS For in-vivo study, 5 % imiquimod cream was used to induce psoriasis-like skin inflammation in BALB/c mice to mimic human psoriatic conditions. PASI scores, CosCam score, histopathology, immunohistochemistry, qRT-PCR, and ELISA were done to evaluate the anti-psoriatic potential of topically applied apigenin. For in-vitro studies, LPS-induced inflammation in RAW264.7 was done, and qRT-PCR, ELISA, and immunofluorescence were conducted to evaluate the anti-inflammatory activity of apigenin. Migration and cell doubling assay in HaCaT cells were performed to assess the anti-proliferative effect of apigenin. Acute dermal toxicity profile of apigenin has also been done as per OECD guidelines. KEY FINDINGS Results showed that apigenin significantly reduce the PASI and CosCam scores, ameliorate the deteriorating histopathology, and effectively downregulated the expression of CCR 6, IL-17A, and NF-κB. Apigenin effectively downregulated the expression and secretion of pro-inflammatory cytokines through IL-23/IL-17/IL-22 axis. Apigenin suppressed nuclear translocation of NF-κB in LPS-induced RAW 264.7 cells. Cell migration and cell doubling assay in HaCaT cells showing the anti-proliferative potential of apigenin. Apigenin was found safe in acute dermal toxicity study. SIGNIFICANCE Apigenin was found effective against psoriasis in both in-vitro and in-vivo models suggesting apigenin as a potential candidate for the development of anti-psoriatic agent.
Collapse
Affiliation(s)
- Vipin Kumar Singh
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Debasish Sahoo
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India
| | - Kirti Agrahari
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India
| | - Ammar Khan
- Plant Biotechnology division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Pradipto Mukhopadhyay
- Plant Biotechnology division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Debabrata Chanda
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Narayan Prasad Yadav
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, Uttar Pradesh 226015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
60
|
Zhang G, Xia Z, Tian C, Xia A, You J, Liu J, Yang S, Li L. Discovery of 5-((1H-indazol-3-yl) methylene)-2-thioxoimidazolidin-4-one derivatives as a new class of AHR agonists with anti-psoriasis activity in a mouse model. Bioorg Med Chem Lett 2023:129383. [PMID: 37348572 DOI: 10.1016/j.bmcl.2023.129383] [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: 05/21/2023] [Revised: 06/15/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023]
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand dependent transcription factor and participates in the regulation of the immune balance of Th17/22 and Treg cells. It has been found to be widely expressed in the skin, and involved in the pathology of psoriasis. Therefore, AHR is thought as a potential intervention target for psoriasis. Here, we report the discovery of 5-((1H-indazol-3-yl) methylene)-2-thioxoimidazolidin-4-one derivatives as a new class of AHR agonists. Structure-activity relationship analyses led to the identification of the most active compound, 5- ((1H-indazol-3-yl)methylene) -3- (prop-2-yn-1-yl) -2-thiooimidazolidin-4-one (24e), which exhibited an EC50 value of 0.015 µM against AHR. Mechanism of action studies showed that 24e regulated the expression of CYP1A1 by activating the AHR pathway. Topical administration of 24e substantially alleviated imiquimod (IMQ)-induced psoriasis-like skin lesion. Overall, compound 24e could be a good lead compound for drug discovery against psoriasis, and hence deserving further in-depth studies.
Collapse
Affiliation(s)
- Guo Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziyi Xia
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chenyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Anjie Xia
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing You
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shengyong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linli Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
61
|
Maeda K, Tanioka T, Takahashi R, Watanabe H, Sueki H, Takimoto M, Hashimoto SI, Ikeo K, Miwa Y, Kasama T, Iwamoto S. MCAM+CD161- Th17 Subset Expressing CD83 Enhances Tc17 Response in Psoriasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1867-1881. [PMID: 37186262 DOI: 10.4049/jimmunol.2200530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 03/27/2023] [Indexed: 05/17/2023]
Abstract
Recent studies have highlighted the pathogenic roles of IL-17-producing CD8+ T cells (T-cytotoxic 17 [Tc17]) in psoriasis. However, the underlying mechanisms of Tc17 induction remain unclear. In this study, we focused on the pathogenic subsets of Th17 and their mechanism of promotion of Tc17 responses. We determined that the pathogenic Th17-enriched fraction expressed melanoma cell adhesion molecule (MCAM) and CCR6, but not CD161, because this subset produced IL-17A abundantly and the presence of these cells in the peripheral blood of patients has been correlated with the severity of psoriasis. Intriguingly, the serial analysis of gene expression revealed that CCR6+MCAM+CD161-CD4+ T cells displayed the gene profile for adaptive immune responses, including CD83, which is an activator for CD8+ T cells. Coculture assay with or without intercellular contact between CD4+ and CD8+ T cells showed that CCR6+MCAM+CD161-CD4+ T cells induced the proliferation of CD8+ T cells in a CD83-dependent manner. However, the production of IL-17A by CD8+ T cells required exogenous IL-17A, suggesting that intercellular contact via CD83 and the production of IL-17A from activated CD4+ T cells elicit Tc17 responses. Intriguingly, the CD83 expression was enhanced in the presence of IL-15, and CD83+ cells stimulated with IL-1β, IL-23, IL-15, and IL-15Rα did not express FOXP3. Furthermore, CCR6+MCAM+CD161-CD4+ T cells expressing CD83 were increased in the peripheral blood of patients, and the CD83+ Th17-type cells accumulated in the lesional skin of psoriasis. In conclusion, pathogenic MCAM+CD161- Th17 cells may be involved in the Tc17 responses via IL-17A and CD83 in psoriasis.
Collapse
Affiliation(s)
- Kohei Maeda
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan
| | - Toshihiro Tanioka
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan
| | - Rei Takahashi
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan
| | - Hideaki Watanabe
- Department of Dermatology, Showa University School of Medicine, Tokyo, Japan
| | - Hirohiko Sueki
- Department of Dermatology, Showa University School of Medicine, Tokyo, Japan
| | - Masafumi Takimoto
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shin-Ichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kazuho Ikeo
- DNA Data Analysis Laboratory, National Institute of Genetics, Shizuoka, Japan
| | - Yusuke Miwa
- Department of Internal Medicine, Division of Rheumatology, Showa University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Kasama
- Department of Internal Medicine, Division of Rheumatology, Showa University School of Medicine, Tokyo, Japan
| | - Sanju Iwamoto
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology, and Therapeutics, Showa University School of Pharmacy, Tokyo, Japan
| |
Collapse
|
62
|
Guo Y, Jin L, Dong L, Zhang M, Kuang Y, Chen X, Zhu W, Yin M. NHWD-1062 ameliorates inflammation and proliferation by the RIPK1/NF-κB/TLR1 axis in Psoriatic Keratinocytes. Biomed Pharmacother 2023; 162:114638. [PMID: 37011486 DOI: 10.1016/j.biopha.2023.114638] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/04/2023] Open
Abstract
Psoriasis is a common chronic inflammatory skin disease. RIPK1 plays an important role in inflammatory diseases. At present, the clinical efficacy of the RIPK1 inhibitor is limited and the regulatory mechanism is unclear in the treatment of psoriasis. Therefore, our team developed a new RIPK1 inhibitor, NHWD-1062, which showed a slightly lower IC50 in U937 cells than that of GSK'772 (a RIPK1 inhibitor in clinical trials) (11 nM vs. 14 nM), indicating that the new RIPK1 inhibitor was no less inhibitory than GSK'772. In this study, we evaluated the therapeutic effects of NHWD-1062 using an IMQ-induced mouse model of psoriasis and explored the precise regulatory mechanism involved. We found that gavage of NHWD-1062 significantly ameliorated the inflammatory response and inhibited the abnormal proliferation of the epidermis in IMQ-induced psoriatic mice. We then elucidated the mechanism of NHWD-1062, which was that suppressed the proliferation and inflammation of keratinocytes in vitro and in vivo through the RIPK1/NF-κB/TLR1 axis. Dual-luciferase reporter assay indicated that P65 can directly target the TLR1 promoter region and activate TLR1 expression, leading to inflammation. In summary, our study demonstrates that NHWD-1062 alleviates psoriasis-like inflammation by inhibiting the activation of the RIPK1/NF-κB/TLR1 axis, which has not been previously reported and further provides evidence for the clinical translation of NHWD-1062 in the treatment of psoriasis.
Collapse
Affiliation(s)
- Yiyan Guo
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan, China; Furong Laboratory, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha 410008, Hunan, China
| | - Liping Jin
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan, China; Furong Laboratory, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha 410008, Hunan, China
| | - Liang Dong
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan, China; Furong Laboratory, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha 410008, Hunan, China
| | - Mi Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan, China; Furong Laboratory, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha 410008, Hunan, China
| | - Yehong Kuang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan, China; Furong Laboratory, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha 410008, Hunan, China
| | - Xiang Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan, China; Furong Laboratory, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha 410008, Hunan, China
| | - Wu Zhu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan, China; Furong Laboratory, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha 410008, Hunan, China.
| | - Mingzhu Yin
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha 410008, Hunan, China; Furong Laboratory, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
63
|
Galluzzo M, Vellucci L, Marcelli L, Paganini C, Bianchi L, Talamonti M. Deucravacitinib, a selective tyrosine kinase 2 inhibitor, for the treatment of moderate-to-severe plaque psoriasis. Expert Opin Pharmacother 2023; 24:981-988. [PMID: 37147879 DOI: 10.1080/14656566.2023.2211764] [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: 05/07/2023]
Abstract
INTRODUCTION Protein kinases play a crucial role in intracellular signaling pathways that lead to inflammation and cell proliferation. New understandings of the involvement of these metabolic pathways in the pathogenesis of psoriasis allowed the development of a new class of drugs. Unlike biologics, these compounds work by blocking intracellular targets involved in the immune response. AREAS COVERED Deucravacitinib is an oral small molecule inhibitor of TYK2 that binds the pseudokinase domain and locks the kinase in an inactive state by an allosteric mechanism, arresting TYK2- mediated signaling cascades and disabling the upregulation of proinflammatory genes implicated in psoriasis. The authors present the results of phase I - III clinical trials of deucravacitinib for the treatment of psoriasis. EXPERT OPINION At week 16 about 56% of patients treated with deucravacitinib achieved PASI75. No serious infections were reported, nor were thromboembolic events or laboratory abnormalities. Efficacy was reported to be persistent and safety profiles were shown to be consistent for up to 2 years. Deucravacitinib can potentially become a safe, effective, well-tolerated treatment for patients suffering from moderate-to-severe disease. Future studies and real-life experiences will be important to determine the exact role of this drug in the treatment of psoriasis.
Collapse
Affiliation(s)
- Marco Galluzzo
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Dermatology Unit, Fondazione Policlinico "Tor Vergata", Rome, Italy
| | - Laura Vellucci
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Dermatology Unit, Fondazione Policlinico "Tor Vergata", Rome, Italy
| | - Lorenzo Marcelli
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Dermatology Unit, Fondazione Policlinico "Tor Vergata", Rome, Italy
| | - Claudia Paganini
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Dermatology Unit, Fondazione Policlinico "Tor Vergata", Rome, Italy
| | - Luca Bianchi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Dermatology Unit, Fondazione Policlinico "Tor Vergata", Rome, Italy
| | - Marina Talamonti
- Dermatology Unit, Fondazione Policlinico "Tor Vergata", Rome, Italy
| |
Collapse
|
64
|
Doppegieter M, van der Beek N, Bakker ENTP, Neumann MHA, van Bavel E. Effects of pulsed dye laser treatment in psoriasis: A nerve-wrecking process? Exp Dermatol 2023. [PMID: 37083107 DOI: 10.1111/exd.14816] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/31/2023] [Accepted: 04/09/2023] [Indexed: 04/22/2023]
Abstract
Pulsed dye laser (PDL) therapy can be effective in treating psoriasis, with a long duration of remission. Although PDL therapy, albeit on a modest scale, is being used for decades now, the underlying mechanisms responsible for the long-term remission of psoriasis remain poorly understood. The selective and rapid absorption of energy by the blood causes heating of the vascular wall and surrounding structures, like perivascular nerves. Several studies indicate the importance of nerves in psoriatic inflammation. Interestingly, denervation leads to a spontaneous remission of the psoriatic lesion. Among all dermal nerves, the perivascular nerves are the most likely to be affected during PDL treatment, possibly impairing the neuro-inflammatory processes that promote T-cell activation, expression of adhesion molecules, leukocyte infiltration and cytokine production. Repeated PDL therapy could cause a prolonged loss of innervation through nerve damage, or result in a 'reset' of neurogenic inflammation after temporary denervation. The current hypothesis provides strong arguments that PDL treatment affects nerve fibres in the skin and thereby abrogates the persistent and exaggerated inflammatory process underlying psoriasis, causing a long-term remission of psoriasis.
Collapse
Affiliation(s)
- Meagan Doppegieter
- Department of Biomedical Engineering & Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nick van der Beek
- ZBC MultiCare, Independent Treatment Center for Dermatology, Hilversum, The Netherlands
| | - Erik N T P Bakker
- Department of Biomedical Engineering & Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Martino H A Neumann
- ZBC MultiCare, Independent Treatment Center for Dermatology, Hilversum, The Netherlands
| | - Ed van Bavel
- Department of Biomedical Engineering & Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
65
|
Luo X, Su Y, Zhong L, Kuang Q, Zhu Y, Zhou X, Tang G, Fu Y, Li S, Wu R. Auranofin ameliorates psoriasis-like dermatitis in an imiquimod-induced mouse by inhibiting of inflammation and upregulating FA2H expression. Biomed Pharmacother 2023. [DOI: 10.1016/j.biopha.2023.114421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023] Open
|
66
|
Conundrum for Psoriasis and Thyroid Involvement. Int J Mol Sci 2023; 24:ijms24054894. [PMID: 36902323 PMCID: PMC10003398 DOI: 10.3390/ijms24054894] [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: 02/04/2023] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Strategies concerning thyroid anomalies in patients confirmed with psoriasis, either on clinical level or molecular levels, and their genetic findings remain an open issue. Identification of the exact subgroup of individuals that are candidates to endocrine assessments is also controversial. Our purpose in this work was to overview clinical and pathogenic data concerning psoriasis and thyroid comorbidities from a dual perspective (dermatologic and endocrine). This was a narrative review of English literature between January 2016 and January 2023. We included clinically relevant, original articles with different levels of statistical evidence published on PubMed. We followed four clusters of conditions: thyroid dysfunction, autoimmunity, thyroid cancer, and subacute thyroiditis. A new piece of information in this field was the fact that psoriasis and autoimmune thyroid diseases (ATD) have been shown to be related to the immune-based side effects of modern anticancer drugs-namely, immune checkpoint inhibitors (ICP). Overall, we identified 16 confirmatory studies, but with heterogeneous data. Psoriatic arthritis had a higher risk of positive antithyroperoxidase antibodies (TPOAb) (25%) compared to cutaneous psoriasis or control. There was an increased risk of thyroid dysfunction versus control, and hypothyroidism was the most frequent type of dysfunction (subclinical rather than clinical), among thyroid anomalies correlated with >2-year disease duration, peripheral > axial and polyarticular involvement. With a few exceptions, there was a female predominance. Hormonal imbalance included, most frequently, low thyroxine (T4) and/or triiodothyronine (T3) with normal thyroid stimulating hormone (TSH), followed by high TSH (only one study had higher total T3). The highest ratio of thyroid involvement concerning dermatologic subtypes was 59% for erythrodermic psoriasis. Most studies found no correlation between thyroid anomalies and psoriasis severity. Statistically significant odds ratios were as follows: hypothyroidism: 1.34-1.38; hyperthyroidism: 1.17-1.32 (fewer studies than hypo); ATD: 1.42-2.05; Hashimoto's thyroiditis (HT): 1.47-2.09; Graves' disease: 1.26-1.38 (fewer studies than HT). A total of 8 studies had inconsistent or no correlations, while the lowest rate of thyroid involvement was 8% (uncontrolled studies). Other data included 3 studies on patients with ATD looking for psoriasis, as well as 1 study on psoriasis and thyroid cancer. ICP was shown to potentially exacerbate prior ATD and psoriasis or to induce them both de novo (5 studies). At the case report level, data showed subacute thyroiditis due to biological medication (ustekinumab, adalimumab, infliximab). Thyroid involvement in patients with psoriasis thus remained puzzling. We observed significant data that confirmed a higher risk of identifying positive antibodies and/or thyroid dysfunction, especially hypothyroidism, in these subjects. Awareness will be necessary to improve overall outcomes. The exact profile of individuals diagnosed with psoriasis who should be screened by the endocrinology team is still a matter of debate, in terms of dermatological subtype, disease duration, activity, and other synchronous (especially autoimmune) conditions.
Collapse
|
67
|
Zhou T, Du X, Zhang L, Zheng Y, Jia T, Song X, Che D, Geng S. Suprabasin-derived polypeptides: SBSN(50-63) induces inflammatory response via TLR4-mediated mast cell activation. Inflammopharmacology 2023; 31:1329-1339. [PMID: 36745245 DOI: 10.1007/s10787-023-01137-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/11/2023] [Indexed: 02/07/2023]
Abstract
Psoriasis is a chronic, relapsing, immune-mediated, and papulosquamous skin disorder. Excessive mast cell activation, in psoriatic lesions, contributes to inflammation. Various endogenous peptides can participate in the pathogenesis of inflammatory diseases by activating mast cells. Suprabasin (SBSN) is expressed in multiple epithelial tissues and it regulates the normal epidermal barrier function. We have recently shown that suprabasin-derived polypeptides, SBSN(50-63), are significantly increased in psoriatic lesions, through differential peptide analysis. This study was conducted to clarify whether SBSN(50-63) plays a pivotal role in activating mast cells and mediating proinflammatory cytokines and chemokines production in psoriasis. The increased expression of SBSN in psoriatic lesions was confirmed by bioinformatics analysis, PCR and ELISA. Wild-type mice injected subcutaneously with SBSN(50-63) exhibited infiltration of inflammatory cells and the release of cytokines in vivo. SBSN(50-63) stimulated mouse primary mast cells (MPMC) and the laboratory of allergic disease 2 (LAD2) human mast cells to produce more inflammatory mediators than the control group, which were measured ex vivo and in vitro. Toll-like receptor 4 was identified as the receptor of SBSN on mast cells by molecular docking analysis, molecular dynamics simulation, and siRNA transfection. Collectively, SBSN(50-63) could activate mast cells through TLR4, which may increase the inflammatory response in psoriasis.
Collapse
Affiliation(s)
- Tong Zhou
- Department of Dermatology, Northwest Hospital, The Second Hospital Affiliated to Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xueshan Du
- Department of Dermatology, Northwest Hospital, The Second Hospital Affiliated to Xi'an Jiaotong University, Xi'an, 710061, China.,Center for Dermatology Disease, Precision Medical Institute, Xi'an, China
| | - Lei Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Yi Zheng
- Department of Dermatology, Northwest Hospital, The Second Hospital Affiliated to Xi'an Jiaotong University, Xi'an, 710061, China.,Center for Dermatology Disease, Precision Medical Institute, Xi'an, China
| | - Tao Jia
- Department of Dermatology, Northwest Hospital, The Second Hospital Affiliated to Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiangjin Song
- Department of Dermatology, Northwest Hospital, The Second Hospital Affiliated to Xi'an Jiaotong University, Xi'an, 710061, China
| | - Delu Che
- Department of Dermatology, Northwest Hospital, The Second Hospital Affiliated to Xi'an Jiaotong University, Xi'an, 710061, China. .,Center for Dermatology Disease, Precision Medical Institute, Xi'an, China.
| | - Songmei Geng
- Department of Dermatology, Northwest Hospital, The Second Hospital Affiliated to Xi'an Jiaotong University, Xi'an, 710061, China. .,Center for Dermatology Disease, Precision Medical Institute, Xi'an, China.
| |
Collapse
|
68
|
Tectorigenin inhibits inflammation in keratinocytes by inhibition of NLRP3 inflammasome regulated by the TLR4/NF-κB pathway. Allergol Immunopathol (Madr) 2023; 51:82-89. [PMID: 36916091 DOI: 10.15586/aei.v51i2.780] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/19/2022] [Indexed: 03/07/2023]
Abstract
BACKGROUND Psoriasis is a prevalent inflammatory skin disease characterized by excessive proliferation and abnormal differentiation of keratinocytes, and infiltration of inflammatory cells into the epidermis. However, the underlying mechanisms remain unclear. Tectorigenin is an active ingredient in traditional medicines and has anti-inflammatory activity. This research explored the effects of tectorigenin on the anti-inflammatory property, autophagy, and the underlying mechanisms in M5 ([IL-22, IL-17A, oncostatin M, IL-1α, and TNF-α])-stimulated HaCaT cells. METHODS The in vitro model of mixed M5 cytokines-stimulated HaCaT keratinocytes was established to investigate the phenotypic features in psoriasis. Cell viability was assessed by MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assay, cell proliferative rate by EdU (5-ethynyl-2'-deoxyuridine) assay, and autophagy was detected by immunofluorescence staining. After M5 exposure, the proliferative rate, protein expression of autophagy, and signaling activities of NLR family pyrin domain containing 3 (NLRP3) inflammasome and toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) were measured. The latter were quantitated using quantitative PCR and western blot, respectively. The inflammatory response was detected by enzyme-linked immunosorbent assay (ELISA). RESULTS Tectorigenin exerted a protective effect in ameliorating the hyperproliferation and inflammation of HaCaT keratinocytes induced by M5 cytokines. Furthermore, tectorigenin on keratinocytes seemed to inactivate NLRP3 inflammasome and inhibit cell proliferation and inflammation response via suppression of TLR4/NF-κB pathway. CONCLUSION This study proves that tectorigenin may be a potential therapeutic candidate for psoriasis treatment in future.
Collapse
|
69
|
Ahmad MZ, Mohammed AA, Algahtani MS, Mishra A, Ahmad J. Nanoscale Topical Pharmacotherapy in Management of Psoriasis: Contemporary Research and Scope. J Funct Biomater 2022; 14:jfb14010019. [PMID: 36662067 PMCID: PMC9867016 DOI: 10.3390/jfb14010019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
Psoriasis is a typical dermal condition that has been anticipated since prehistoric times when it was mistakenly implicit in being a variant of leprosy. It is an atypical organ-specific autoimmune disorder, which is triggered by the activation of T-cells and/or B-cells. Until now, the pathophysiology of this disease is not completely explicated and still, many research investigations are ongoing. Different approaches have been investigated to treat this dreadful skin disease using various anti-psoriatic drugs of different modes of action through smart drug-delivery systems. Nevertheless, there is no ideal therapy for a complete cure of psoriasis owing to the dearth of an ideal drug-delivery system for anti-psoriatic drugs. The conventional pharmacotherapy approaches for the treatment of psoriasis demand various classes of anti-psoriatic drugs with optimum benefit/risk ratio and insignificant untoward effects. The advancement in nanoscale drug delivery had a great impact on the establishment of a nanomedicine-based therapy for better management of psoriasis in recent times. Nanodrug carriers are exploited to design and develop nanomedicine-based therapy for psoriasis. It has a promising future in the improvement of the therapeutic efficacy of conventional anti-psoriatic drugs. The present manuscript aims to discuss the pathophysiology, conventional pharmacotherapy, and contemporary research in the area of nanoscale topical drug delivery systems for better management of psoriasis including the significance of targeted pharmacotherapy in psoriasis.
Collapse
Affiliation(s)
- Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia
| | - Abdul Aleem Mohammed
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia
| | - Mohammed S. Algahtani
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia
| | - Awanish Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, Assam, India
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia
- Correspondence: or
| |
Collapse
|
70
|
Khueangchiangkhwang S, Wu Z, Nagano I, Maekawa Y. Trichinella pseudospiralis-secreted 53 kDa protein ameliorates imiquimod-induced psoriasis by inhibiting the IL-23/IL-17 axis in mice. Biochem Biophys Rep 2022; 33:101415. [PMID: 36620087 PMCID: PMC9813687 DOI: 10.1016/j.bbrep.2022.101415] [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: 09/14/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Trichinella infection can experimentally ameliorate many autoimmune diseases. However, the immune mechanism of the amelioration and the identification of corresponding Trichinella-derived molecule(s) are still not fully elucidated. Fifty-three kDa excretory-secretory (ES) protein from Trichinella pseudospiralis (Tpp53) is a molecule like TsP53 reported as a protein exerting immune-inhibitory effect in T. spiralis. In this study, we investigated the immunomodulatory effect of Tpp53 using imiquimod (IMQ)-induced psoriasis-like dermatitis model, which is a mouse model of autoimmune disease with the pathogenic interleukin 17 (IL-17) producing CD4+ T cells (Th17) via IL-23/IL17 axis. Administrating the recombinant Tpp53 (rTpp53) mixed with IMQ cream on the skin of mice ameliorated psoriatic lesions, as revealed by the improvement of erythema, scaling, skin thickening, epidermis hyperplasia and parakeratosis, thickening of acanthosis cell layer, epidermal extension of dermis, less infiltration of inflammatory cells, and decreased expression of inflammatory marker. The increased expression of the factors related to the IL-23/IL-17 axis, including IL-17A, IL-6, Il17F and Il23a, in the skins of IMQ-treated mice was inhibited by rTpp53 treatment. Moreover, the expression of activated keratinocyte-produced cytokines, chemokines, and antimicrobial peptides in the skin was also down-regulated in rTpp53-treated IMQ-treated mice. Co-culture of splenocytes with rTpp53 inhibited IL-17A and treatment of macrophages with rTpp53 reduced IL-6 production. Overall, our study revealed that the Trichinella-secreted 53 kDa ES protein could ameliorate IMQ-induced psoriasis by inhibiting the IL-23/IL-17 axis, suggesting that Tpp53 might involve in regulating host Th17 for immune evasion and have an alternative potential for psoriasis therapy.
Collapse
Affiliation(s)
| | - Zhiliang Wu
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan,Cocorresponding author. 1-1 Yanagido, Gifu, 501-1194, Japan.
| | - Isao Nagano
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yoichi Maekawa
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan,Preemptive Food Research Center, Gifu University, Gifu, Japan,Corresponding author. 1-1 Yanagido, Gifu, 501-1194, Japan.
| |
Collapse
|
71
|
Mechanisms and Effects of Isorhamnetin on Imiquimod-Induced Psoriasiform Dermatitis in Mice. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122107. [PMID: 36556472 PMCID: PMC9786590 DOI: 10.3390/life12122107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/21/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Isorhamnetin (IRh), which has a wide range of pharmacological effects, is one of the most significant active components in the fruits of Hippophae rhamnoides L. and the leaves of Ginkgo biloba L. It protects the heart and brain, in addition to possessing anti-tumor, anti-inflammatory, antioxidant, organ protection, and anti-obesity properties. We sought to assess IRh's anti-psoriatic activity, explore its immunomodulatory properties in reducing the severity of psoriatic symptoms, and evaluate its potential immunotherapeutic effects. We used IRh to treat imiquimod (IMQ)-induced psoriasis in BALB/C mice and examined the underlying mechanisms. The outcomes demonstrated that IRh reduced epidermal hyperplasia, lowered PASI scores, and improved histopathological psoriasiform lesions in IMQ-induced mice. IRh attenuated the accumulation of malondialdehyde (MDA), and also reversed the reduction caused by IMQ of superoxide dismutase (SOD) and catalase (CAT) in skin tissues. Additionally, IRh effectively inhibited IMQ's ability to increase proinflammatory cytokines such as TNF-α, IL-6, IL-17A, and transcription factor NF-κB. Furthermore, IRh significantly reduced the percentage of Th1 and Th17 in the spleens of mice treated with IMQ and suppressed the maturation of splenic dendritic cells. Overall, our research suggests that IRh protects against oxidative stress and inflammation in the pathogenesis of psoriasis, with potential for the development of new and potent medication for the treatment of psoriasis.
Collapse
|
72
|
Vu A, Ulschmid C, Gordon KB. Anti-IL 23 biologics for the treatment of plaque psoriasis. Expert Opin Biol Ther 2022; 22:1489-1502. [PMID: 36243011 DOI: 10.1080/14712598.2022.2132143] [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] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Psoriasis is a chronic inflammatory disease that can drastically affect a patient's quality-of-life and is associated with multiple comorbid conditions. The most common form of psoriasis is plaque psoriasis, commonly presenting as sharply demarcated, erythematous plaques with overlying silvery scale on the trunk, extensor surfaces, limbs, and scalp. Although initially limited to oral therapies, the choices in systemic therapies for moderate-to-severe plaque psoriasis have evolved with biologic immunotherapies being the main focus. AREAS COVERED In this review, we describe the IL-23/Th17 axis and IL-23 inhibitors as targets for a growing family of biologics. This family includes the FDA-approved medications ustekinumab, guselkumab, tildrakizumab, and risankizumab. We will review the safety and efficacy of these medications throughout various Phase 1,2, and 3, trials for moderate-to-severe psoriasis. A literature search of PubMed was utilized for the following terms: 'psoriasis and IL-23,' 'ustekinumab,' 'guselkumab,' 'tildrakizumab,' and 'risankizumab.' We also searched for clinical trials involving IL-23 inhibitors registered at ClinicalTrials.gov. EXPERT OPINION Anti-IL 23 therapy, especially anti-p19 monoclonal antibodies, should be considered first-line therapy for moderate-to-severe plaque psoriasis due to their efficacy and relative safety. More research is required to expand the scope of anti-p19 therapy to pediatric populations and additional indications such as psoriatic arthritis.
Collapse
Affiliation(s)
- Alan Vu
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Caden Ulschmid
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kenneth B Gordon
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
73
|
Xu J, Chen H, Qian H, Wang F, Xu Y. Advances in the modulation of ROS and transdermal administration for anti-psoriatic nanotherapies. J Nanobiotechnology 2022; 20:448. [PMID: 36242051 PMCID: PMC9569062 DOI: 10.1186/s12951-022-01651-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: 08/11/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
Reactive oxygen species (ROS) at supraphysiological concentration have a determinate role in contributing to immuno-metabolic disorders in the epithelial immune microenvironment (EIME) of psoriatic lesions. With an exclusive focus on the gene-oxidative stress environment interaction in the EIME, a comprehensive strategy based on ROS-regulating nanomedicines is greatly anticipated to become the mainstay of anti-psoriasis treatment. This potential therapeutic modality could inhibit the acceleration of psoriasis via remodeling the redox equilibrium and reshaping the EIME. Herein, we present a marked overview of the current progress in the pathomechanisms of psoriasis, with particular concerns on the potential pathogenic role of ROS, which significantly dysregulates redox metabolism of keratinocytes (KCs) and skin-resident or -infiltrating cells. Meanwhile, the emergence of versatile nanomaterial-guided evolution for transdermal drug delivery has been attractive for the percutaneous administration of antipsoriatic therapies in recent years. We emphasize the underlying molecular mechanism of ROS-based nanoreactors for improved therapeutic outcomes against psoriasis and summarize up-to-date progress relating to the advantages and limitations of nanotherapeutic application for transdermal administration, as well as update an insight into potential future directions for nanotherapies in ROS-related skin diseases.
Collapse
Affiliation(s)
- Jiangmei Xu
- Department of Dermatovenerology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China.,Department of Dermatology and Rheumatology Immunology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Hao Chen
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui, People's Republic of China.
| | - Fei Wang
- Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China.
| | - Yunsheng Xu
- Department of Dermatovenerology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
74
|
Shmarov F, Smith GR, Weatherhead SC, Reynolds NJ, Zuliani P. Individualised computational modelling of immune mediated disease onset, flare and clearance in psoriasis. PLoS Comput Biol 2022; 18:e1010267. [PMID: 36178923 PMCID: PMC9524682 DOI: 10.1371/journal.pcbi.1010267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/30/2022] [Indexed: 11/18/2022] Open
Abstract
Despite increased understanding about psoriasis pathophysiology, currently there is a lack of predictive computational models. We developed a personalisable ordinary differential equations model of human epidermis and psoriasis that incorporates immune cells and cytokine stimuli to regulate the transition between two stable steady states of clinically healthy (non-lesional) and disease (lesional psoriasis, plaque) skin. In line with experimental data, an immune stimulus initiated transition from healthy skin to psoriasis and apoptosis of immune and epidermal cells induced by UVB phototherapy returned the epidermis back to the healthy state. Notably, our model was able to distinguish disease flares. The flexibility of our model permitted the development of a patient-specific “UVB sensitivity” parameter that reflected subject-specific sensitivity to apoptosis and enabled simulation of individual patients’ clinical response trajectory. In a prospective clinical study of 94 patients, serial individual UVB doses and clinical response (Psoriasis Area Severity Index) values collected over the first three weeks of UVB therapy informed estimation of the “UVB sensitivity” parameter and the prediction of individual patient outcome at the end of phototherapy. An important advance of our model is its potential for direct clinical application through early assessment of response to UVB therapy, and for individualised optimisation of phototherapy regimes to improve clinical outcome. Additionally by incorporating the complex interaction of immune cells and epidermal keratinocytes, our model provides a basis to study and predict outcomes to biologic therapies in psoriasis. We present a new computer model for psoriasis, an immune-mediated disabling skin disease which presents with red, raised scaly plaques that can appear over the whole body. Psoriasis affects millions of people in the UK alone and causes significant impairment to quality of life, and currently has no cure. Only a few treatments (including UVB phototherapy) can induce temporary remission. Despite our increased understanding about psoriasis, treatments are still given on a ‘trial and error’ basis and there are no reliable computer models that can a) elucidate the mechanisms behind psoriasis onset or flare and b) predict a patient’s response to a course of treatment (e.g., phototherapy) and the likelihood of inducing a period of remission. Our computer model addresses both these needs. First, it explicitly describes the interaction between the immune system and skin cells. Second, our model captures response to therapy at the individual patient level and enables personalised prediction of clinical outcomes. Notably, our model also supports prediction of amending individual UVB phototherapy regimes based on the patient’s initial response that include for example personalised delivery schedules (i.e., 3x weekly vs. 5x weekly phototherapy). Therefore, our work is a crucial step towards precision medicine for psoriasis treatment.
Collapse
Affiliation(s)
- Fedor Shmarov
- School of Computing, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Computer Science, University of Manchester, Manchester, United Kingdom
| | - Graham R. Smith
- Bioinformatics Support Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Sophie C. Weatherhead
- Department of Dermatology, Royal Victoria Infirmary, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Nick J. Reynolds
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- * E-mail: (NJR); (PZ)
| | - Paolo Zuliani
- Department of Computer Science, University of Manchester, Manchester, United Kingdom
- * E-mail: (NJR); (PZ)
| |
Collapse
|
75
|
Bakulev AL, Kruglova LS. Efficacy and safety of treatment of moderate and severe psoriasis with the interleukin 23 inhibitor risankizumab. VESTNIK DERMATOLOGII I VENEROLOGII 2022. [DOI: 10.25208/vdv1349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
A review of the literature on the use of the genetically engineered biological drug Skyrizi (INN: risankizumab) for the treatment of psoriasis in patients is presented. The problem of high dissatisfaction of patients with the available means (methods) of therapy for this dermatosis is discussed.
Inhibition of regulatory IL-23 seems to be a very promising direction of therapy, which makes it possible to block the immunopathogenesis of the disease.
The IL-23 inhibitor risankizumab is a highly effective GEBA for the treatment of moderate to severe psoriasis in patients. The use of this drug is accompanied by a very rapid and stable therapeutic response in the form of clear or almost clear skin. Long-term use of risankizumab demonstrates the maintenance of a stable therapeutic response without a tendency to decrease.
Risankizumab has a favorable safety profile with a low risk of infections and malignancies. At the same time, such adverse events as the development of tuberculosis or inflammatory bowel disease are not typical for therapy with these GIBDs.
Collapse
|
76
|
Gene Profiling of a 3D Psoriatic Skin Model Enriched in T Cells: Downregulation of PTPRM Promotes Keratinocyte Proliferation through Excessive ERK1/2 Signaling. Cells 2022; 11:cells11182904. [PMID: 36139479 PMCID: PMC9497242 DOI: 10.3390/cells11182904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 12/03/2022] Open
Abstract
Psoriasis is a complex, immune-mediated skin disease involving a wide range of epithelial and immune cells. The underlying mechanisms that govern the epidermal defects and immunological dysfunction observed in this condition remain largely unknown. In recent years, the emergence of new, more sophisticated models has allowed the evolution of our knowledge of the pathogenesis of psoriasis. The development of psoriatic skin biomaterials that more closely mimic native psoriatic skin provides advanced preclinical models that will prove relevant in predicting clinical outcomes. In this study, we used a tissue-engineered, two-layered (dermis and epidermis) human skin substitute enriched in T cells as a biomaterial to study both the cellular and molecular mechanisms involved in psoriasis’ pathogenesis. Gene profiling on microarrays revealed significant changes in the profile of genes expressed by the psoriatic skin substitutes compared with the healthy ones. Two genes, namely, PTPRM and NELL2, whose products influence the ERK1/2 signaling pathway have been identified as being deregulated in psoriatic substitutes. Deregulation of these genes supports excessive activation of the ERK1/2 pathway in psoriatic skin substitutes. Most importantly, electrophoresis mobility shift assays provided evidence that the DNA-binding properties of two downstream nuclear targets of ERK1/2, both the NF-κB and Sp1 transcription factors, are increased under psoriatic conditions. Moreover, the results obtained with the inhibition of RSK, a downstream effector of ERK1/2, supported the therapeutic potential of inhibiting this signaling pathway for psoriasis treatment. In conclusion, this two-layered human psoriatic skin substitute enriched in T cells may prove particularly useful in deciphering the mechanistic details of psoriatic pathogenesis and provide a relevant biomaterial for the study of potential therapeutic targets.
Collapse
|
77
|
Ferreri A, Lang V, Kaufmann R, Buerger C. mTORC1 Activity in Psoriatic Lesions Is Mediated by Aberrant Regulation through the Tuberous Sclerosis Complex. Cells 2022; 11:cells11182847. [PMID: 36139422 PMCID: PMC9497233 DOI: 10.3390/cells11182847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
In the basal, proliferative layer of healthy skin, the mTOR complex 1 (mTORC1) is activated, thus regulating proliferation while preventing differentiation. When cells leave the proliferative, basal compartment, mTORC1 signaling is turned off, which allows differentiation. Under inflammatory conditions, this switch is hijacked by cytokines and prevents proper differentiation. It is currently unknown how mTORC1 is regulated to mediate these effects on keratinocyte differentiation. In other tissues, mTORC1 activity is controlled through various pathways via the tuberous sclerosis complex (TSC). Thus, we investigated whether the TS complex is regulated by proinflammatory cytokines and contributes to the pathogenesis of psoriasis. TNF-α as well as IL-1β induced the phosphorylation of TSC2, especially on S939 via the PI3-K/AKT and MAPK pathway. Surprisingly, increased TSC2 phosphorylation could not be detected in psoriasis patients. Instead, TSC2 was strongly downregulated in lesional psoriatic skin compared to non-lesional skin of the same patients or healthy skin. In vitro inflammatory cytokines induced dissociation of TSC2 from the lysosome, followed by destabilization of the TS complex and degradation. Thus, we assume that in psoriasis, inflammatory cytokines induce strong TSC2 phosphorylation, which in turn leads to its degradation. Consequently, chronic mTORC1 activity impairs ordered keratinocyte differentiation and contributes to the phenotypical changes seen in the psoriatic epidermis.
Collapse
|
78
|
Xi L, Han Y, Liu C, Liu Y, Wang Z, Wang R, Zheng Y. Sonodynamic therapy by phase-transition nanodroplets for reducing epidermal hyperplasia in psoriasis. J Control Release 2022; 350:435-447. [PMID: 36030991 DOI: 10.1016/j.jconrel.2022.08.038] [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: 03/31/2022] [Revised: 07/31/2022] [Accepted: 08/21/2022] [Indexed: 11/29/2022]
Abstract
The cross-talk between hyperproliferative keratinocytes and activated immune cells is responsible for the progression of psoriasis. The strategy to alleviate psoriasis through inhibiting the abnormal proliferation of keratinocytes remains challenging due to limited therapeutic effects and low skin penetration of drugs. Herein we designed an ultrasound-triggered phase-transition nanodroplet that could produce cavitation to enhance skin penetration and effectively generate reactive oxygen species (ROS) to induce keratinocyte apoptosis for psoriasis treatment. After ultrasound stimulation, the perfluoro-n-pentane (PFP) liquid core of the nanodroplets vaporized, and the Haematoporphyrin monomethyl ether (HMME) encapsulated in the nanodroplets generated plenty of intracellular ROS which caused the apoptosis of HaCat cells through inducing mitochondrial dysfunction. In addition, the blank nanodroplets successfully inhibited the secretion of IL-6 and TNF-α from macrophages and dendritic cells in vitro due to the anti-inflammatory effect of POPG. For the skin penetration test, the phase-transition nanodroplets could effectively accumulate in the epidermis of the skin and generate intracellular ROS. The in-vivo anti-psoriasis experiment demonstrated that the phase-transition nanodroplets relieved the symptoms of psoriasis lesion and inhibited epidermal hyperplasia through induction of cell apoptosis under ultrasound irritation. Meanwhile, the inflammatory cytokines in the skin lesion almost decreased to the normal baseline level after SDT. Collectively, this study demonstrated a new strategy to inhibit keratinocyte hyperproliferation for psoriasis management based on sonodynamic responded nanodroplets.
Collapse
Affiliation(s)
- Long Xi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Yunfeng Han
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Chang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Yihan Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, CA 92093, USA
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China.
| |
Collapse
|
79
|
Hari G, Kishore A, Karkala SRP. Treatments for psoriasis: A journey from classical to advanced therapies. How far have we reached? Eur J Pharmacol 2022; 929:175147. [PMID: 35820531 DOI: 10.1016/j.ejphar.2022.175147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/29/2022] [Accepted: 07/06/2022] [Indexed: 11/03/2022]
Abstract
Psoriasis is considered an autoimmune, inflammatory disorder with a genetic basis. The underlying aetiology is yet unclear. Evidence suggests the congregation of immune cells and their secreted inflammatory cytokines, leukocytes, and other inflammation-promoting factors in large amounts within the epidermal layers of the skin, driving an inflammatory milieu. Although psoriasis is not a fatal condition, patients experience severe pain and suffering. It has a debilitating effect on the physiological and psychological state of the patient. Its distinguishing features are inflammation, formation of plaques on the skin and hyperproliferation of keratinocytes. Therapeutic strategies for treating psoriasis witnessed a radical improvement from traditional therapies to the approval of specific therapies like biologics and small molecules. The emerging evidence about new pharmacological targets and mechanisms in psoriasis has widened the scope for expanding therapeutic strategies. Our review discusses the existing treatments for plaque psoriasis and updates on therapies based on novel pharmacological targets in clinical development.
Collapse
Affiliation(s)
- Gangadhar Hari
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sreedhara Ranganath Pai Karkala
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
80
|
Belpaire A, van Geel N, Speeckaert R. From IL-17 to IFN-γ in inflammatory skin disorders: Is transdifferentiation a potential treatment target? Front Immunol 2022; 13:932265. [PMID: 35967358 PMCID: PMC9367984 DOI: 10.3389/fimmu.2022.932265] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The targeted inhibition of effector cytokines such as interleukin 17 (IL-17) in psoriasis and IL-13 in atopic dermatitis offers impressive efficacy with a favorable side effect profile. In contrast, the downregulation of interferon gamma (IFN-γ) in T helper (Th) 1-dominant skin disorders may lead to more adverse events, given the crucial role of IFN-γ in antiviral and antitumoral immunity. Modulating Th17 and Th2 cell differentiation is performed by blocking IL-23 and IL-4, respectively, whereas anti-IL-12 antibodies are only moderately effective in downregulating Th1 lymphocyte differentiation. Therefore, a targeted approach of IFN-γ-driven disorders remains challenging. Recent literature suggests that certain pathogenic Th17 cell subsets with Th1 characteristics, such as CD4+CD161+CCR6+CXCR3+IL-17+IFN-y+ (Th17.1) and CD4+CD161+CCR6+CXCR3+IL-17-IFN-y+ (exTh17), are important contributors in Th1-mediated autoimmunity. Differentiation to a Th17.1 or exTh17 profile results in the upregulation of IFN-y. Remarkably, these pathogenic Th17 cell subsets are resistant to glucocorticoid therapy and the dampening effect of regulatory T cells (Treg). The identification of Th17.1/exTh17 cells in auto-immune disorders may explain the frequent treatment failure of conventional immunosuppressants. In this review, we summarize the current evidence regarding the cellular plasticity of Th17 cells in inflammatory skin disorders. A deeper understanding of this phenomenon may lead to better insights into the pathogenesis of various skin diseases and the discovery of a potential new treatment target.
Collapse
|
81
|
Tang X. The risk of organ-based comorbidities in psoriasis: a systematic review and meta-analysis. An Bras Dermatol 2022; 97:612-623. [PMID: 35850940 PMCID: PMC9453528 DOI: 10.1016/j.abd.2021.10.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023] Open
Abstract
Background The close relationship between psoriasis and concomitant diseases is widely accepted. However, a comprehensive analysis of organ-based comorbidities in psoriasis is still lacking. Objective The authors aimed to present the risk of organ-based comorbidities in psoriasis by comparing the general population. Methods The authors retrieved a search of Pubmed, EMBASE, and Cochrane databases for studies reporting organ-based comorbidities in psoriasis versus the general population. Observational studies that met the following criteria were assessed: 1) Psoriasis diagnosis; 2) Cardiovascular or kidney or liver or respiratory or cerebrovascular outcomes; 3) Comparison group of individuals without psoriasis. Pooled Relative Risks (pRRs) and 95% Confidence Intervals (CIs) were calculated by using the random-effect model. Results Fifteen observational studies with 216,348 psoriatic patients and 9,896,962 individuals from the general population were included. Psoriasis showed a greater risk of organ-based comorbidities. Compared to the general population, pRR for all organ-based comorbidities was 1.20 (95% CI 1.11‒1.31) in psoriasis, and pRR was lower in mild 0.61 (95% CI 0.46‒0.81) than in moderate/severe patients. pRR was 1.20 (95% CI 1.11‒1.30) for cardiovascular, 1.56 (95% CI 1.20‒2.04), and 1.75 (95% CI 1.33‒2.29) for cerebrovascular and liver diseases, respectively. pRR for coexisting renal and cardiovascular events was 1.09 (95% CI 1.01‒1.18). pRR for coexisting renal and cerebrovascular events was 1.28 (95% CI 0.99‒1.66). pRR for coexisting renal and liver diseases was 1.46 (95% CI 1.10‒1.94). pRR for coexisting cardiovascular and liver diseases was 1.41 (95% CI 1.11‒1.80). Study limitations There is heterogeneity. Conclusion Psoriasis has a higher risk of single and multiple organ-based comorbidities than the general population. The present study will further improve attention to psoriasis as a systemic inflammatory disease.
Collapse
Affiliation(s)
- Xuemei Tang
- Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
82
|
Babaie F, Omraninava M, Gorabi AM, Khosrojerdi A, Aslani S, Yazdchi A, Torkamandi S, Mikaeili H, Sathyapalan T, Sahebkar A. Etiopathogenesis of Psoriasis from Genetic Perspective: An updated Review. Curr Genomics 2022; 23:163-174. [PMID: 36777004 PMCID: PMC9878828 DOI: 10.2174/1389202923666220527111037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/11/2022] [Accepted: 04/12/2022] [Indexed: 11/22/2022] Open
Abstract
Psoriasis is an organ-specific autoimmune disease characterized by the aberrant proliferation and differentiation of keratinocytes, leading to skin lesions. Abnormal immune responses mediated by T cells and dendritic cells and increased production of inflammatory cytokines have been suggested as underlying mechanisms in the pathogenesis of psoriasis. Emerging evidence suggests that there is a heritable basis for psoriatic disorders. Moreover, numerous gene variations have been associated with the disease risk, particularly those in innate and adaptive immune responses and antigen presentation pathways. Herein, this article discusses the genetic implications of psoriatic diseases' etiopathogenesis to develop novel investigative and management options.
Collapse
Affiliation(s)
- Farhad Babaie
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Melodi Omraninava
- Department of Infectious Diseases, Faculty of Medical Sciences, Sari Branch, Islamic Azad University, Sari, Iran
| | - Armita Mahdavi Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Arezou Khosrojerdi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Aslani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arsalan Yazdchi
- Student Research Committee, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Torkamandi
- Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Haleh Mikaeili
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Western Australia, Mashhad, Iran
| |
Collapse
|
83
|
Zhou HF, Wang FX, Sun F, Liu X, Rong SJ, Luo JH, Yue TT, Xiao J, Yang CL, Lu WY, Luo X, Zhou Q, Zhu H, Yang P, Xiong F, Yu QL, Zhang S, Wang CY. Aloperine Ameliorates IMQ-Induced Psoriasis by Attenuating Th17 Differentiation and Facilitating Their Conversion to Treg. Front Pharmacol 2022; 13:778755. [PMID: 35721119 PMCID: PMC9198605 DOI: 10.3389/fphar.2022.778755] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 04/11/2022] [Indexed: 11/26/2022] Open
Abstract
Aloperine is an anti-inflammatory compound isolated from the Chinese herb Sophora alopecuroides L. Previously, our group has reported that the generation of induced Treg was promoted by aloperine treatment in a mouse colitis model. However, the effect of aloperine on effector T cell subsets remains unclear. We therefore carefully examined the effect of aloperine on the differentiation of major subsets of T helper cells. Based on our results, psoriasis, a Th17 dominant skin disease, is selected to explore the potential therapeutic effect of aloperine in vivo. Herein, we demonstrated that topical application of aloperine suppressed epidermal proliferation, erythema, and infiltration of inflammatory cells in skin lesions. Mechanistic studies revealed that aloperine suppressed the differentiation of Th17 cells directly through inhibiting the phosphorylation of STAT3 or indirectly through impairing the secretion of Th17-promoting cytokines by dendritic cells. Moreover, aloperine enhanced the conversion of Th17 into Treg via altering the pSTAT3/pSTAT5 ratio. Collectively, our study supported that aloperine possesses the capacity to affect Th17 differentiation and modulates Th17/Treg balance, thereby alleviating imiquimod (IMQ)-induced psoriasis in mice.
Collapse
Affiliation(s)
- Hai-Feng Zhou
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Fa-Xi Wang
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Fei Sun
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xin Liu
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Shan-Jie Rong
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Jia-Hui Luo
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Tian-Tian Yue
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Jun Xiao
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China.,Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Chun-Liang Yang
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Wan-Ying Lu
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xi Luo
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Qing Zhou
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - He Zhu
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Ping Yang
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Fei Xiong
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Qi-Lin Yu
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Shu Zhang
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Cong-Yi Wang
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| |
Collapse
|
84
|
Jiang R, Xu J, Zhang Y, Liu J, Wang Y, Chen M, Chen X, Yin M. Ligustrazine alleviates psoriasis-like inflammation through inhibiting TRAF6/c-JUN/NFκB signaling pathway in keratinocyte. Biomed Pharmacother 2022; 150:113010. [PMID: 35468584 DOI: 10.1016/j.biopha.2022.113010] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 11/02/2022] Open
Abstract
Ligusticum chuanxiong Hort (Ligusticum; Apiaceae) (accepted name, Ligusticum striatum DC, on "The Plant List" for the latest version) is a Chinese herbal medicine (CHM) which mainly distributed in Sichuan Basin, China. Chuanxiong is the dried rhizome of Ligusticum chuanxiong Hort. Ligustrazine, also known as tetramethylpyrazine (TMP), is a main active fraction of chuanxiong. The aim of this study was to clarify the underlying mechanisms by which TMP protect against psoriasis-like inflammation in keratinocytes. Here, we demonstrated that TMP alleviated the severity and PASI scores of IMQ-induced psoriasis-like skin lesion in vivo. For the histopathology level, TMP inhibited the over-proliferation of keratinocytes in the epidermis and the substantial immune cells influx in dermis. For the mechanism of the ability of TMP on regulating inflammation, we confirmed that TMP regulate the TRAF6/c-JUN/NFκB signaling pathway through analyzing the proteomics profiling and verifying the expression of TRAF6, pho-c-Jun, pho-NFκB, so that the downstream psoriasis-relevant genes transcribed by c-JUN or NFκB were down-regulated. Furthermore, we predicted TRAF6 as the potential binding point of TMP. Accordingly, our study demonstrated that TMP regulated psoriasis-like inflammation through inhibiting TRAF6/c-JUN/NFκB signaling pathway in keratinocytes, which potentially provides evidence of the mechanism of TMP in the treatment and prevention of psoriasis.
Collapse
Affiliation(s)
- Rundong Jiang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jiaqi Xu
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yuezhong Zhang
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jiachen Liu
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yutong Wang
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mingliang Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Xiang Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Mingzhu Yin
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
85
|
TRPV1 + sensory nerves modulate corneal inflammation after epithelial abrasion via RAMP1 and SSTR5 signaling. Mucosal Immunol 2022; 15:867-881. [PMID: 35680973 DOI: 10.1038/s41385-022-00533-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 04/25/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023]
Abstract
Timely initiation and termination of inflammatory response after corneal epithelial abrasion is critical for the recovery of vision. The cornea is innervated with rich sensory nerves with highly dense TRPV1 nociceptors. However, the roles of TRPV1+ sensory neurons in corneal inflammation after epithelial abrasion are not completely understood. Here, we found that depletion of TRPV1+ sensory nerves using resiniferatoxin (RTX) and blockade of TRPV1 using AMG-517 delayed corneal wound closure and enhanced the infiltration of neutrophils and γδ T cells to the wounded cornea after epithelial abrasion. Furthermore, depletion of TRPV1+ sensory nerves increased the number and TNF-α production of corneal CCR2+ macrophages and decreased the number of corneal CCR2- macrophages and IL-10 production. In addition, the TRPV1+ sensory nerves inhibited the recruitment of neutrophils and γδ T cells to the cornea via RAMP1 and SSTR5 signaling, decreased the responses of CCR2+ macrophages via RAMP1 signaling, and increased the responses of CCR2- macrophages via SSTR5 signaling. Collectively, our results suggest that the TRPV1+ sensory nerves suppress inflammation to support corneal wound healing via RAMP1 and SSTR5 signaling, revealing potential approaches for improving defective corneal wound healing in patients with sensory neuropathy.
Collapse
|
86
|
Gut–Skin Axis: Unravelling the Connection between the Gut Microbiome and Psoriasis. Biomedicines 2022; 10:biomedicines10051037. [PMID: 35625774 PMCID: PMC9138548 DOI: 10.3390/biomedicines10051037] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/23/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022] Open
Abstract
Evidence has shown that gut microbiome plays a role in modulating the development of diseases beyond the gastrointestinal tract, including skin disorders such as psoriasis. The gut–skin axis refers to the bidirectional relationship between the gut microbiome and skin health. This is regulated through several mechanisms such as inflammatory mediators and the immune system. Dysregulation of microbiota has been seen in numerous inflammatory skin conditions such as atopic dermatitis, rosacea, and psoriasis. Understanding how gut microbiome are involved in regulating skin health may lead to development of novel therapies for these skin disorders through microbiome modulation, in particularly psoriasis. In this review, we will compare the microbiota between psoriasis patients and healthy control, explain the concept of gut–skin axis and the effects of gut dysbiosis on skin physiology. We will also review the current evidence on modulating gut microbiome using probiotics in psoriasis.
Collapse
|
87
|
Xu J, Chen H, Chu Z, Li Z, Chen B, Sun J, Lai W, Ma Y, He Y, Qian H, Wang F, Xu Y. A multifunctional composite hydrogel as an intrinsic and extrinsic coregulator for enhanced therapeutic efficacy for psoriasis. J Nanobiotechnology 2022; 20:155. [PMID: 35331238 PMCID: PMC8943972 DOI: 10.1186/s12951-022-01368-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/10/2022] [Indexed: 12/20/2022] Open
Abstract
Background Psoriasis is a chronic relapsing immunological skin disease characterized by multiple cross-talk inflammatory circuits which are relevantly associated with abnormal cross-reactivity between immune cells and keratinocytes (KCs). It may be inadequate to eradicate complicated pathogenesis only via single-mode therapy. To provide optimal combinatory therapeutics, a nanocomposite-based hydrogel was constructed by loading methotrexate (MTX) into ZnO/Ag to realize combined multiple target therapy of psoriasis. Results In this composite hydrogel, ZnO hybrid mesoporous microspheres were utilized both as drug carriers and reactive oxygen species (ROS)-scavenging nanoparticles. A proper amount of Ag nanoparticle-anchored ZnO nanoparticles (ZnO/Ag) was functionalized with inherent immunoregulatory property. The experiments showed that ZnO/Ag nanoparticles could exhibit a self-therapeutic effect that was attributed to reducing innate cytokine profiles by inactivating p65 in proinflammatory macrophages and abrogating secretion of adaptive cytokines in KCs by downregulating ROS-mediated STAT3-cyclin D1 signaling. A preferable antipsoriatic efficacy was achieved via topical administration of this hydrogel on the imiquimod (IMQ)-induced psoriasis mice model, demonstrating the superior transdermal delivery and combined enhancement of therapeutic efficacy caused by intrinsic nanoparticles and extrinsic MTX. Conclusion This composite hydrogel could serve as a multifunctional, nonirritating, noninvasive and effective transcutaneous nanoagent against psoriasis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01368-y.
Collapse
Affiliation(s)
- Jiangmei Xu
- Department of Dermatovenerology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, People's Republic of China
| | - Hao Chen
- School of Basic Medical Sciences, School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, People's Republic of China
| | - Zhaoyou Chu
- School of Basic Medical Sciences, School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, People's Republic of China
| | - Zhu Li
- Department of Dermatovenerology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, People's Republic of China
| | - Benjin Chen
- School of Basic Medical Sciences, School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, People's Republic of China
| | - Jianan Sun
- School of Basic Medical Sciences, School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, People's Republic of China
| | - Wei Lai
- Department of Dermatovenerology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yan Ma
- School of Basic Medical Sciences, School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, People's Republic of China
| | - Yulong He
- Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, People's Republic of China
| | - Haisheng Qian
- School of Basic Medical Sciences, School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Hefei, Anhui, People's Republic of China.
| | - Fei Wang
- Center for Digestive Disease, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, People's Republic of China.
| | - Yunsheng Xu
- Department of Dermatovenerology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
88
|
Wi D, Wilson A, Satgé F, Murrell DF. Osteoporosis and Psoriasis: A Literature Review. Clin Exp Dermatol 2022; 47:1438-1445. [PMID: 35279869 PMCID: PMC9543251 DOI: 10.1111/ced.15174] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/09/2022] [Indexed: 12/01/2022]
Abstract
Psoriasis is a chronic inflammatory skin disease with complex comorbidities. Recent evidence has revealed how the inflammatory nature of psoriasis affects bone mineral density and may lead to osteoporosis. This review outlines the current understanding and advances on the association between psoriasis and osteoporosis. The current literature suggests an increased risk of osteopenia and osteoporosis in patients with extensive and chronic psoriasis, compounded by other lifestyle and genetic factors. It suggests that prophylactic measures such as vitamin D supplementation and increasing weight‐bearing exercises can help, but in patients with extensive psoriasis, prolonged systemic inflammation may require long‐term management. Although there have been many short‐term RCTs on the efficacy and safety of biologics in psoriasis, clinical studies looking at the long‐term effects of biologics, such as whether they might improve bone mineral density in these patients with psoriasis are yet to be conducted.
Collapse
Affiliation(s)
- Dohyen Wi
- Faculty of Medicine University of New South Wales Sydney NSW Australia
| | - Anna Wilson
- Faculty of Medicine University of New South Wales Sydney NSW Australia
- Department of Dermatology, St George Hospital Sydney NSW Australia
| | - Fanny Satgé
- Department of Dermatology, St George Hospital Sydney NSW Australia
- Paris‐East Créteil Val de Marne University Paris France
| | - Dédée F. Murrell
- Faculty of Medicine University of New South Wales Sydney NSW Australia
- Department of Dermatology, St George Hospital Sydney NSW Australia
| |
Collapse
|
89
|
Bolt JW, van Kuijk AW, Teunissen MBM, van der Coelen D, Aarrass S, Gerlag DM, Tak PP, van de Sande MG, Lebre MC, van Baarsen LGM. Impact of Adalimumab Treatment on Interleukin-17 and Interleukin-17 Receptor Expression in Skin and Synovium of Psoriatic Arthritis Patients with Mild Psoriasis. Biomedicines 2022; 10:biomedicines10020324. [PMID: 35203534 PMCID: PMC8869729 DOI: 10.3390/biomedicines10020324] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Interleukin (IL)-17 and tumor necrosis factor-alpha (TNF)-α are key players in psoriatic arthritis (PsA) pathogenesis. While both cytokines can be therapeutically targeted with beneficial clinical outcome, it is unclear whether inhibiting one cytokine will affect the other at sites of inflammation. If both act independently, this might provide a rationale for dual or combined inhibition of both cytokines. Here, we evaluated the effect of TNF blockade in PsA patients on IL-17 levels in both skin and synovial tissue biopsies. PsA patients with mild psoriatic skin lesions were randomized to receive either adalimumab or placebo for four weeks. Synovial and skin biopsies were obtained at weeks zero and four. Skin from healthy donors (HDs) was used for comparison. Expression of IL-17A, IL-17F, IL-17RA and IL-17RC was assessed by immunohistochemistry and analyzed with digital image analysis. We found relatively low levels of IL-17 and its receptors in the skin of PsA patients compared to HD, and only IL-17F in the dermis of lesional psoriatic skin was significantly higher compared to HD skin (p = 0.0002). Histologically IL-17A, IL-17F, IL-17RA and IL-17RC in skin and synovial tissue were not downregulated by adalimumab treatment. Thus, in this cohort of PsA patients with mild psoriasis, TNF blockade did not affect the protein levels of IL-17 cytokines and its receptors in skin and synovium, despite reduced cellular inflammation and improved clinical outcome for joint involvement.
Collapse
Affiliation(s)
- Janne W. Bolt
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
| | - Arno W. van Kuijk
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
- Department of Rheumatology, Reade, 1056 AB Amsterdam, The Netherlands
| | - Marcel B. M. Teunissen
- Department of Dermatology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Dennis van der Coelen
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
| | - Saïda Aarrass
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
| | - Daniëlle M. Gerlag
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
| | - Paul P. Tak
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
- Candel Therapeutics, Needham, MA 02494, USA
- Internal Medicine, Cambridge University, Cambridge CB2 1TN, UK
| | - Marleen G. van de Sande
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
| | - Maria C. Lebre
- The Netherlands Cancer Institute, Division of Pharmacology, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands;
| | - Lisa G. M. van Baarsen
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (J.W.B.); (D.v.d.C.); (S.A.); (D.M.G.); (P.P.T.); (M.G.v.d.S.)
- Department of Rheumatology, Amsterdam Rheumatology & Immunology Center (ARC)-Reade, 1040 HG Amsterdam, The Netherlands;
- Correspondence:
| |
Collapse
|
90
|
Zhou X, Chen Y, Cui L, Shi Y, Guo C. Advances in the pathogenesis of psoriasis: from keratinocyte perspective. Cell Death Dis 2022; 13:81. [PMID: 35075118 PMCID: PMC8786887 DOI: 10.1038/s41419-022-04523-3] [Citation(s) in RCA: 282] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 12/14/2021] [Accepted: 01/11/2022] [Indexed: 02/08/2023]
Abstract
Psoriasis is a complex long-lasting inflammatory skin disease with high prevalence and associated comorbidity. It is characterized by epidermal hyperplasia and dermal infiltration of immune cells. Here, we review the role of keratinocytes in the pathogenesis of psoriasis, focusing on factors relevant to genetics, cytokines and receptors, metabolism, cell signaling, transcription factors, non-coding RNAs, antimicrobial peptides, and proteins with other different functions. The critical role of keratinocytes in initiating and maintaining the inflammatory state suggests the great significance of targeting keratinocytes for the treatment of psoriasis.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China
| | - Youdong Chen
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China
| | - Lian Cui
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China
| | - Yuling Shi
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China.
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China.
| | - Chunyuan Guo
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China.
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China.
| |
Collapse
|
91
|
Murzina E, Dosenko V, Drevytska T, Litus O, Bardova K, Vozianova S. Relationship between mir-126 expression in children with psoriasis, disease progression and therapeutic response. J Med Life 2022; 14:667-675. [PMID: 35027969 PMCID: PMC8742889 DOI: 10.25122/jml-2021-0115] [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: 06/09/2021] [Accepted: 09/09/2021] [Indexed: 11/17/2022] Open
Abstract
This study aimed to investigate the expression level of miR-126 in children with psoriasis in the epidermis affected by psoriasis and intact buccal epithelium, establish the impact on the characteristics of the course of psoriasis and the results of therapy in children with psoriasis of initial expression levels of miR-126. miR-126 expression levels in psoriatic keratinocytes and buccal epithelium were determined in 54 children with psoriasis on the severity of psoriasis, treatment efficacy. miR-126 levels in the buccal epithelium in children with psoriasis were reduced compared to healthy children (AUC=0.776±0.048, p<0.001). There were no discrepancies between miR-126 expression levels in psoriatic keratinocytes and buccal epithelium (p=0.097). There are statistically significant discrepancies between miR-126 expression levels in the psoriatic epidermis depending on the clinical form of psoriasis (AUC=0.637±0.056; p=0.014) and severity according to BSA (AUC=0.634±0.063; p=0.034). Depending on the miR-126 level in the buccal epithelium, the response to treatment (PASI<75) in children with high miR-126 is worse than in children with expected miR-126 levels (OR 2.79; 95%; CI: 1.19 - 6.51). Treatment failures were observed in children with high levels of miR-126 in the buccal epithelium compared to miR-126 in the psoriatic epidermis: children aged 12/13 to 17 years (OR 2.44; 95% CI: 1.02 - 5.85), children with PGA=4 (OR 3.16; 95% CI: 1.34 - 7.43). The location and level of miR-126 expression affects the course of psoriasis and the outcome of treatment. High levels of miR-126 in psoriatic keratinocytes lead to manifestations of plaque psoriasis with a course of moderate to severe forms. Initial miR-126 levels in the buccal epithelium in children with psoriasis are a prognostic criterion for response to therapy and can be used as a marker for prescribing systemic treatment.
Collapse
Affiliation(s)
- Elvina Murzina
- Department of Dermatovenereology, Allergology, Clinical and Laboratory Immunology, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
| | - Victor Dosenko
- Department of General and Molecular Pathophysiology of Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine
| | - Tetiana Drevytska
- Department of General and Molecular Pathophysiology of Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine
| | - Oleksandr Litus
- Department of Dermatovenereology, Allergology, Clinical and Laboratory Immunology, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
| | - Kateryna Bardova
- Department of Dermatovenereology, Allergology, Clinical and Laboratory Immunology, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
| | - Svitlana Vozianova
- Department of Dermatovenereology, Allergology, Clinical and Laboratory Immunology, Shupyk National Healthcare University of Ukraine, Kyiv, Ukraine
| |
Collapse
|
92
|
Kelemen E, Ádám É, Sági SM, Göblös A, Kemény L, Bata-Csörgő Z, Széll M, Danis J. Psoriasis-Associated Inflammatory Conditions Induce IL-23 mRNA Expression in Normal Human Epidermal Keratinocytes. Int J Mol Sci 2022; 23:540. [PMID: 35008970 PMCID: PMC8745281 DOI: 10.3390/ijms23010540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 12/14/2022] Open
Abstract
Psoriasis is a multifactorial, chronic inflammatory skin disease, the development of which is affected by both genetic and environmental factors. Cytosolic nucleic acid fragments, recognized as pathogen- and danger-associated molecular patterns, are highly abundant in psoriatic skin. It is known that psoriatic skin exhibits increased levels of IL-23 compared to healthy skin. However, the relationship between free nucleic acid levels and IL-23 expression has not been clarified yet. To examine a molecular mechanism by which nucleic acids potentially modulate IL-23 levels, an in vitro system was developed to investigate the IL-23 mRNA expression of normal human epidermal keratinocytes under psoriasis-like circumstances. This system was established using synthetic nucleic acid analogues (poly(dA:dT) and poly(I:C)). Signaling pathways, receptor involvement and the effect of PRINS, a long non-coding RNA previously identified and characterized by our research group, were analyzed to better understand the regulation of IL-23 in keratinocytes. Our results indicate that free nucleic acids regulate epithelial IL-23 mRNA expression through the TLR3 receptor and specific signaling pathways, thereby, contributing to the development of an inflammatory milieu favorable for the appearance of psoriatic symptoms. A moderate negative correlation was confirmed between the nucleic-acid-induced IL-23 mRNA level and the rate of its decrease upon PRINS overexpression.
Collapse
Affiliation(s)
- Evelyn Kelemen
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (E.K.); (S.M.S.); (L.K.); (Z.B.-C.)
- Department of Medical Genetics, University of Szeged, 6720 Szeged, Hungary;
| | - Éva Ádám
- Department of Medical Genetics, University of Szeged, 6720 Szeged, Hungary;
| | - Stella Márta Sági
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (E.K.); (S.M.S.); (L.K.); (Z.B.-C.)
| | - Anikó Göblös
- MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, 6720 Szeged, Hungary; (A.G.); (J.D.)
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (E.K.); (S.M.S.); (L.K.); (Z.B.-C.)
- MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, 6720 Szeged, Hungary; (A.G.); (J.D.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Zsuzsanna Bata-Csörgő
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (E.K.); (S.M.S.); (L.K.); (Z.B.-C.)
- MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, 6720 Szeged, Hungary; (A.G.); (J.D.)
| | - Márta Széll
- Department of Medical Genetics, University of Szeged, 6720 Szeged, Hungary;
- MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, 6720 Szeged, Hungary; (A.G.); (J.D.)
| | - Judit Danis
- MTA-SZTE Dermatological Research Group, Eötvös Loránd Research Network, 6720 Szeged, Hungary; (A.G.); (J.D.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| |
Collapse
|
93
|
Identification of triazolopyridine derivatives as a new class of AhR agonists and evaluation of anti-psoriasis effect in a mouse model. Eur J Med Chem 2022; 231:114122. [DOI: 10.1016/j.ejmech.2022.114122] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/31/2021] [Accepted: 01/10/2022] [Indexed: 12/24/2022]
|
94
|
Rioux G, Simard M, Morin S, Lorthois I, Guérin SL, Pouliot R. Development of a 3D psoriatic skin model optimized for infiltration of IL-17A producing T cells: Focus on the crosstalk between T cells and psoriatic keratinocytes. Acta Biomater 2021; 136:210-222. [PMID: 34547515 DOI: 10.1016/j.actbio.2021.09.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/20/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022]
Abstract
Psoriasis is a chronic inflammatory skin disease involving several cell types, including T cells, via the IL-23/IL-17 axis. IL-17A acts on the surrounding epithelial cells thus resulting in an inflammatory feedback loop. The development of immunocompetent models that correctly recapitulate the complex phenotype of psoriasis remains challenging, which also includes both the T cell isolation and activation methods. The purpose of this work was to develop an advanced in vitro 3D psoriatic skin model that enables the study of the impact of T cells on psoriatic epithelial cells. To reach that aim, healthy and psoriatic fibroblasts and keratinocytes were used to reproduce this tissue-engineered skin model in which activated T cells, isolated beforehand from human whole blood, have been incorporated. Our study showed that isolation of T cells with the EasySep procedure, followed by activation with PMA/ionomycin, mimicked the psoriatic characteristics in an optimal manner with the production of inflammatory cytokines important in the pathogenesis of psoriasis, as well as increased expression of Ki67, S100A7, elafin and involucrin. This psoriatic model enriched in activated T cells displayed enhanced production of IL-17A, IFN-ƴ, CCL2, CXCL10, IL-1ra, IL-6 and CXCL8 compared with the healthy model and whose increased secretion was maintained over time. In addition, anti-IL17A treatment restored some psoriatic features, including epidermal thickness and basal keratinocytes proliferation, as well as a downregulation of S100A7, elafin and involucrin expression. Altogether, our study demonstrated that this model reflects a proper psoriatic inflammatory environment and is effective for the investigation of epidermal and T cell interaction over time. STATEMENT OF SIGNIFICANCE: The aim of this study was to provide an innovative 3D immunocompetent human psoriatic skin model. To our knowledge, this is the first immunocompetent model that uses skin cells from psoriatic patients to study the impact of IL-17A on pathological cells. Through the use of this model, we demonstrated that the T-cell enriched psoriatic model differs from T-cell enriched healthy model, highlighting efficient crosstalk between pathologic epithelial cells and T cells. This advanced preclinical model further mimics the original psoriatic skin and will prove relevant in predicting clinical outcomes, thereby decreasing inaccurate predictions of compound effects.
Collapse
|
95
|
Shehwana H, Ijaz S, Fatima A, Walton S, Sheikh ZI, Haider W, Naz S. Transcriptome Analysis of Host Inflammatory Responses to the Ectoparasitic Mite Sarcoptes scabiei var. hominis. Front Immunol 2021; 12:778840. [PMID: 34925353 PMCID: PMC8671885 DOI: 10.3389/fimmu.2021.778840] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/02/2021] [Indexed: 11/30/2022] Open
Abstract
Scabies, a human skin infestation caused by the ectoparasitic mite Sarcoptes scabiei var. hominis, affects more than 200 million people globally. The prevailing knowledge of the disease process and host immune response mechanisms is limited. A better understanding of the host-parasite relationship is essential for the identification of novel vaccine and drug targets. Here we aimed to interrogate the transcriptomic profiles of mite-infested human skin biopsies with clinical manifestations of ordinary scabies subjects ("OS"; n = 05) and subjects naive to scabies ("control"; n = 03) using RNASeq data analysis. A combined clustering, network, and pathway mapping approach enabled us to identify key signaling events in the host immune and pro-inflammatory responses to S. scabiei infestation. The clustering patterns showed various differentially expressed genes including inflammatory responses and innate immunity genes (DEFB4A, IL-19, CXCL8, CSF3, SERPINB4, S100A7A, HRNR) and notably upregulation of the JAK-STAT pathway in scabies-infested samples. Mite-infested human skin biopsies (GSE178563) were compared with an ex-vivo porcine infested model (E-MTAB-6433) and human skin equivalents (GSE48459). Marked enrichment of immune response pathways (JAK-STAT signaling, IL-4 and IL-13 pathway, and Toll receptor cascade), chemokine ligands and receptors (CCL17, CCL18, CCL3L1, CCL3L3, CCR7), and cytokines (IL-13 and IL-20) were observed. Additionally, genes known for their role in psoriasis and atopic dermatitis were upregulated, e.g., IL-19. The detailed transcriptomic profile has provided an insight into molecular functions, biological processes, and immunological responses and increased our understanding about transcriptomic regulation of scabies in human.
Collapse
Affiliation(s)
- Huma Shehwana
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Sadaf Ijaz
- Research Centre for Modelling & Simulation, National University of Science and Technology, Islamabad, Pakistan
| | - Abeera Fatima
- Research Centre for Modelling & Simulation, National University of Science and Technology, Islamabad, Pakistan
| | - Shelley Walton
- Inflammation and Healing Research Cluster, School of Health and Sport Sciences, University of the Sunshine Coast, Maroochydore, QLD, Australia
| | - Zafar Iqbal Sheikh
- Department of Dermatology, Pak-Emirates Military Hospital, Rawalpindi, Pakistan
| | - Waseem Haider
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Shumaila Naz
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| |
Collapse
|
96
|
Immune Checkpoint Blockade and Skin Toxicity Pathogenesis. J Invest Dermatol 2021; 142:951-959. [PMID: 34844731 DOI: 10.1016/j.jid.2021.06.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022]
Abstract
Immune checkpoint blockade has revolutionized the treatment of multiple tumor types, including melanoma and nonmelanoma skin cancers. The use of immune checkpoint blockade is curtailed by tissue toxicities termed immune-related adverse events (irAEs), which occur most quickly and most often in the skin. We review the rationale for immune checkpoint blockade use, current agents, use in skin cancers, autoimmune manifestations in the skin, and considerations for predictive biomarkers and treatment options on the basis of skin pathogenesis. We also highlight major gaps in the field and the lack of preclinical modeling in the skin. A deeper understanding of irAE pathophysiology may help to uncouple toxicity and efficacy but mandates an interdisciplinary approach, including foundational skin immunology and autoimmune pathogenesis.
Collapse
|
97
|
Liu X, Hu Z, Zhang J, Ma T, Wu W, Wei X, Wang Z, Zhen H, Zhou H, Huang N, Li J. IL-30 ameliorates imiquimod and K14-VEGF induced psoriasis-like disease by inhibiting both innate and adaptive immunity disorders. Biochem Biophys Res Commun 2021; 579:97-104. [PMID: 34597998 DOI: 10.1016/j.bbrc.2021.09.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 09/18/2021] [Indexed: 02/08/2023]
Abstract
Psoriasis is a severe skin disease with significant physical and psychological health consequences. As a typical type of immune disease, both innate and adaptive immunity disorders play key roles in the development of psoriasis. Interleukin (IL)-30 was thought as a natural antagonist of gp130-mediated signaling that affects T helper type 1 and 17 cell polarization by inhibiting IL-6 and IL-27 signaling pathways. Here, we found that, in vitro, IL-30 reduced cytokine levels of HaCaT keratinocytes and dendritic cells (DCs), weakened the maturationS of DCs, inhibited DC-mediated T cell proliferation, and blocked the activation of nuclear factor-κB. In vivo, IL-30 inhibited the development of skin disease in two animal models: Krt14-Vegfa and imiquimod (IMQ)-induced psoriasis-like skin disease. Thus, IL-30 may be useful as a therapeutic agent for controlling psoriasis.
Collapse
Affiliation(s)
- Xiao Liu
- Human Sperm Bank, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhonglan Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Jun Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Teng Ma
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Wenlin Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaoqiong Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhen Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Huaping Zhen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Hong Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Nongyu Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
98
|
Samuelov L, Bochner R, Magal L, Malovitski K, Sagiv N, Nousbeck J, Keren A, Fuchs-Telem D, Sarig O, Gilhar A, Sprecher E. Vorinostat, a histone deacetylase inhibitor, as a potential novel treatment for psoriasis. Exp Dermatol 2021; 31:567-576. [PMID: 34787924 DOI: 10.1111/exd.14502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 10/05/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Psoriasis is characterized by aberrant activation of several pro-inflammatory circuits as well as abnormal hyperproliferation and dysregulated apoptosis of keratinocytes (KCs). Most currently available therapeutic options primarily target psoriasis-associated immunological defects rather than epidermal abnormalities. OBJECTIVE To investigate the efficacy of the histone deacetylase (HDAC) inhibitor, Vorinostat, in targeting hyperproliferation and impaired apoptosis in psoriatic skin. METHODS Vorinostat effect was investigated in primary KCs cell cultures using cell cycle analysis by flow cytometry, apoptosis assays (Annexin V-FICH and caspase-3/7) and antibody arrays, qRT-PCR and immunohistochemistry. Vorinostat impact on clinical manifestations of psoriasis was investigated in a chimeric mouse model. RESULTS Vorinostat was found to inhibit KCs proliferation and to induce their differentiation and apoptosis. Using a chimeric mouse model, vorinostat was found to result in marked attenuation of a psoriasiform phenotype with a significant decrease in epidermal thickness and inhibition of epidermal proliferation. CONCLUSIONS Our results support the notion that vorinostat, a prototypic HDAC inhibitor, may be of potential use in the treatment of psoriasis and other hyperproliferative skin disorders.
Collapse
Affiliation(s)
- Liat Samuelov
- Division of Dermatology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ron Bochner
- Division of Dermatology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Lee Magal
- Division of Dermatology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Kiril Malovitski
- Division of Dermatology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Nadav Sagiv
- Division of Dermatology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Janna Nousbeck
- Division of Dermatology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Aviad Keren
- Skin Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Dana Fuchs-Telem
- Division of Dermatology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Ofer Sarig
- Division of Dermatology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Amos Gilhar
- Skin Research Laboratory, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Eli Sprecher
- Division of Dermatology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
99
|
Bregnhøj A, Thuesen KKH, Emmanuel T, Litman T, Grek CL, Ghatnekar GS, Johansen C, Iversen L. HSP90 inhibitor RGRN-305 for oral treatment of plaque type psoriasis: efficacy, safety and biomarker results in an open-label proof-of-concept study. Br J Dermatol 2021; 186:861-874. [PMID: 34748646 DOI: 10.1111/bjd.20880] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND HSP90 is a downstream regulator of tumor necrosis factor α (TNFα) and interleukin (IL)-17A signaling and may therefore serve as a novel target in the treatment of psoriasis. OBJECTIVE This phase 1b proof-of-concept study was undertaken to evaluate the safety and efficacy of a novel HSP90 inhibitor (RGRN-305) in the treatment of plaque psoriasis. METHODS An open-label, single-arm, dose-selection, single-center proof-of-concept study. Patients with plaque psoriasis were treated with 250 mg or 500 mg RGRN-305 daily for 12 weeks. Efficacy was evaluated clinically using Psoriasis Area and Severity Index (PASI), body surface area (BSA), and Physician Global Assessment (PGA) scores and by Dermatology Life Quality Index (DLQI). Skin biopsies collected at baseline and at 4, 8, and 12 weeks after treatment start were used for immunohistochemical staining and for gene expression analysis. Safety was monitored via laboratory tests, vital signs, ECG, and physical examinations. RESULTS Six of the eleven patients completing the study responded to RGRN-305 with a PASI improvement between 71% and 94%, whereas five patients were considered nonresponders with a PASI response < 50%. No severe adverse events were reported. Four of seven patients treated with 500 mg RGRN-305 daily experienced a mild to moderate exanthematous drug induced eruption due to study treatment. Two patients chose to discontinue the study due to this exanthematous eruption. RGRN-305 treatment resulted in pronounced inhibition of the IL-23, TNFα, and IL-17A signaling pathways and normalization of both histological changes and psoriatic lesion gene expression profiles in patients responding to treatment. CONCLUSION Treatment with RGRN-305 showed an acceptable safety, especially in the low-dose group, and was associated with clinically meaningful improvement in a subset of patients with plaque psoriasis, indicating that HSP90 may serve as a novel future target in psoriasis treatment.
Collapse
Affiliation(s)
- A Bregnhøj
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - K K H Thuesen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - T Emmanuel
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - T Litman
- Department of Immunology and Microbiology, University of Copenhagen, 2200, Copenhagen, Denmark
| | - C L Grek
- FirstString Research, Mount Pleasant, SC, 29464, USA
| | | | - C Johansen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - L Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
100
|
Shokrian Zeini M, Haddadi NS, Shayan M, Shokrian Zeini M, Kazemi K, Solaimanian S, Abdollahifar MA, Hedayatyanfard K, Dehpour AR. Losartan ointment attenuates imiquimod-induced psoriasis-like inflammation. Int Immunopharmacol 2021; 100:108160. [PMID: 34583123 DOI: 10.1016/j.intimp.2021.108160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Psoriasis is a chronic skin condition associated with interleukin-23/interleukin-17 (IL-23/IL-17) pathway. Recent evidence declares that angiotensin II (Ang II) induces a potent IL-17-related inflammation. Meanwhile, Losartan, an angiotensin one receptor (AT1R) antagonist, attenuates the TH17-related responses. Therefore, we investigated the possible beneficial effects of topically applied Losartan1% ointment on imiquimod (IMQ)-induced psoriasis in mice. METHOD Psoriasis was induced in mice consecutively for five days by topical IMQ on the shaved back. The IMQ-induced psoriasis was treated via topical administration of Losartan1% twice a day. The severity of skin inflammation was evaluated employing Psoriasis Area and Severity Index (PASI) scores. Subsequently, the skin samples were assessed using Baker's scoring system, stereological studies, and biochemical assessment with real-time PCR and immunohistochemistry. RESULTS IMQ administration induced plaque-type psoriasis and skin inflammation. We characterized psoriatic lesions by hyperkeratosis, Munro abscess, rete ridges, and marked T-cell infiltrates. IMQ significantly increased epidermal volume, mRNA expression of IL-17a, IL-23, Ang II, AT1R, and TNF-α levels compared with the Placebo group. Topical administration of Losartan1% on IMQ-induced psoriasis significantly reduced the PASI scores and alleviated the erythema and scaling. The treatment significantly decreased the psoriatic thickness and dermal T-cell infiltration. Regarding biochemical assessment, Losartan1% considerably reduced the IMQ-induced increase of IL-17a, Ang II, and AT1R expression in the skin. CONCLUSION Topical Losartan1% significantly alleviates psoriasis by reducing AT1R and IL-17a expression. Our results introduce AT1Rs as a promising therapeutic target in psoriasis and represent a link between angiotensin and TH17-related inflammation. However, the effects of AngII-AT1R systems on IL-17 signaling need to be confirmed by further investigations.
Collapse
Affiliation(s)
- Maryam Shokrian Zeini
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazgol-Sadat Haddadi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Dermatology, University of Massachusetts School of Medicine, Worchester, MA, USA
| | - Maryam Shayan
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohadese Shokrian Zeini
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiarash Kazemi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahabaddin Solaimanian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Keshvad Hedayatyanfard
- Department of Physiology and Pharmacology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran; Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran.
| | - Ahmad-Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|