1
|
Liu H, Chen Y, Xu S, Chen H, Qiu F, Liang CL, Mo X, Liu J, Lu C, Dai Z. Electroacupuncture and methotrexate cooperate to ameliorate psoriasiform skin inflammation by regulating the immune balance of Th17/Treg. Int Immunopharmacol 2024; 140:112702. [PMID: 39094355 DOI: 10.1016/j.intimp.2024.112702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 06/02/2024] [Accepted: 07/14/2024] [Indexed: 08/04/2024]
Abstract
Psoriasis is an autoinflammatory dermatosis, while methotrexate (MTX) is an immunosuppressant used to treat psoriasis. However, conventional immunosuppressants may cause various side effects. Acupuncture has potential benefits in treating psoriasis based on its anti-inflammatory effects. However, the immune mechanisms underlying its effects remain unclear. In this study, imiquimod-induced psoriatic mice were used to investigate the effects and mechanisms of electroacupuncture (EA) and, in particular, its joint treatment with MTX. We found that treatment with either EA or MTX ameliorated psoriasiform skin lesions, improved skin pathology and reduced proinflammatory cytokines in the skin, while joint treatment with both EA and MTX further alleviated the skin lesions and inflammation compared to either one alone. Moreover, percentages of CD4+ IL-17A+ Th17 cells in the skin and lymph nodes were decreased by EA or MTX and further lowered by combined EA+MTX treatment. Similarly, EA or MTX also reduced their RORγt expression. On the contrary, CD4+ FoxP3+ Treg frequency in psoriatic mice was augmented by EA or MTX and further increased by the joint treatment. However, depleting Tregs mostly reversed the therapeutic effects of EA or EA plus MTX. Additionally, the phosphorylated NF-κB (p65) expression was suppressed by treatment with EA, MTX or better with EA+MTX. Meanwhile, the anti-inflammatory effects of EA plus MTX were offset by an NF-κB agonist. Thus, this study has revealed that EA cooperates with MTX to balance Th17/Treg responses and to ameliorate psoriasiform skin inflammation through suppressing NF-κB activation. Our findings may be implicated for treating human psoriasis.
Collapse
Affiliation(s)
- Huazhen Liu
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Yuchao Chen
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Siyuan Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Haiming Chen
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Feifei Qiu
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Chun-Ling Liang
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Xiumei Mo
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Junfeng Liu
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Chuanjian Lu
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Zhenhua Dai
- Joint Immunology Program, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
2
|
Bubna AK, Viplav V. Revisiting risankizumab: a newer biologic drug in dermatology. Ital J Dermatol Venerol 2024; 159:543-554. [PMID: 39422528 DOI: 10.23736/s2784-8671.24.07971-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Risankizumab is an interleukin 23p19 inhibitor, approved by the USA Food and Drug Administration (FDA) for the management of moderate to severe plaque psoriasis. Apart from its utility in psoriasis, there are a number of other dermatologic conditions where risankizumab has demonstrated value. The aim of this narrative review is to describe the utility of risankizumab in psoriasis, as well as its implication in off-label dermatologic disorders. EVIDENCE ACQUISITION PubMed, Google Scholar, Scopus and ResearchGate were searched for scholarly articles related to risankizumab and its utility in dermatology using the search terms "Risankizumab" AND "Psoriasis" AND "other dermatological disorders." EVIDENCE SYNTHESIS Risankizumab is a valuable biologic agent for the management of psoriasis and psoriatic arthropathy. It has also been used successfully for other dermatologic disorders like hidradenitis suppurativa, pityriasis rubra pilaris and pyoderma gangrenosum. CONCLUSIONS Risankizumab's usage is not limited to psoriasis. Its benefit extends to many more dermatologic conditions. Besides, it has an acceptable safety profile.
Collapse
Affiliation(s)
- Aditya K Bubna
- Department of Dermatology, Katihar Medical College, Al-Karim University, Katihar, India -
| | - Vinayak Viplav
- Department of Dermatology, Katihar Medical College, Al-Karim University, Katihar, India
| |
Collapse
|
3
|
Xu J, Xie R, Ji Y, Qian C, Zhang X, Todd K, Wang F, Cui Y. Safety and Pharmacokinetics of Single-Dose Mirikizumab in Chinese Healthy Participants: Results From a Phase 1 Study. Clin Pharmacol Drug Dev 2024; 13:1143-1150. [PMID: 39230208 DOI: 10.1002/cpdd.1449] [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: 03/26/2024] [Accepted: 06/17/2024] [Indexed: 09/05/2024]
Abstract
The objective of this phase 1 single-dose study was to evaluate the safety, tolerability, and pharmacokinetics of mirikizumab in Chinese healthy adults. Sixty participants were randomized within 5 planned dose cohorts: intravenous (IV) 300 mg, IV 600 mg, IV 1200 mg, subcutaneous (SC) 200 mg, and SC 400 mg to receive mirikizumab (10 participants in each cohort) or placebo (2 participants in each cohort). No death or serious adverse events occurred. Twenty-eight (56.0%) participants who received mirikizumab reported 49 treatment-emergent adverse events (TEAEs) and 8 (80.0%) participants who received placebo reported 18 TEAEs. The majority of TEAEs were mild in severity. Following IV 300-1200 mg mirikizumab, the arithmetic mean of both area under the concentration versus time curve from time 0 to infinity (AUC0-∞) and maximum observed drug concentration (Cmax) increased by approximately 3.5-fold, and the arithmetic mean half-life (t1/2) ranged from 9.64 to 12.0 days. Following SC 200 and 400 mg mirikizumab, the arithmetic mean of both AUC0-∞ and Cmax increased by approximately 1.6-fold, the median time to Cmax (tmax) was 2.98 days for both, and the arithmetic mean t1/2 was 10.6 and 10.5 days, respectively. Absolute bioavailability based on pooled SC and IV dose data was 38.2%. In this study, the safety and pharmacokinetic profile of mirikizumab were consistent with what has been reported in other studies.
Collapse
Affiliation(s)
- Junyu Xu
- Drug Clinical Trial Institution, Peking University First Hospital, Beijing, China
| | - Ran Xie
- Drug Clinical Trial Institution, Peking University First Hospital, Beijing, China
| | | | | | - Xin Zhang
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Kris Todd
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Feng Wang
- Eli Lilly and Company, Shanghai, China
| | - Yimin Cui
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| |
Collapse
|
4
|
Catlett IM, Gao L, Hu Y, Banerjee S, Krueger JG. Pharmacodynamic Response to Deucravacitinib, an Oral, Selective, Allosteric TYK2 Inhibitor, in a Global, Phase 2, Randomized, Double-Blind, Placebo-Controlled Psoriasis Trial. Dermatol Ther (Heidelb) 2024; 14:2827-2839. [PMID: 39283417 PMCID: PMC11480296 DOI: 10.1007/s13555-024-01262-5] [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: 05/28/2024] [Accepted: 08/20/2024] [Indexed: 10/17/2024] Open
Abstract
BACKGROUND Psoriasis, a chronic, immune-mediated, inflammatory disease, affects 2‒3% of the population. Tyrosine kinase 2 (TYK2) mediates cytokine signaling involved in adaptive [interleukin (IL)-12, IL-23] and innate (type-I interferons) immune responses; IL-23-driven T-helper (Th)17 pathways play a key role in chronic inflammation in psoriasis. In a phase 2 trial, deucravacitinib, an oral, selective, allosteric TYK2 inhibitor, reduced IL-23/Th17 and type-I interferon pathway expression in the skin of patients with moderate to severe plaque psoriasis, reductions that were accompanied by clinical improvement of psoriatic lesions. OBJECTIVES The aim of this study was to identify biomarkers of psoriatic disease in serum from patients enrolled in the phase 2 trial and to assess the effects of deucravacitinib on those biomarkers. METHODS Serum biomarkers from Olink proteomics and other quantitative assays were evaluated for a pharmacodynamic response to deucravacitinib treatment and correlation with psoriasis disease activity measures. RESULTS Serum biomarkers associated with the IL-23/Th17 pathway [IL-17A, IL-17C, IL-19, IL-20, beta-defensin, and peptidase inhibitor 3 (PI3)] were upregulated in patients with psoriasis versus healthy controls. Deucravacitinib treatment reduced IL-17A (adjusted mean change from baseline at Day 85; 12 mg once daily versus placebo; -0.240 versus -0.067), IL-17C (-14.850 versus -1.664), IL-19 (-96.445 versus -8.119), IL-20 (-0.265 versus -0.064), beta-defensin (-65,025.443 versus -7553.961), and PI3 (-14.005 versus -1.360) expression. Reductions in serum biomarker expression occurred in a dose- and time-dependent manner, with significant reductions from baseline seen with deucravacitinib doses ≥ 3 mg twice daily (P ≤ 0.05). Biomarker expression correlated with disease activity measures such as Psoriasis Area and Severity Index (PASI) at baseline. Biomarker expression also correlated with PASI scores at Week 12. CONCLUSION IL-23/Th17 pathway expression in the serum of patients with psoriasis is an indicator of disease activity and response to deucravacitinib treatment. TRIAL REGISTRATION NUMBER NCT02931838.
Collapse
Affiliation(s)
- Ian M Catlett
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA
| | - Lu Gao
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA.
| | - Yanhua Hu
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA
| | - Subhashis Banerjee
- Bristol Myers Squibb, 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA
| | | |
Collapse
|
5
|
DeBerg HA, Fahning ML, Schlenker JD, Schmitt WP, Gratz IK, Carlin JS, Campbell DJ, Morawski PA. T cells promote distinct transcriptional programs of cutaneous inflammatory disease in human skin structural cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606077. [PMID: 39131334 PMCID: PMC11312529 DOI: 10.1101/2024.07.31.606077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
T cells and structural cells coordinate appropriate inflammatory responses and restoration of barrier integrity following insult. Dysfunctional T cell activity precipitates tissue pathology that occurs alongside disease-associated alterations of structural cell subsets, but the mechanisms by which T cells promote these changes remain unclear. We show that subsets of circulating and skin-resident CD4+ T cells promote distinct transcriptional outcomes in human keratinocytes and dermal fibroblasts that correspond with divergent T cell cytokine production. Using these transcriptional signatures, we identify T cell-dependent outcomes associated with inflammatory skin disease, including a set of Th17 cell-induced genes in keratinocytes that are enriched in the skin during psoriasis and normalized by anti-IL-17 therapy, and a skin-resident T cell-induced gene module enriched in scleroderma-associated fibroblasts. Interrogating clinical data using T cell-derived structural cell gene networks enables investigation of the immune-dependent contribution to inflammatory disease and the heterogeneous patient response to biologic therapy.
Collapse
Affiliation(s)
- Hannah A. DeBerg
- Center for Systems Immunology, Benaroya Research Institute, Seattle, Washington, USA
| | - Mitch L. Fahning
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, Washington, USA
| | - James D. Schlenker
- Plastic and Reconstructive Surgery, Virginia Mason Medical Center, Seattle, Washington, USA
| | - William P. Schmitt
- Plastic and Reconstructive Surgery, Virginia Mason Medical Center, Seattle, Washington, USA
| | - Iris K. Gratz
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, Washington, USA
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
- EB House Austria, Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- Center for Tumor Biology and Immunology, University of Salzburg, Salzburg, Austria
| | - Jeffrey S. Carlin
- Center for Translational Immunology, Benaroya Research Institute, Seattle, Washington, USA
- Division of Rheumatology, Virginia Mason Medical Center, Seattle, Washington, USA
| | - Daniel J. Campbell
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, Washington, USA
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Peter A. Morawski
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, Washington, USA
| |
Collapse
|
6
|
Lv Y, Qi J, Babon JJ, Cao L, Fan G, Lang J, Zhang J, Mi P, Kobe B, Wang F. The JAK-STAT pathway: from structural biology to cytokine engineering. Signal Transduct Target Ther 2024; 9:221. [PMID: 39169031 PMCID: PMC11339341 DOI: 10.1038/s41392-024-01934-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/12/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway serves as a paradigm for signal transduction from the extracellular environment to the nucleus. It plays a pivotal role in physiological functions, such as hematopoiesis, immune balance, tissue homeostasis, and surveillance against tumors. Dysregulation of this pathway may lead to various disease conditions such as immune deficiencies, autoimmune diseases, hematologic disorders, and cancer. Due to its critical role in maintaining human health and involvement in disease, extensive studies have been conducted on this pathway, ranging from basic research to medical applications. Advances in the structural biology of this pathway have enabled us to gain insights into how the signaling cascade operates at the molecular level, laying the groundwork for therapeutic development targeting this pathway. Various strategies have been developed to restore its normal function, with promising therapeutic potential. Enhanced comprehension of these molecular mechanisms, combined with advances in protein engineering methodologies, has allowed us to engineer cytokines with tailored properties for targeted therapeutic applications, thereby enhancing their efficiency and safety. In this review, we outline the structural basis that governs key nodes in this pathway, offering a comprehensive overview of the signal transduction process. Furthermore, we explore recent advances in cytokine engineering for therapeutic development in this pathway.
Collapse
Affiliation(s)
- You Lv
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100080, China
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Longxing Cao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Guohuang Fan
- Immunophage Biotech Co., Ltd, No. 10 Lv Zhou Huan Road, Shanghai, 201112, China
| | - Jiajia Lang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jin Zhang
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Pengbing Mi
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland, 4072, Australia.
| | - Faming Wang
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China.
| |
Collapse
|
7
|
Florian TL, Florian IA, Vesa SC, Beni L, Orăsan M. Inflammatory Cytokines and Clinical Outcome Following Biological Therapy in Adult Bio-Naïve Psoriasis Patients. Curr Issues Mol Biol 2024; 46:7719-7729. [PMID: 39057098 PMCID: PMC11276069 DOI: 10.3390/cimb46070457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Inflammatory cytokines may hold the key to the clinical evolution of psoriasis. The aims of this study are to find a correlation between levels of inflammatory cytokines such as TNF-α, IL-23, IL-17A, and IL-17F and disease duration and severity scores in psoriasis; to test if the decrease in any of the aforementioned cytokines is correlated with an amelioration in disease severity scores; and to analyze if any of the four biologic agents used are linked with a greater decrease in overall cytokine levels. We enrolled 23 adult patients under treatment with ixekizumab, secukinumab, guselkumab, or adalimumab and measured psoriasis disease severity scores PASI (Psoriasis Area Severity Index) and DLQI (Dermatology Life Quality Index), as well as the levels of the aforementioned cytokines at the start of therapy and after 3 months of continuous treatment. Inclusion criteria were the presence of psoriasis, age above 18 years and the need to initiate biological therapy (lack of response to standard treatment). Biological therapies resulted in an amelioration of PASI and DLQI scores, as well as levels of TNF-α, IL-23 and IL-17F. Disease duration and PASI and DLQI scores did not correlate with cytokine levels except DLQI and IL-23 score, in a paradoxically inversely proportional manner. IL-23, in particular, could be a useful biomarker for checking treatment response in psoriasis.
Collapse
Affiliation(s)
- Teodora-Larisa Florian
- Department of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Ioan-Alexandru Florian
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Stefan Cristian Vesa
- Department of Pharmacology, “Iuliu Hatieganu” University of Medicine and Pharmacy, No. 23 Marinescu Street, 400337 Cluj-Napoca, Romania;
| | - Lehel Beni
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Meda Orăsan
- Department of Pathophysiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
8
|
Bubna AK, Viplav V. Guselkumab - In Psoriasis and Beyond. Dermatol Pract Concept 2024; 14:dpc.1403a181. [PMID: 39122539 PMCID: PMC11314551 DOI: 10.5826/dpc.1403a181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2024] [Indexed: 08/12/2024] Open
Abstract
INTRODUCTION Guselkumab is an interleukin 23p19 inhibitor, and the first in this group, to be approved by the US Food and Drug Administration for the management of moderate to severe psoriasis. Apart from its utility in psoriasis, there are a number of other dermatologic conditions where guselkumab has demonstrated value. OBJECTIVES The aim of this narrative review is to describe the utility of guselkumab in psoriasis as well as its implication in off-label dermatologic disorders. METHODS Pubmed, Google Scholar, Scopus and ResearchGate were searched for scholarly articles related to guselkumab and its utility in dermatology using the search terms "Guselkumab" AND "Psoriasis" AND "other dermatological disorders". RESULTS Guselkumab is a valuable biologic agent for the management of psoriasis and psoriatic arthropathy. It has also been used successfully for other dermatologic disorders like hidradenitis suppurativa, lichen planus, pityriasis rubra pilaris and pyoderma gangrenosum. Recently, its utility in Stewart-Treves angiosarcoma (STA) has been exemplified. CONCLUSION Guselkumab usage is not limited to psoriasis. Its benefit extends to many more dermatologic conditions. Its utility in STA could open an avenue for its application in the field of oncology. Furthermore, it has an acceptable safety profile.
Collapse
Affiliation(s)
- Aditya Kumar Bubna
- Department of Dermatology, Katihar Medical College, Karim Bagh, Katihar, Bihar, India
| | - Vinayak Viplav
- Department of Dermatology, Katihar Medical College, Karim Bagh, Katihar, Bihar, India
| |
Collapse
|
9
|
Wang L, Dou YX, Yu QX, Hu Z, Ip SP, Xian YF, Lin ZX. Improvement effects of a novel Chinese herbal formula in imiquimod and IL-23-stimulated mouse models of psoriasis. Chin Med 2024; 19:81. [PMID: 38858762 PMCID: PMC11165727 DOI: 10.1186/s13020-024-00951-9] [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: 04/03/2024] [Accepted: 05/22/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Psoriasis is a long-term inflammatory skin disease. A novel herbal formula containing nine Chinese herbal medicines, named Inflammation Skin Disease Formula (ISDF), has been prescribed in clinics for decades. AIMS To investigate the efficacy and action mechanisms of ISDF on psoriasis using imiquimod (IMQ) and Interleukin-23 (IL-23)-induced models in mice and reveal the pharmacokinetics profile of ISDF in rats. METHODS Topical administration of IMQ and intradermal injection with IL-23 respectively induced skin lesions like psoriasis on the dorsal area of Balb/c and C57 mice. The mice's body weight, skin thickness, and psoriasis area and severity index (PASI) were assessed weekly. SD rats were used in the pharmacokinetics study and the contents of berberine and baicalin were determined. RESULTS The PASI scores and epidermal thickness of mice were markedly decreased after ISDF treatment in both models. ISDF treatment significantly decreased the contents of IL-17A and IL-22 in the serum of IMQ- and IL-23-treated mice. Importantly, ISDF markedly downregulated IL-4, IL-6, IL-1β, and tumor necrosis factor α (TNF-α) gene expression, and the phosphorylation of NF-κB p65, JNK, ERKs and MAPK p38 in IMQ-treated mice. The protein phosphorylation of Jak1, Jak2, Tyk2 and Stat3 was significantly mitigated in the ISDF-treated groups. The absorption of baicalin and berberine of ISDF through the gastrointestinal tract of rats was limited, and their distribution and metabolism in rats were also very slow, which suggested ISDF could be used in the long-term application. CONCLUSIONS ISDF has a strong anti-psoriatic therapeutic effect on mouse models induced with psoriasis through IMQ and IL-23, which is achieved by inhibiting the activation of the Jak/Stat3-activated IL-23/Th17 axis and the downstream NF-κB signalling and MAPK signalling pathways. ISDF holds great potential to be a therapy for psoriasis and should be further developed for this purpose.
Collapse
Affiliation(s)
- Lan Wang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Room 101-J, 1/F, Li Wai Chun Building, Shatin , Hong Kong SAR, NT, China
| | - Yao-Xing Dou
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiu-Xia Yu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Room 101-J, 1/F, Li Wai Chun Building, Shatin , Hong Kong SAR, NT, China
| | - Zhen Hu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Room 101-J, 1/F, Li Wai Chun Building, Shatin , Hong Kong SAR, NT, China
| | - Siu-Po Ip
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Room 101-J, 1/F, Li Wai Chun Building, Shatin , Hong Kong SAR, NT, China
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Room 101-J, 1/F, Li Wai Chun Building, Shatin , Hong Kong SAR, NT, China.
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Room 101-J, 1/F, Li Wai Chun Building, Shatin , Hong Kong SAR, NT, China.
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
10
|
Nazimek K, Bryniarski K. Macrophage Functions in Psoriasis: Lessons from Mouse Models. Int J Mol Sci 2024; 25:5306. [PMID: 38791342 PMCID: PMC11121292 DOI: 10.3390/ijms25105306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Psoriasis is a systemic autoimmune/autoinflammatory disease that can be well studied in established mouse models. Skin-resident macrophages are classified into epidermal Langerhans cells and dermal macrophages and are involved in innate immunity, orchestration of adaptive immunity, and maintenance of tissue homeostasis due to their ability to constantly shift their phenotype and adapt to the current microenvironment. Consequently, both macrophage populations play dual roles in psoriasis. In some circumstances, pro-inflammatory activated macrophages and Langerhans cells trigger psoriatic inflammation, while in other cases their anti-inflammatory stimulation results in amelioration of the disease. These features make macrophages interesting candidates for modern therapeutic strategies. Owing to the significant progress in knowledge, our review article summarizes current achievements and indicates future research directions to better understand the function of macrophages in psoriasis.
Collapse
Affiliation(s)
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University Medical College, 31-121 Krakow, Poland;
| |
Collapse
|
11
|
Riedl R, Kühn A, Hupfer Y, Hebecker B, Peltner LK, Jordan PM, Werz O, Lorkowski S, Wiegand C, Wallert M. Characterization of Different Inflammatory Skin Conditions in a Mouse Model of DNCB-Induced Atopic Dermatitis. Inflammation 2024; 47:771-788. [PMID: 38150167 DOI: 10.1007/s10753-023-01943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/10/2023] [Accepted: 12/04/2023] [Indexed: 12/28/2023]
Abstract
The mouse model of 2,4-dinitrochlorbenzene (DNCB)-induced human-like atopic dermatitis (hlAD) has been widely used to test novel treatment strategies and compounds. However, the study designs and methods are highly diverse, presenting different hlAD disease patterns that occur after sensitization and repeated challenge with DNCB on dorsal skin. In addition, there is a lack of information about the progression of the disease during the experiment and the achieved pheno- and endotypes, especially at the timepoint when therapeutic treatment is initiated. We here examine hlAD in a DNCB-induced BALB/cJRj model at different timepoints: (i) before starting treatment with dexamethasone, representing a standard drug control (day 12) and (ii) at the end of the experiment (day 22). Both timepoints display typical AD-associated characteristics: skin thickening, spongiosis, hyper- and parakeratosis, altered cytokine and gene expression, increased lipid mediator formation, barrier protein and antimicrobial peptide abnormalities, as well as lymphoid organ hypertrophy. Increased mast cell infiltration into the skin and elevated immunoglobulin E plasma concentrations indicate a type I allergy response. The DNCB-treated skin showed an extrinsic moderate sub-acute hlAD lesion at day 12 and an extrinsic mild sub-acute to chronic pheno- and endotype at day 22 with a dominating Th2 response. A dependency of the filaggrin formation and expression in correlation to the disease severity in the DNCB-treated skin was found. In conclusion, our study reveals a detailed classification of a hlAD at two timepoints with different inflammatory skin conditions and pheno- and endotypes, thereby providing a better understanding of the DNCB-induced hlAD model in BALB/cJRj mice.
Collapse
Affiliation(s)
- Rebecca Riedl
- Department of Dermatology, Dermatological Research Laboratory, Jena University Hospital, 07747, Jena, Germany
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Annika Kühn
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Yvonne Hupfer
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
| | - Betty Hebecker
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, 07743, Jena, Germany
| | - Lukas K Peltner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743, Jena, Germany
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University, 07743, Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University, 07743, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University, 07743, Jena, Germany
| | - Stefan Lorkowski
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, 07743, Jena, Germany
| | - Cornelia Wiegand
- Department of Dermatology, Dermatological Research Laboratory, Jena University Hospital, 07747, Jena, Germany
| | - Maria Wallert
- Department of Biochemistry and Physiology of Nutrition, Institute of Nutritional Science, Friedrich Schiller University, 07743, Jena, Germany.
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, 07743, Jena, Germany.
| |
Collapse
|
12
|
Daniele SG, Eldirany SA, Damiani G, Ho M, Bunick CG. Structural Basis for p19 Targeting by Anti-IL-23 Biologics: Correlations with Short- and Long-Term Efficacy in Psoriasis. JID INNOVATIONS 2024; 4:100261. [PMID: 38445231 PMCID: PMC10914523 DOI: 10.1016/j.xjidi.2024.100261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 03/07/2024] Open
Abstract
IL-23 is central to psoriasis pathogenesis. Biologics targeting IL-23 are important therapies against psoriasis. IL-23 inhibitors risankizumab, tildrakizumab, and guselkumab bind the IL-23 p19 subunit, whereas ustekinumab binds p40; however, the structural composition of the IL-23-binding epitopes and how these molecular properties relate to clinical efficacy are not known. Utilizing epitope data derived from hydrogen-deuterium exchange or crystallographic experiments, we mapped inhibitor epitope locations, hydrophobicity, and surface charge onto the IL-23 surface. Molecular properties of each inhibitor epitope, including solvent-accessible surface area, were correlated to binding affinity, kinetic values, and clinical efficacy scores for plaque psoriasis through linear regression analysis. Each IL-23 inhibitor binds an epitope with a unique size, composition, and location except for a 10-residue overlap region outside of the IL-23 receptor epitope. We observed strong correlations between epitope surface area and KD and koff but not kon. Epitope surface area, KD, and koff were further associated with short-term (10-16 weeks) and long-term (44-60 weeks) clinical efficacy according to PASI-90 responses, with risankizumab demonstrating highest efficacy among IL-23 biologics. In contrast, kon, epitope hydrophobicity, polarity, and charge content did not correlate with efficacy. These data exemplify how molecular principles of medications within a therapeutic class can explain their differential clinical responses.
Collapse
Affiliation(s)
| | - Sherif A. Eldirany
- Department of Dermatology, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Giovanni Damiani
- Italian Center of Precision Medicine and Chronic Inflammation, Milan, Italy
- Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
- Graduate Program (PhD) in Pharmacological Sciences, Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Minh Ho
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Program in Translational Biomedicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christopher G. Bunick
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Program in Translational Biomedicine, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
13
|
Drakos A, Torres T, Vender R. Emerging Oral Therapies for the Treatment of Psoriasis: A Review of Pipeline Agents. Pharmaceutics 2024; 16:111. [PMID: 38258121 PMCID: PMC10819460 DOI: 10.3390/pharmaceutics16010111] [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: 11/24/2023] [Revised: 12/19/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The introduction of biologic agents for the treatment of psoriasis has revolutionized the current treatment landscape, targeting cytokines in the interleukin (IL)-23/IL-17 pathway and demonstrating strong efficacy and safety profiles in clinical trials. These agents however are costly, are associated with a risk of immunogenicity, and require administration by intravenous or subcutaneous injection, limiting their use among patients. Oral therapies, specifically small molecule and microbiome therapeutics, have the potential to be more convenient and cost-effective agents for patients and have been a focus of development in recent years, with few targeted oral medications available for the disease. In this manuscript, we review pipeline oral therapies for psoriasis identified through a search of ClinicalTrials.gov (30 June 2022-1 October 2023). Available preclinical and clinical trial data on each therapeutic agent are discussed. Small molecules under development include tumor necrosis factor inhibitors, IL-23 inhibitors, IL-17 inhibitors, phosphodiesterase-4 inhibitors, Janus kinase inhibitors, A3 adenosine receptor agonists, and sphingosine-1-phosphate receptor 1 agonists, several of which are entering phase III trials. Oral microbials have also demonstrated success in early phase studies. As new oral therapies emerge for the treatment of psoriasis, real-world data and comparative trials are needed to better inform their use among patients.
Collapse
Affiliation(s)
- Anastasia Drakos
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Tiago Torres
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal;
- Department of Dermatology, Centro Hospitalar de Santo António, 4099-001 Porto, Portugal
| | - Ronald Vender
- Dermatrials Research Inc. & Venderm Consulting, Hamilton, ON L8N 1Y2, Canada
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| |
Collapse
|
14
|
Park HW, Lee HS. IL-23 contributes to Particulate Matter induced allergic asthma in the early life of mice and promotes asthma susceptibility. J Mol Med (Berl) 2024; 102:129-142. [PMID: 37994911 DOI: 10.1007/s00109-023-02393-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/10/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
Air pollutant exposure leads to and exacerbates respiratory diseases. Particulate Matter (PM) is a major deleterious factor in the pathophysiology of asthma. Nonetheless, studies on the effects and mechanisms of exposure in the early life of mice remain unresolved. This study aimed to investigate changes in allergic phenotypes and effects on allergen-specific memory T cells resulting from co-exposure of mice in the early life to PM and house dust mites (HDM) and to explore the role of interleukin-23 (IL-23) in this process. PM and low-dose HDM were administered intranasally in 4-day-old C57BL/6 mice. After confirming an increase in IL-23 expression in mouse lung tissues, changes in the asthma phenotype and lung effector/memory Th2 or Th17 cells were evaluated after intranasal administration of anti-IL-23 antibody (Ab) during co-exposure to PM and HDM. Evaluation was performed up to 7 weeks after the last administration. Co-exposure to PM and low-dose HDM resulted in increases in airway hyperresponsiveness (AHR), eosinophils, neutrophils, and persistent Th2/Th17 effector/memory cells, which were all inhibited by anti-IL-23 Ab administration. When low-dose HDM was administered twice after a 7-week rest, mice exposed to PM and HDM during the previous early life period exhibited re-increases AHR, eosinophil count, HDM-specific IgG1, and effector/memory Th2 and Th17 cell populations. However, anti-IL-23 Ab administration during the early life period resulted in inhibition. Co-exposure to PM and low-dose HDM reinforced the allergic phenotypes and allergen-specific memory responses in early life of mice. During this process, IL-23 contributes to the enhancement of effector/memory Th2/Th17 cells and allergic phenotypes. KEY MESSAGES: PM-induced IL-23 expression, allergic responses in HDMinstilled mice of early life period. PM-induced effector/memory Th2/Th17 cells in HDMinstilled mice of early life period. Inhibition of IL-23 reduced the increase in allergic responses. Inhibition of IL-23 reduced the increase in allergic responses. After the resting period, HDM administration showed re-increase in allergic responses. Inhibition of IL-23 reduced the HDM-recall allergic responses.
Collapse
Affiliation(s)
- Heung-Woo Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Seung Lee
- Institute of Allergy and Clinical Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, Seoul, 110-744, Republic of Korea.
| |
Collapse
|
15
|
Amalia SN, Baral H, Fujiwara C, Uchiyama A, Inoue Y, Yamazaki S, Ishikawa M, Kosaka K, Sekiguchi A, Yokoyama Y, Ogino S, Torii R, Hosoi M, Shibasaki K, Motegi SI. TRPV4 Regulates the Development of Psoriasis by Controlling Adenosine Triphosphate Expression in Keratinocytes and the Neuroimmune System. J Invest Dermatol 2023; 143:2356-2365.e5. [PMID: 37263487 DOI: 10.1016/j.jid.2023.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 04/26/2023] [Accepted: 05/01/2023] [Indexed: 06/03/2023]
Abstract
TRPV4 is a calcium ion channel that is widely expressed in various cells. It is also involved in physiological and pathological processes. However, the role of TRPV4 in psoriasis remains unknown. We aimed to investigate the role of TRPV4 in psoriasis using human psoriasis skin samples and an imiquimod-induced psoriasis-like mouse model. Keratinocytes in human psoriasis skin had high TRPV4 expression. Trpv4-knockout mice had less severe dermatitis than wild-type mice in the imiquimod-induced mouse model. Knockout mice had significantly reduced epidermal thickness and a low number of infiltrated CD3+ T cells and CD68+ macrophages on the basis of histopathological studies and decreased mRNA expression of Il17a, Il17f, and Il23, as detected through qPCR. Furthermore, knockout mice had a significantly low expression of neuropeptides and the neuron marker PGP9.5. Adenosine triphosphate release was significantly suppressed by TRPV4 knockdown in both human and mouse keratinocytes in vitro. Finally, treatment with TRPV4 antagonist was significantly effective in preventing the progression of psoriasis-like dermatitis. In conclusion, TRPV4 mediates the expression of keratinocyte-derived adenosine triphosphate and increases the secretion of neuropeptides, resulting in the activation and amplification of IL-23/Th17 responses. Hence, TRPV4 can serve as a novel therapeutic target in psoriasis.
Collapse
Affiliation(s)
- Syahla Nisaa Amalia
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hritu Baral
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Chisako Fujiwara
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akihiko Uchiyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan.
| | - Yuta Inoue
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Sahori Yamazaki
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Mai Ishikawa
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Keiji Kosaka
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akiko Sekiguchi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoko Yokoyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Sachiko Ogino
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ryoko Torii
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Mari Hosoi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Koji Shibasaki
- Laboratory of Neurochemistry, Graduate School of Human Health Science, University of Nagasaki, Nagasaki, Japan
| | - Sei-Ichiro Motegi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
16
|
Ruggiero A, Portarapillo A, Megna M, Patruno C, Napolitano M. Management Strategies for Pediatric Moderate-to-Severe Plaque Psoriasis: Spotlight on Biologics. Pediatric Health Med Ther 2023; 14:435-451. [PMID: 38024562 PMCID: PMC10656860 DOI: 10.2147/phmt.s389108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Although psoriasis onset has been reported at any ages, in up to one-third of cases, it begins during childhood, with an estimated prevalence of about 2% in pediatric population. The management of moderate-to-severe forms of childhood psoriasis may represent a challenge for dermatologists, especially for parents' concerns about the need of systemic treatments. However, a prompt safe and effective treatment is mandatory in these patients, due to the significative impact that psoriasis may have on their quality of life, with well-known consequences on psychological health of both patients and caregivers. Due to the relatively frequent parents' refusal of systemic treatments, probably due to the fear of eventual adverse events, difficulties of oral or injective route, the management of moderate-to-severe forms still represents a challenge. Herein, we report a narrative review, aiming to resume the systemic treatments for pediatric psoriasis, focusing on the use of biologics and small molecules in the pediatric ages. The most widely used therapeutic strategies today for the pediatric population with moderate-severe psoriasis are traditional systemic therapies, while more innovative drugs such as biologics and small molecules now represent a somewhat unexplored but certainly promising field for unresponsive patients.
Collapse
Affiliation(s)
- Angelo Ruggiero
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, 80131, Italy
| | - Antonio Portarapillo
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, 80131, Italy
| | - Matteo Megna
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, 80131, Italy
| | - Cataldo Patruno
- Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Maddalena Napolitano
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, 80131, Italy
| |
Collapse
|
17
|
Gu SL, Maier T, Moy AP, Dusza S, Faleck DM, Shah NJ, Lacouture ME. IL12/23 Blockade with Ustekinumab as a Treatment for Immune-Related Cutaneous Adverse Events. Pharmaceuticals (Basel) 2023; 16:1548. [PMID: 38004414 PMCID: PMC10674871 DOI: 10.3390/ph16111548] [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: 09/12/2023] [Revised: 10/18/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
Background: Immune-related cutaneous adverse events (ircAEs) are frequent and may reduce quality of life and consistent dosing. IL12/23 has been implicated in psoriasis, which is reminiscent of the psoriasiform/lichenoid ircAE phenotype. We report the use of ustekinumab as a therapeutic option. Methods: Patients at Memorial Sloan Kettering Cancer Center, New York, who received immune checkpoint inhibitors and were treated with ustekinumab or had the keywords "ustekinumab" or "Stelara" in their clinical notes between 1 March 2017 and 1 December 2022 were retrospectively identified via a database query. Documentation from initial and follow-up visits was manually reviewed, and response to ustekinumab was categorized into complete cutaneous response (CcR, decrease to CTCAE grade 0), partial cutaneous response (PcR, any decrease in CTCAE grade exclusive of decrease to grade 0), and no cutaneous response (NcR, no change in CTCAE grade or worsening). Labs including complete blood count (CBC), cytokine panels, and IgE were obtained in a subset of patients as standard of care. Skin biopsies were reviewed by a dermatopathologist. Results: Fourteen patients with psoriasiform (85.7%), maculopapular (7.1%), and pyoderma gangrenosum (7.1%) ircAEs were identified. Ten (71.4%) receiving ustekinumab had a positive response to treatment. Among these 10 responders, 4 (40%) demonstrated partial cutaneous response and 6 (60%) demonstrated complete cutaneous resolution. Six patients (42.9%) experienced interruptions to their checkpoint inhibitor treatment as a result of intolerable ircAEs, and following ircAE management with ustekinumab, two (33.3%) were successfully rechallenged with their checkpoint inhibitors. On histopathology, patients primarily had findings of interface or psoriasiform dermatitis. No patients reported an adverse event related to ustekinumab. Conclusions: Ustekinumab showed a benefit in a subset of patients with psoriasiform/lichenoid ircAEs. No safety signals were identified. However, further prospective randomized controlled trials are needed to confirm our findings.
Collapse
Affiliation(s)
- Stephanie L. Gu
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; (S.L.G.)
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Tara Maier
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; (S.L.G.)
| | - Andrea P. Moy
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen Dusza
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; (S.L.G.)
| | - David M. Faleck
- Gastroenterology, Hepatology, and Nutrition Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Neil J. Shah
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mario E. Lacouture
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; (S.L.G.)
- Department of Dermatology, Weill Cornell Medical College, New York, NY 10021, USA
| |
Collapse
|
18
|
Xiao Q, Mears J, Nathan A, Ishigaki K, Baglaenko Y, Lim N, Cooney LA, Harris KM, Anderson MS, Fox DA, Smilek DE, Krueger JG, Raychaudhuri S. Immunosuppression causes dynamic changes in expression QTLs in psoriatic skin. Nat Commun 2023; 14:6268. [PMID: 37805522 PMCID: PMC10560299 DOI: 10.1038/s41467-023-41984-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 09/25/2023] [Indexed: 10/09/2023] Open
Abstract
Psoriasis is a chronic, systemic inflammatory condition primarily affecting skin. While the role of the immune compartment (e.g., T cells) is well established, the changes in the skin compartment are more poorly understood. Using longitudinal skin biopsies (n = 375) from the "Psoriasis Treatment with Abatacept and Ustekinumab: A Study of Efficacy"(PAUSE) clinical trial (n = 101), we report 953 expression quantitative trait loci (eQTLs). Of those, 116 eQTLs have effect sizes that were modulated by local skin inflammation (eQTL interactions). By examining these eQTL genes (eGenes), we find that most are expressed in the skin tissue compartment, and a subset overlap with the NRF2 pathway. Indeed, the strongest eQTL interaction signal - rs1491377616-LCE3C - links a psoriasis risk locus with a gene specifically expressed in the epidermis. This eQTL study highlights the potential to use biospecimens from clinical trials to discover in vivo eQTL interactions with therapeutically relevant environmental variables.
Collapse
Affiliation(s)
- Qian Xiao
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joseph Mears
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aparna Nathan
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Kazuyoshi Ishigaki
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, Japan
| | - Yuriy Baglaenko
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Noha Lim
- Immune Tolerance Network, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Laura A Cooney
- Immune Tolerance Network, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
- Division of Rheumatology, Department of Internal Medicine and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, USA
| | - Kristina M Harris
- Immune Tolerance Network, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Mark S Anderson
- Immune Tolerance Network, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - David A Fox
- Division of Rheumatology, Department of Internal Medicine and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, USA
| | - Dawn E Smilek
- Immune Tolerance Network, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA.
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK.
| |
Collapse
|
19
|
Silva-Abreu M, Sosa L, Espinoza LC, Fábrega MJ, Rodríguez-Lagunas MJ, Mallandrich M, Calpena AC, Garduño-Ramírez ML, Rincón M. Efficacy of Apremilast Gels in Mouse Model of Imiquimod-Induced Psoriasis Skin Inflammation. Pharmaceutics 2023; 15:2403. [PMID: 37896163 PMCID: PMC10610068 DOI: 10.3390/pharmaceutics15102403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Apremilast (APM) is a novel drug for the treatment of psoriasis and psoriatic arthritis. APM is a phosphodiesterase 4 (PDE4) inhibitor, raising intracellular cAMP levels and thereby decreasing the inflammatory response by modulating the expression of TNF-α, IL-17, IL-23, and other inflammatory cytokines. The goal of this study is to develop APM gels as a new pharmaceutical formulation for the treatment of topical psoriasis. APM was solubilized in Transcutol-P and incorporated into Pluronic F127, Sepigel, and carbomer bases at different proportions. All formulations were characterized physiochemically. A biopharmaceutical study (release profile) was performed, and ex vivo permeation was evaluated using a human skin model. A toxicity assay was carried out on the HaCaT cell line. A mouse model of imiquimod-induced psoriasis skin inflammation was carried out to determine its efficacy by histological analysis, RNA extraction, and RT-qPCR assays. APM gel formulations showed good physicochemical characteristics and a sustained release profile. There was no permeation of any gel measured through human skin, indicating a high retained amount of APM on the skin. Cell viability was greater than 80% at most dilution concentrations. APM gels treated the psoriasis mouse model, and it shows a reduction in the proinflammatory cytokines (IL-8, IL-17A, IL-17F, and IL-23). APM gels could be a new approach for the treatment of topical psoriasis.
Collapse
Affiliation(s)
- Marcelle Silva-Abreu
- Departament de Farmàcia, Tecnologia Farmacèutica, i Fisicoquímica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; (M.M.); (A.C.C.)
- Institut de Nanociència i Nanotecnologia IN2UB, University of Barcelona, 08028 Barcelona, Spain; (L.C.E.); (M.R.)
| | - Lilian Sosa
- Research Institute of Applied Sciences and Technology, National Autonomous University of Honduras (UNAH), Tegucigalpa 11101, Honduras;
- Microbiology Research Institute, National Autonomous University of Honduras (UNAH), Tegucigalpa 11101, Honduras
| | - Lupe Carolina Espinoza
- Institut de Nanociència i Nanotecnologia IN2UB, University of Barcelona, 08028 Barcelona, Spain; (L.C.E.); (M.R.)
- Departamento de Química y Ciencias Exactas, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador
| | - María-José Fábrega
- Department of Experimental and Health Sciences, Parc de Recerca Biomèdica de Barcelona, University Pompeu Fabra (UPF), 08005 Barcelona, Spain;
| | - María J. Rodríguez-Lagunas
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain;
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Barcelona, Spain
| | - Mireia Mallandrich
- Departament de Farmàcia, Tecnologia Farmacèutica, i Fisicoquímica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; (M.M.); (A.C.C.)
- Institut de Nanociència i Nanotecnologia IN2UB, University of Barcelona, 08028 Barcelona, Spain; (L.C.E.); (M.R.)
| | - Ana Cristina Calpena
- Departament de Farmàcia, Tecnologia Farmacèutica, i Fisicoquímica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; (M.M.); (A.C.C.)
- Institut de Nanociència i Nanotecnologia IN2UB, University of Barcelona, 08028 Barcelona, Spain; (L.C.E.); (M.R.)
| | - María Luisa Garduño-Ramírez
- Center for Chemical Research, Institute for Research Basic and Applied Sciences, Autonomous University of the State of Morelos, Av. Universidad 1001, Cuernavaca 62209, Mexico;
| | - María Rincón
- Institut de Nanociència i Nanotecnologia IN2UB, University of Barcelona, 08028 Barcelona, Spain; (L.C.E.); (M.R.)
- Departament de Ciència de Materials i Química Física, Facultat de Química, Universitat de Barcelona (UB), C. Martí i Franquès 1-11, 08028 Barcelona, Spain
| |
Collapse
|
20
|
Wu M, Dai C, Zeng F. Cellular Mechanisms of Psoriasis Pathogenesis: A Systemic Review. Clin Cosmet Investig Dermatol 2023; 16:2503-2515. [PMID: 37727872 PMCID: PMC10506593 DOI: 10.2147/ccid.s420850] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023]
Abstract
Psoriasis is a common inflammatory skin disease characterized by abnormal proliferation of epidermal keratinocytes and massive infiltration of inflammatory cells. Many kinds of cells, including keratinocytes, T lymphocytes, dendritic cells, neutrophils, and macrophages, are reported to play critical roles in the pathogenesis and progression of psoriasis. However, to date, the role of each kind of cell in the pathogenesis and development of psoriasis has not been systematically reviewed. In addition, although antibodies developed targeting cytokines (e.g. IL-23, IL-17A, and TNF-α) released by these cells have shown promising results in the treatment of psoriasis patients, these targeted antibodies still do not cure psoriasis and only provide short-term relief of symptoms. Furthermore, long-term use of these antibodies has been reported to have adverse physical and psychological effects on psoriasis patients. Therefore, gaining a deeper understanding of the cellular and molecular pathogenesis of psoriasis and providing new thoughts on the development of psoriasis therapeutic drugs is of great necessity. In this review, we summarize the roles of various cells involved in psoriasis, aiming to provide new insights into the pathogenesis and development of psoriasis at the cellular level and hoping to provide new ideas for exploring new and effective psoriasis treatments.
Collapse
Affiliation(s)
- Mengjun Wu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Chan Dai
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Fanfan Zeng
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| |
Collapse
|
21
|
Sosa L, Espinoza LC, Valarezo E, Bozal N, Calpena A, Fábrega MJ, Baldomà L, Rincón M, Mallandrich M. Therapeutic Applications of Essential Oils from Native and Cultivated Ecuadorian Plants: Cutaneous Candidiasis and Dermal Anti-Inflammatory Activity. Molecules 2023; 28:5903. [PMID: 37570874 PMCID: PMC10420932 DOI: 10.3390/molecules28155903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Essential oils are a complex mixture of aromatic substances whose pharmacological actions, including antimicrobial, antioxidant, anticancer, and anti-inflammatory activities, have been widely reported. This study aimed to evaluate the anti-Candida and dermal anti-inflammatory activity of essential oils from native and cultivated Ecuadorian plants. Essential oils from Bursera graveolens, Dacryodes peruviana, Mespilodaphne quixos, and Melaleuca armillaris were isolated by hydrodistillation and were characterized physically and chemically. Its tolerance was analyzed by in vitro and in vivo studies. The antifungal activity was studied against Candida albicans, Candida glabrata, and Candida parapsilosis, whereas the anti-inflammatory effect was evaluated by a mouse ear edema model. The main compounds were limonene, α-phellandrene, (E)-methyl cinnamate, and 1,8-cineole, respectively. All essential oils showed high tolerability for skin application, antifungal activity against the three Candida strains, and anti-inflammatory efficacy by decreasing edema and overexpression of pro-inflammatory cytokines. Dacryodes peruviana essential oil showed the highest antifungal activity. On the other hand, Dacryodes peruviana and Melaleuca armillaris showed the greatest anti-inflammatory potential, decreasing edema by 53.3% and 65.25%, respectively, and inhibiting the overexpression of TNF-α, IL-8, IL-17A, and IL-23. The results suggest that these essential oils could be used as alternative therapies in the treatment of both cutaneous candidiasis and dermal inflammation.
Collapse
Affiliation(s)
- Lilian Sosa
- Microbiological Research Institute (IIM), National Autonomous University of Honduras (UNAH), Tegucigalpa 11101, Honduras;
- Research Institute of Applied Sciences and Technology, National Autonomous University of Honduras (UNAH), Tegucigalpa 11101, Honduras
| | - Lupe Carolina Espinoza
- Departamento de Química, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador; (L.C.E.); (E.V.)
- Institut de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona (UB), 08028 Barcelona, Spain;
| | - Eduardo Valarezo
- Departamento de Química, Universidad Técnica Particular de Loja, Loja 1101608, Ecuador; (L.C.E.); (E.V.)
| | - Núria Bozal
- Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain;
| | - Ana Calpena
- Institut de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona (UB), 08028 Barcelona, Spain;
- Departament Farmàcia, Tecnologia Farmacèutica, i Físicoquímica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| | - María-José Fábrega
- Department of Experimental and Health Sciences, Parc of Biomedical Research of Barcelona, Pompeu Fabra University, 08003 Barcelona, Spain;
| | - Laura Baldomà
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain;
| | - María Rincón
- Departament de Ciència de Materials i Química Física, Facultat de Química, Universitat de Barcelona (UB), 08028 Barcelona, Spain;
| | - Mireia Mallandrich
- Institut de Nanociència i Nanotecnologia (IN2UB), Universitat de Barcelona (UB), 08028 Barcelona, Spain;
- Departament Farmàcia, Tecnologia Farmacèutica, i Físicoquímica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain
| |
Collapse
|
22
|
Jeong Y, Song J, Lee Y, Choi E, Won Y, Kim B, Jang W. A Transcriptome-Wide Analysis of Psoriasis: Identifying the Potential Causal Genes and Drug Candidates. Int J Mol Sci 2023; 24:11717. [PMID: 37511476 PMCID: PMC10380797 DOI: 10.3390/ijms241411717] [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: 05/31/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by cutaneous eruptions and pruritus. Because the genetic backgrounds of psoriasis are only partially revealed, an integrative and rigorous study is necessary. We conducted a transcriptome-wide association study (TWAS) with the new Genotype-Tissue Expression version 8 reference panels, including some tissue and multi-tissue panels that were not used previously. We performed tissue-specific heritability analyses on genome-wide association study data to prioritize the tissue panels for TWAS analysis. TWAS and colocalization (COLOC) analyses were performed with eight tissues from the single-tissue panels and the multi-tissue panels of context-specific genetics (CONTENT) to increase tissue specificity and statistical power. From TWAS, we identified the significant associations of 101 genes in the single-tissue panels and 64 genes in the multi-tissue panels, of which 26 genes were replicated in the COLOC. Functional annotation and network analyses identified that the genes were associated with psoriasis and/or immune responses. We also suggested drug candidates that interact with jointly significant genes through a conditional and joint analysis. Together, our findings may contribute to revealing the underlying genetic mechanisms and provide new insights into treatments for psoriasis.
Collapse
Affiliation(s)
- Yeonbin Jeong
- Department of Life Sciences, Dongguk University, Seoul 04620, Republic of Korea
| | - Jaeseung Song
- Department of Life Sciences, Dongguk University, Seoul 04620, Republic of Korea
| | - Yubin Lee
- Department of Life Sciences, Dongguk University, Seoul 04620, Republic of Korea
| | - Eunyoung Choi
- Department of Life Sciences, Dongguk University, Seoul 04620, Republic of Korea
| | - Youngtae Won
- Department of Life Sciences, Dongguk University, Seoul 04620, Republic of Korea
| | - Byunghyuk Kim
- Department of Life Sciences, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Wonhee Jang
- Department of Life Sciences, Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
23
|
Radulska A, Pelikant-Małecka I, Jendernalik K, Dobrucki IT, Kalinowski L. Proteomic and Metabolomic Changes in Psoriasis Preclinical and Clinical Aspects. Int J Mol Sci 2023; 24:ijms24119507. [PMID: 37298466 DOI: 10.3390/ijms24119507] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Skin diseases such as psoriasis (Ps) and psoriatic arthritis (PsA) are immune-mediated inflammatory diseases. Overlap of autoinflammatory and autoimmune conditions hinders diagnoses and identifying personalized patient treatments due to different psoriasis subtypes and the lack of verified biomarkers. Recently, proteomics and metabolomics have been intensively investigated in a broad range of skin diseases with the main purpose of identifying proteins and small molecules involved in the pathogenesis and development of the disease. This review discusses proteomics and metabolomics strategies and their utility in research and clinical practice in psoriasis and psoriasis arthritis. We summarize the studies, from in vivo models conducted on animals through academic research to clinical trials, and highlight their contribution to the discovery of biomarkers and targets for biological drugs.
Collapse
Affiliation(s)
- Adrianna Radulska
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland
| | - Iwona Pelikant-Małecka
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland
| | - Kamila Jendernalik
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland
| | - Iwona T Dobrucki
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405N Mathews Ave., MC-251, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 7 Debinki Street, 80-211 Gdansk, Poland
- BioTechMed Centre/Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Narutowicza Street, 80-233 Gdansk, Poland
| |
Collapse
|
24
|
Petrić M, Radić M. Is Th17-Targeted Therapy Effective in Systemic Lupus Erythematosus? Curr Issues Mol Biol 2023; 45:4331-4343. [PMID: 37232744 DOI: 10.3390/cimb45050275] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a broad spectrum of clinical manifestations. The proposed pathophysiological hypotheses of SLE are numerous, involving both innate and adaptive abnormal immune responses. SLE is characterized by the overproduction of different autoantibodies that form immune complexes, which cause damage in different organs. Current therapeutic modalities are anti-inflammatory and immunosuppressive. In the last decade, we have witnessed the development of many biologicals targeting different cytokines and other molecules. One of them is interleukin-17 (IL-17), a central cytokine of a proinflammatory process that is mediated by a group of helper T cells called Th17. Direct inhibitors of IL-17 are used in psoriatic arthritis, spondyloarthritis, and other diseases. Evidence about the therapeutic potential of Th17-targeted therapies in SLE is scarce, and probably the most promising is related to lupus nephritis. As SLE is a complex heterogeneous disease with different cytokines involved in its pathogenesis, it is highly unlikely that inhibition of only one molecule, such as IL-17, will be effective in the treatment of all clinical manifestations. Future studies should identify SLE patients that are eligible for Th17-targeted therapy.
Collapse
Affiliation(s)
- Marin Petrić
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, University Hospital of Split, Center of Excellence for Systemic Sclerosis Ministry of Health Republic of Croatia, Šoltanska 1, 21000 Split, Croatia
| | - Mislav Radić
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, University Hospital of Split, Center of Excellence for Systemic Sclerosis Ministry of Health Republic of Croatia, Šoltanska 1, 21000 Split, Croatia
- Department of Internal Medicine, School of Medicine, University of Split, Šoltanska 2, 21000 Split, Croatia
| |
Collapse
|
25
|
Torosian K, Lal E, Kavanaugh A, Loomba R, Ajmera V, Guma M. Psoriatic disease and non-alcoholic fatty liver disease shared pathogenesis review. Semin Arthritis Rheum 2023; 59:152165. [PMID: 36716599 PMCID: PMC9992353 DOI: 10.1016/j.semarthrit.2023.152165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/03/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023]
Abstract
Psoriatic disease (PD) and non-alcoholic fatty liver disease (NAFLD) potentially share disease pathways given the numerous inflammatory pathways involved in both diseases and a higher prevalence of NAFLD in PD patients. Metabolic syndrome and obesity are a key link between the two diseases, but even when controlling for this, associations between both diseases are still seen. Therapeutics that impact metabolic or inflammatory pathways may be impactful in both PD and NAFLD. In this review, we describe common inflammatory pathways contributing to both PD and NAFLD and critically review the potential impact of treatments for and on both diseases.
Collapse
Affiliation(s)
- Kelly Torosian
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Esha Lal
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Arthur Kavanaugh
- Department of Rheumatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; NAFLD Research Center, Department of Medicine, University of California at San Diego, La Jolla, USA; Division of Epidemiology, Department of Family and Preventative Medicine, University of California at San Diego, La Jolla, USA
| | - Veeral Ajmera
- Division of Gastroenterology and Hepatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; NAFLD Research Center, Department of Medicine, University of California at San Diego, La Jolla, USA.
| | - Monica Guma
- Department of Rheumatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona, Spain; San Diego VA Healthcare Service, San Diego, CA, 92161, USA.
| |
Collapse
|
26
|
Daniele SG, Eldirany SA, Ho M, Bunick CG. Structural basis for differential p19 targeting by IL-23 biologics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.09.531913. [PMID: 36945513 PMCID: PMC10029002 DOI: 10.1101/2023.03.09.531913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Background IL-23 is central to the pathogenesis of psoriasis, and is structurally comprised of p19 and p40 subunits. "Targeted" IL-23 inhibitors risankizumab, tildrakizumab, and guselkumab differ mechanistically from ustekinumab because they bind p19, whereas ustekinumab binds p40; however, a knowledge gap exists regarding the structural composition of their epitopes and how these molecular properties relate to their clinical efficacy. Objectives To characterize and differentiate the structural epitopes of the IL-23 inhibitors risankizumab, guselkumab, tildrakinumab, and ustekinumab, and correlate their molecular characteristics with clinical response in plaque psoriasis therapy. Methods We utilized epitope data derived from hydrogen-deuterium exchange studies for risankizumab, tildrakizumab, and guselkumab, and crystallographic data for ustekinumab to map drug epitope locations, hydrophobicity, and surface charge onto the IL-23 molecular surface (Protein Data Bank ID Code 3D87) using UCSF Chimera. PDBePISA was used to calculate solvent accessible surface area (SASA). Epitope composition was determined by classifying residues as acidic, basic, polar, or hydrophobic and calculating their contribution to epitope SASA. Linear regression and analysis of variance was performed. Results All the p19-specific inhibitor epitopes differ in location and size, with risankizumab and guselkumab having large epitope surface areas (SA), and tildrakizumab and ustekinumab having smaller SA. The tildrakizumab epitope was mostly hydrophobic (56%), while guselkumab, risankizumab, and ustekinumab epitopes displayed >50% non-hydrophobic residues. Risankizumab and ustekinumab exhibited acidic surface charges, while tildrakizumab and guselkumab were net neutral. Each inhibitor binds an epitope with a unique size and composition, and with mostly distinct locations except for a 10-residue overlap region that lies outside of the IL-23 receptor epitope. We observed a strong correlation between epitope SA and PASI-90 rates (R2 = 0.9969, p = 0.0016), as well as between epitope SA and KD (R2 = 0.9772, p = 0.0115). In contrast, we found that total epitope hydrophobicity, polarity, and charge content do not correlate with clinical efficacy. Conclusions Structural analysis of IL-23 inhibitor epitopes reveals strong association between epitope SA and early drug efficacy in plaque psoriasis therapy, exemplifying how molecular data can explain clinical observations, inform future innovation, and help clinicians in specific drug selection for patients.
Collapse
Affiliation(s)
- Stefano G. Daniele
- Medical Scientist Training Program (MD/PhD), Yale School of Medicine, New Haven, CT 06511, USA
| | - Sherif A. Eldirany
- Department of Dermatology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Minh Ho
- Department of Dermatology and Program in Translational Biomedicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Christopher G. Bunick
- Department of Dermatology and Program in Translational Biomedicine, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
27
|
The p300/CBP Inhibitor A485 Normalizes Psoriatic Fibroblast Gene Expression In Vitro and Reduces Psoriasis-Like Skin Inflammation In Vivo. J Invest Dermatol 2023; 143:431-443.e19. [PMID: 36174717 DOI: 10.1016/j.jid.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/23/2022]
Abstract
Psoriasis is a chronic inflammatory skin disease that often recurs at the same locations, indicating potential epigenetic changes in lesional skin cells. In this study, we discovered that fibroblasts isolated from psoriatic skin lesions retain an abnormal phenotype even after several passages in culture. Transcriptomic profiling revealed the upregulation of several genes, including the extra domain A splice variant of fibronectin and ITGA4 in psoriatic fibroblasts. A phenotypic library screening of small-molecule epigenetic modifier drugs revealed that selective CBP/p300 inhibitors were able to rescue the psoriatic fibroblast phenotype, reducing the expression levels of extra domain A splice variant of fibronectin and ITGA4. In the imiquimod-induced mouse model of psoriasis-like skin inflammation, systemic treatment with A485, a potent CBP/p300 blocker, significantly reduced skin inflammation, immune cell recruitment, and inflammatory cytokine production. Our findings indicate that epigenetic reprogramming might represent a new approach for the treatment and/or prevention of relapses of psoriasis.
Collapse
|
28
|
Habanjar O, Bingula R, Decombat C, Diab-Assaf M, Caldefie-Chezet F, Delort L. Crosstalk of Inflammatory Cytokines within the Breast Tumor Microenvironment. Int J Mol Sci 2023; 24:4002. [PMID: 36835413 PMCID: PMC9964711 DOI: 10.3390/ijms24044002] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Several immune and immunocompetent cells, including dendritic cells, macrophages, adipocytes, natural killer cells, T cells, and B cells, are significantly correlated with the complex discipline of oncology. Cytotoxic innate and adaptive immune cells can block tumor proliferation, and others can prevent the immune system from rejecting malignant cells and provide a favorable environment for tumor progression. These cells communicate with the microenvironment through cytokines, a chemical messenger, in an endocrine, paracrine, or autocrine manner. These cytokines play an important role in health and disease, particularly in host immune responses to infection and inflammation. They include chemokines, interleukins (ILs), adipokines, interferons, colony-stimulating factors (CSFs), and tumor necrosis factor (TNF), which are produced by a wide range of cells, including immune cells, such as macrophages, B-cells, T-cells, and mast cells, as well as endothelial cells, fibroblasts, a variety of stromal cells, and some cancer cells. Cytokines play a crucial role in cancer and cancer-related inflammation, with direct and indirect effects on tumor antagonistic or tumor promoting functions. They have been extensively researched as immunostimulatory mediators to promote the generation, migration and recruitment of immune cells that contribute to an effective antitumor immune response or pro-tumor microenvironment. Thus, in many cancers such as breast cancer, cytokines including leptin, IL-1B, IL-6, IL-8, IL-23, IL-17, and IL-10 stimulate while others including IL-2, IL-12, and IFN-γ, inhibit cancer proliferation and/or invasion and enhance the body's anti-tumor defense. Indeed, the multifactorial functions of cytokines in tumorigenesis will advance our understanding of cytokine crosstalk pathways in the tumor microenvironment, such as JAK/STAT, PI3K, AKT, Rac, MAPK, NF-κB, JunB, cFos, and mTOR, which are involved in angiogenesis, cancer proliferation and metastasis. Accordingly, targeting and blocking tumor-promoting cytokines or activating and amplifying tumor-inhibiting cytokines are considered cancer-directed therapies. Here, we focus on the role of the inflammatory cytokine system in pro- and anti-tumor immune responses, discuss cytokine pathways involved in immune responses to cancer and some anti-cancer therapeutic applications.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Rea Bingula
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Caroline Decombat
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Lebanon
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
29
|
Gracia-Cazaña T, Bernal-Masferrer L, Morales-Callaghan AM, Almenara-Blasco M, Gilaberte Y. Risankizumab for the Treatment of Moderate to Severe Psoriasis: Impact on Health-Related Quality of Life and Psychological Wellbeing. Clin Cosmet Investig Dermatol 2023; 16:221-229. [PMID: 36721838 PMCID: PMC9884449 DOI: 10.2147/ccid.s296544] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023]
Abstract
Biologic treatments are increasingly being used in the management of moderate to severe plaque psoriasis (PSO). Risankizumab (RZB) is a humanized monoclonal antibody that specifically blocks the p19 subunit of interleukin 23, which in turn regulates the activation, differentiation, and survival of Th17. RZB has proved their efficacy and their safety compared to anti-TNF. However, studies that assess and compare the improvement in other secondary PROs such as the patient's quality of life are still scarce. Health-related quality of life (HRQoL) is the sum of physical health, well-being, and participation; it defines the functional effect of a disease or its treatment and how it is perceived by the patient. The objective of this paper is to analyze the literature on the impact of treatment with RZB on the quality of life of patients with PSO and their psychological well-being. A bibliographic search was carried out to identify all the papers published from July 2015 to June 1, 2022, on RZB treatment in psoriasis and its impact on health-related quality of life and psychological well-being, finally twenty articles have been evaluated in full text, of which 8 were excluded because they did not meet the inclusion criteria. Risankizumab has shown not only to have very relevant data on effectiveness and safety, but all of this is associated with an improvement in quality of life related to health and psychological well-being measured on generic and specific quality of life scales, both in pivotal trials, ad hoc analysis, and data in real clinical practice.
Collapse
Affiliation(s)
- Tamara Gracia-Cazaña
- Dermatology Service, Hospital Miguel Servet, IIS Aragon, Zaragoza University, Zaragoza, Spain,Correspondence: Tamara Gracia-Cazaña, Dermatology Service, Hospital Miguel Servet, Zaragoza, Paseo Isabel la Católica 1-3, P.O.Box: 50009, Zaragoza, Spain, Tel +34 6657571403, Email
| | - Laura Bernal-Masferrer
- Dermatology Service, Hospital Miguel Servet, IIS Aragon, Zaragoza University, Zaragoza, Spain
| | | | - Manuel Almenara-Blasco
- Dermatology Service, Hospital Miguel Servet, IIS Aragon, Zaragoza University, Zaragoza, Spain
| | - Yolanda Gilaberte
- Dermatology Service, Hospital Miguel Servet, IIS Aragon, Zaragoza University, Zaragoza, Spain
| |
Collapse
|
30
|
Balato A, Scala E, Eyerich K, Brembilla NC, Chiricozzi A, Sabat R, Ghoreschi K. Management of Infections in Psoriatic Patients Treated with Systemic Therapies: A Lesson from the Immunopathogenesis of Psoriasis. Dermatol Pract Concept 2023; 13:dpc.1301a16. [PMID: 36892377 PMCID: PMC9946081 DOI: 10.5826/dpc.1301a16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Modern treatments continue to be developed based on identifying targets within the innate and adaptive immune pathways associated with psoriasis. Whilst there is a sound biologic rationale for increased risk of infection following treatment with immunomodulators, the clinical evidence is confounded by these agents being used in patients affected with several comorbidities. In an era characterized by an ever greater and growing risk of infections, it is necessary to always be updated on this risk. In this mini-review, we will discuss recent updates in psoriasis immunopathogenesis as a rationale for systemic therapy, outline the risk of infections linked to the disease itself and systemic therapy as well, and provide an overview of the prevention and management of infections.
Collapse
Affiliation(s)
- Anna Balato
- Dermatology Unit, University of Campania, Naples, Italy
| | - Emanuele Scala
- Division of Dermatology and Venereology, Department of Medicine Solna, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Dermatology and Venereology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kilian Eyerich
- Division of Dermatology and Venereology, Department of Medicine Solna, and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Dermatology and Venereology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Dermatology and Venereology, Unit of Dermatology, Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Robert Sabat
- Interdisciplinary Group of Molecular Immunopathology, Dermatology/Medical Immunology, Charité-Universitätsmedizin, Berlin, Germany.,Psoriasis Research and Treatment Center, Department of Dermatology and Allergy and Institute of Medical Immunology, Charité-Universitätsmedizin, Berlin, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin, Berlin, Germany
| |
Collapse
|
31
|
Expression of genes related to inflammation - IL-6, IL-8, and IFN-γ in monitoring ustekinumab therapy: preliminary results. Postepy Dermatol Alergol 2022; 39:1040-1047. [PMID: 36686017 PMCID: PMC9837596 DOI: 10.5114/ada.2022.122602] [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: 10/11/2021] [Accepted: 12/28/2021] [Indexed: 12/24/2022] Open
Abstract
Introduction Psoriasis is classified as an inflammatory and autoimmune disease. Changes in the concentration profile of some cytokines, such as interleukin-12 (IL-12), IL-23, and IL-17, play a key role in its pathogenesis. IL-6, IL-8 and interferon- γ (IFN-γ) are also hallmark cytokines in a psoriatic cytokine network. Cytokine-blocking drugs, which are a part of the inflammatory cascade, are now increasingly popular. One of them is ustekinumab, directed against IL-12 and IL-23, but also indirectly against other interleukins, which take part in the inflammatory reaction. Due to the complexity of inflammation pathways, new molecular markers are still being sought. Regardless of the type of therapy used, they allow to determine its effectiveness, signal the lack or loss of sensitivity to treatment. Aim To evaluate the expression profile of genes related to the inflammatory reaction - IL-6, IL-8, and IFN-γ - in patients with psoriasis, depending on the duration of ustekinumab therapy. Material and methods The material for the study was the PBMCs of 14 patients suffering from psoriasis who were treated with ustekinumab. Monitoring was performed after 16, 28, and 40 weeks of therapy. The gene expression of IL-6, IL-8, and IFN-γ was measured using the RT-qPCR method. Results There was a statistically significant increase in the expression of IL-6 and IFN-γ genes in psoriasis patients, depending on the duration of ustekinumab therapy. Conclusions The increase in mRNA copy numbers of the pro-inflammatory IL-6 and IFN-γ genes in the following weeks of therapy may suggest that patients treated with ustekinumab may progressively develop resistance to biological treatment.
Collapse
|
32
|
Parab S, Doshi G. An update on emerging immunological targets and their inhibitors in the treatment of psoriasis. Int Immunopharmacol 2022; 113:109341. [DOI: 10.1016/j.intimp.2022.109341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/27/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
33
|
Drakos A, Vender R. A Review of the Clinical Trial Landscape in Psoriasis: An Update for Clinicians. Dermatol Ther (Heidelb) 2022; 12:2715-2730. [PMID: 36319883 PMCID: PMC9674811 DOI: 10.1007/s13555-022-00840-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
As our understanding of the pathogenesis of psoriasis has evolved over the past two decades, so has the number of treatment options. The introduction of biologic agents targeting specific cytokines in the interleukin (IL)-23/IL-17 pathway has proven successful in promoting skin clearance among patients. However, their use is often limited owing to cost, parenteral administration, and possible reduced efficacy over time. Topical therapies have also seen limited advancement, with agents such as corticosteroids and vitamin D derivatives remaining the mainstay of treatment, despite side effects limiting their long-term use. New therapeutic agents are needed to improve disease management for patients. In this review, we summarize pipeline and recently approved therapies undergoing clinical trials for psoriasis during a 12-month search period (30 June 2021 to 30 June 2022) using ClinicalTrials.gov. New-generation biologics and oral small molecules in phase II or III development were included, and pivotal data identified through various search modalities (PubMed, conference presentations, etc.) evaluating each drug candidate will be discussed. Topical therapies will also be discussed in line with recent US Food and Drug Administration approvals. As new therapies continue to enter the treatment landscape, long-term data and comparative trials will be needed to better understand their place among existing therapeutic agents.
Collapse
Affiliation(s)
| | - Ronald Vender
- Dermatrials Research Inc., Venderm Innovations in Psoriasis, Hamilton, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
34
|
Mastorino L, Siliquini N, Avallone G, Zenone M, Ortoncelli M, Quaglino P, Dapavo P, Ribero S. Guselkumab shows high efficacy and maintenance in the improvement of response until week 48, a real-life study. Dermatol Ther 2022; 35:e15670. [PMID: 35762118 PMCID: PMC9786539 DOI: 10.1111/dth.15670] [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: 12/30/2021] [Revised: 06/07/2022] [Accepted: 06/25/2022] [Indexed: 12/30/2022]
Abstract
Guselkumab is an IL-23 inhibitor that has been demonstrated to be effective and safe for the treatment of moderate-to-severe plaque psoriasis in clinical trials. The data pool relating to the use of guselkumab in a real-life setting is still lacking. To evaluate the efficacy and safety of guselkumab in a real-life setting, focusing on predictors of early clinical response, a single-center prospective study was conducted enrolling patients with moderate-to-severe psoriasis. The clinical data relating to the efficacy and safety of the drug were acquired at initiation of treatment and at all subsequent clinical follow-ups: the primary endpoint was PASI90 and PASI100 response at week 12, 24, and 48. Out of the total cohort of 74 patients, 62 (83.8) reached a 48-week follow-up 64 (87.8%) reached a 24-week follow-up, while 72 (97.3%) a 12-week follow-up. Treatment with guselkumab reduced the mean PASI from the initial 11 ± 6.3 to 2.5 ± 3.1 at 12 weeks, to 1.2 ± 1.8 at 24 weeks, and to 0.8 ± 1.6 at 48 weeks. At week 12, a PASI 90 and PASI 100 response was achieved by 44.4% and 23.6% of patients, respectively. After 24 weeks, 63% of patients reported a PASI 90 while 46.1% achieved PASI 100. Previous treatment with one or more other biologics did not impact significantly on the achievement of the PASI 90 and 100 at any endpoints analyzed. We reported no difference between bio-naïve and non-naïve patients in the response to guselkumab, high safety, and efficacy was showed in both populations.
Collapse
Affiliation(s)
- Luca Mastorino
- Dermatologic Clinic, Department of Medical SciencesUniversity of TurinTurinItaly
| | - Niccolò Siliquini
- Dermatologic Clinic, Department of Medical SciencesUniversity of TurinTurinItaly
| | - Gianluca Avallone
- Dermatologic Clinic, Department of Medical SciencesUniversity of TurinTurinItaly
| | - Mattia Zenone
- Dermatologic Clinic, Department of Medical SciencesUniversity of TurinTurinItaly
| | - Michela Ortoncelli
- Dermatologic Clinic, Department of Medical SciencesUniversity of TurinTurinItaly
| | - Pietro Quaglino
- Dermatologic Clinic, Department of Medical SciencesUniversity of TurinTurinItaly
| | - Paolo Dapavo
- Dermatologic Clinic, Department of Medical SciencesUniversity of TurinTurinItaly
| | - Simone Ribero
- Dermatologic Clinic, Department of Medical SciencesUniversity of TurinTurinItaly
| |
Collapse
|
35
|
Odnopozova L, Edin A, Sukharev A, Wu T, Aydin K, Kelly M, Khotko A. Risankizumab for the Treatment of Moderate to Severe Plaque Psoriasis in the Russian Federation. Dermatol Ther (Heidelb) 2022; 12:2063-2075. [PMID: 35917057 PMCID: PMC9464287 DOI: 10.1007/s13555-022-00776-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/12/2022] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Risankizumab has demonstrated efficacy and safety in phase 3 studies in patients with moderate to severe plaque psoriasis. This randomized clinical trial assessed the efficacy and safety of risankizumab in patients with moderate to severe plaque psoriasis in the Russian Federation. METHODS Patients with moderate to severe plaque psoriasis were randomized 4:1 to 16 weeks of double-blind treatment with risankizumab 150 mg or placebo (period A; dosing at baseline and week 4) followed by an open-label extension (period B) during which all patients received risankizumab 150 mg at weeks 16, 28, and 40 and were followed up to week 52. The primary study endpoint was the proportion of patients achieving ≥ 90% improvement in the Psoriasis Area and Severity Index (PASI 90) at week 16, and secondary endpoints included Static Physician's Global Assessment scores and the Dermatology Life Quality Index. Treatment-emergent adverse events were monitored throughout the two study periods. RESULTS Of the 50 patients who entered period A, 41 were randomized to receive risankizumab and 9 to receive placebo. Forty-eight patients entered period B, and 47 completed the study. A significantly larger proportion of risankizumab-treated patients achieved PASI 90 at week 16 compared with placebo-treated patients [response rate difference: 38.8% (95% CI 7.8-69.7%; P = 0.035)]. Consistently higher proportions of risankizumab-treated patients achieved secondary endpoints compared with the placebo-treated patients. Safety profiles were similar between the treatment groups, and no patients discontinued the study drug owing to adverse events. CONCLUSION Risankizumab was efficacious and well tolerated in patients with moderate to severe plaque psoriasis in the Russian Federation. TRIAL REGISTRATION ClinicalTrials.gov NCT03518047.
Collapse
Affiliation(s)
- Liudmila Odnopozova
- Alliance Biomedical Ural Group, 89 ul. 10 Let Oktyabrya st, Izhevsk, Udmurtskaya Respublika, 426061, Russian Federation.
| | - Anton Edin
- LLC ArsVitae Severo-Zapad, St. Petersburg, Russia
| | - Alexey Sukharev
- Clinic of Dermatology and Venerology, St. Petersburg, Russia
| | | | - Kerstin Aydin
- AbbVie Deutschland GmbH & Co KG, Ludwigshafen, Germany
| | | | - Alkes Khotko
- GBUZ Clinical Dermatovenerology, Dispensary of MoH of Krasnodar Region, Krasnodar, Russia
| |
Collapse
|
36
|
Purwoko M, Indarto D, Kariosentono H, Purwanto B, Soetrisno S, Cilmiaty R. Chloroform Extract of Plumbago zeylanica Linn. Roots Ameliorates the Epidermal Thickness of Imiquimod-induced Psoriatic Mice through Cell Cycle and Apoptosis. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Introduction: Psoriasis vulgaris is a chronic skin disease which is characterized by recurrent scales on skin. The global prevalence of this disease has increased in ten years. Plumbagin is an active compound in the P. zeylanica Linn. Some recent studies revealed that P. zeylanica Linn extracts have the antiproliferative activity, which is used for treatment of some human diseases. The aim of this study was to investigated the effect of Chloroform extract of P. zeylanica Linn roots (CEP) on epidermal thickness of Imiquimod-induced psoriatic mice.
Methods: This was a post-test only control group design. A total of 42 male BALB/c mice was divided into six groups. Mice in treatment groups orally received 25, 50, and 100 mg/kg body weight CEP, respectively while positive control orally received 1 mg/kg body weight Methotrexate for seven days. Evaluation of epidermal thickness based on histological changes, serum IL-23 level by ELISA, and Cyclin-dependent kinase 2, Cyclin A, and Caspase-3 expressions by immunohistochemistry.
Results: Administrations of CEP decreased the epidermal thickness of psoriatic plaques in all treatment groups (p = 0.002, 0.003, and 0.016 respectively) compared to negative control but it did not reduce the serum IL-23 level. The expressions of CDK2 and Cyclin A reduced in T2 and T3 groups and the expression of Caspase-3 increased was only in T3 group.
Conclusion: Chloroform extract of P. zeylanica Linn roots administrations reduce the epidermal thickness of Imiquimod-induced psoriatic mice by inhibition of keratinocyte cell cycle and induction of Caspase-3 expression.
Collapse
|
37
|
Wei NW, Chi S, Lebwohl MG. Retrospective Analysis in Patients With Moderate to Severe Plaque Psoriasis Treated With Tildrakizumab: Real-Life Clinical Data. JOURNAL OF PSORIASIS AND PSORIATIC ARTHRITIS 2022; 7:55-59. [PMID: 39296827 PMCID: PMC11361525 DOI: 10.1177/24755303221077211] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Background: Interleukin (IL)-23 antagonists have shown great efficacy with minimal side effect profile in clinical trial data for the treatment of moderate to severe plaque psoriasis. As of yet, there have been no published data regarding real-world patients who have received tildrakizumab therapy. Objectives: To analyze real-world efficacy and safety of tildrakizumab in patients with moderate to severe plaque psoriasis. Methods: A retrospective chart review was performed at a large urban academic medical center for all patients treated with tildrakizumab for moderate to severe plaque psoriasis. Demographic information, Psoriasis Area and Severity Indexs(PASIs) from initial presentation and 12-month follow-up, comorbidities, and any possible adverse events were collected and analyzed statistically. Results: 30 patients on tildrakizumab therapy were included in the analysis. Overall, the mean ± standard deviation of the PASIs was 15.8 ± 11.8 at initial visit and 1.5 ± 2.9 at 12-month follow-up (P < .001). No serious adverse events were reported. Conclusions: Tildrakizumab has shown efficacy in clinical trials and this real-world cohort for the treatment of moderate to severe plaque psoriasis with a good safety profile. Future studies should be done to assess the efficacy of tildrakizumab compared with other IL-23 inhibitors.
Collapse
Affiliation(s)
- Nancy W Wei
- The Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Sunny Chi
- The Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Mark G Lebwohl
- The Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
38
|
Mohd Noor AA, Azlan M, Mohd Redzwan N. Orchestrated Cytokines Mediated by Biologics in Psoriasis and Its Mechanisms of Action. Biomedicines 2022; 10:biomedicines10020498. [PMID: 35203707 PMCID: PMC8962336 DOI: 10.3390/biomedicines10020498] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 12/27/2022] Open
Abstract
Psoriasis is an autoimmune disease mediated by disturbed T cells and other immune cells, and is defined by deep-red, well-demarcated skin lesions. Due to its varied etiologies and indefinite standard pathogenesis, it is challenging to consider the right treatment exclusively for each psoriasis patient; thus, researchers yearn to seek even more precise treatments other than topical treatment and systemic therapy. Using biologics to target specific immune components, such as upregulated cytokines secreted by activated immune cells, is the most advanced therapy for psoriasis to date. By inhibiting the appropriate pro-inflammatory cytokines, cellular signaling can be altered and, thus, can inhibit further downstream inflammatory pathways. Herein, the roles of cytokines with their mechanisms of action in progressing psoriasis and how the usage of biologics alleviates cellular inflammation are discussed. In addition, other potential pro-inflammatory cytokines, with their mechanism of action, are presented herein. The authors hope that this gathered information may benefit future research in expanding the discovery of targeted psoriasis therapy.
Collapse
Affiliation(s)
- Aina Akmal Mohd Noor
- Immunology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia;
| | - Maryam Azlan
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia;
| | - Norhanani Mohd Redzwan
- Immunology Department, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia;
- Correspondence: ; Tel.: +60-9767-6130
| |
Collapse
|
39
|
Gooderham M, Pinter A, Ferris LK, Warren RB, Zhan T, Zeng J, Soliman AM, Kaufmann C, Kaplan B, Photowala H, Strober B. Long-Term, Durable, Absolute Psoriasis Area and Severity Index and Health-Related Quality of Life Improvements with Risankizumab Treatment: A Post-Hoc Integrated Analysis of Patients with Moderate-to-Severe Plaque Psoriasis. J Eur Acad Dermatol Venereol 2022; 36:855-865. [PMID: 35174556 PMCID: PMC9314097 DOI: 10.1111/jdv.18010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 12/22/2021] [Accepted: 01/19/2022] [Indexed: 11/29/2022]
Abstract
Background Risankizumab has demonstrated durable, high rates of efficacy in patients with moderate‐to‐severe plaque psoriasis as assessed by the achievement of relative Psoriasis Area and Severity Index (PASI) improvement and Dermatology Life Quality Index (DLQI) 0/1. Objectives The aim of this post hoc analysis is to assess the achievement of absolute PASI thresholds and related improvements in health‐related quality of life (HRQoL) in patients with moderate‐to‐severe plaque psoriasis treated with (i) risankizumab compared with ustekinumab, and (ii) long‐term (>52 weeks to 172 weeks) risankizumab. Methods Data from patients randomised to 150 mg risankizumab or 45 or 90 mg ustekinumab in replicate randomised controlled trials UltIMMa‐1 and UltIMMa‐2 were analysed for the achievement of absolute PASI thresholds PASI ≤ 3, PASI ≤ 1, and PASI = 0, time to achieve these thresholds, and combined PASI and DLQI endpoints. Data from pat ients initially randomised to risankizumab who continued on risankizumab in the open‐label extension study LIMMitless were analysed for the achievement of absolute PASI levels, mean DLQI scores, and DLQI 0/1. Results Significantly greater proportions of patients treated with risankizumab compared with ustekinumab achieved PASI ≤ 3, PASI ≤ 1, and PASI = 0, as well as combined endpoints for absolute PASI and DLQI [(PASI ≤ 3 and DLQI ≤ 5) or (PASI ≤ 1 and DLQI 0/1)]. The median time to first achieve PASI ≤ 3, PASI ≤ 1, and PASI = 0 was significantly lower for risankizumab‐treated patients compared with ustekinumab‐treated patients. Among patients treated with long‐term risankizumab, more than 90% achieved PASI ≤ 3 though week 172 and more than 80% achieved DLQI 0/1. Low absolute PASI scores corresponded with low mean absolute DLQI scores through week 172 of continuous risankizumab treatment. Conclusions Risankizumab treatment demonstrated high rates of rapid and durable efficacy as measured by absolute PASI thresholds and improvements in patient HRQoL.
Collapse
Affiliation(s)
- Melinda Gooderham
- SKiN Centre for Dermatology, Probity Medical Research and Queen's University, Peterborough, Canada
| | - Andreas Pinter
- University Hospital Frankfurt am Main, Dept. of Dermatology, Frankfurt am Main, Germany
| | - Laura K Ferris
- University of Pittsburgh, Department of Dermatology, Pittsburgh, Pennsylvania, USA
| | - Richard B Warren
- The Dermatology Centre, Salford Royal NHS Foundation Trust, Manchester NIHR Biomedical Research Centre, University of Manchester, UK
| | | | | | | | | | | | | | - Bruce Strober
- Yale University, New Haven, CT and Central Connecticut Dermatology Research, Cromwell, CT, USA
| |
Collapse
|
40
|
Phuna ZX, Madhavan P. A CLOSER LOOK AT THE MYCOBIOME IN ALZHEIMER'S DISEASE: FUNGAL SPECIES, PATHOGENESIS AND TRANSMISSION. Eur J Neurosci 2022; 55:1291-1321. [DOI: 10.1111/ejn.15599] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Zhi Xin Phuna
- School of Medicine, Faculty of Health & Medical Sciences, Taylor’s University Malaysia Subang Jaya Selangor
| | - Priya Madhavan
- School of Medicine, Faculty of Health & Medical Sciences, Taylor’s University Malaysia Subang Jaya Selangor
| |
Collapse
|
41
|
Fernández-Ruiz M. Interleukin-12 and -23 Targeted Agents. INFECTIOUS COMPLICATIONS IN BIOLOGIC AND TARGETED THERAPIES 2022:199-217. [DOI: 10.1007/978-3-031-11363-5_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
42
|
Abstract
INTRODUCTION Psoriasis is a chronic immune-mediated disease affecting 125 million people globally. It is characterized by erythematous plaques in the skin, covered by silvery scales. However, non-cutaneous manifestations (e.g., joint symptoms in psoriatic arthritis) and a high prevalence of other immune-mediated diseases such as inflammatory bowel diseases reflect its systemic nature. So far, research on psoriasis pathogenesis has improved our knowledge of the roles of the immune system, and cytokines play significant roles in immune responses. AREAS COVERED Herein, we review cytokine changes in psoriasis patients. Moreover, we will investigate the possible relationships between disease severity and cytokines alongside describing cytokine alterations in psoriasis patients with other comorbidities. Lastly, we will discuss the biologics and their effects on cytokines in psoriasis patients. EXPERT OPINION Psoriasis could develop various clinical types and clinical manifestations in people. It is an immune-mediated disease, and these manifestations are associated with different impaired cytokines. Imbalanced cytokines could lead to abnormal keratinocytes, neovascularization, and inflammation in psoriasis patients. So, a better understanding of the cytokine roles can help one choose a specific cytokine-targeting biologic to treat psoriasis. Moreover, these cytokines may be used as a severity marker for following up with these patients.
Collapse
Affiliation(s)
- Homa Seyedmirzaei
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Tehran, Iran.,Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Harris KM, Smilek DE, Byron M, Lim N, Barry WT, McNamara J, Garcet S, Konrad RJ, Stengelin M, Bathala P, Korman NJ, Feldman SR, Boh EE, Barber K, Laumann AE, Helfrich YR, Krueger GG, Sofen H, Bissonnette R, Krueger JG. Effect of Costimulatory Blockade With Abatacept After Ustekinumab Withdrawal in Patients With Moderate to Severe Plaque Psoriasis: The PAUSE Randomized Clinical Trial. JAMA Dermatol 2021; 157:1306-1315. [PMID: 34643650 PMCID: PMC8515260 DOI: 10.1001/jamadermatol.2021.3492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Question Does blockade of CD28/B7 costimulatory signaling with abatacept suppress the psoriasis molecular signature and prevent psoriasis relapse after ustekinumab withdrawal? Findings In this parallel-design, double-blind randomized clinical trial of 91 adults with moderate to severe plaque psoriasis, costimulatory blockade with abatacept did not prevent psoriasis relapse and did not maintain suppression of the pathogenic psoriasis molecular signature following ustekinumab withdrawal. Meaning In this study, abatacept did not prevent psoriasis relapse, which may rely on alternative, compensatory mechanisms of residual T-cell activation in skin. Importance Psoriasis relapse may involve compensatory T-cell activation pathways in the presence of CD28-CD80/CD86 blockade with abatacept. Objective To determine whether costimulatory signaling blockade with abatacept prevents psoriasis relapse after ustekinumab withdrawal. Design, Setting, and Participants Psoriasis Treatment with Abatacept and Ustekinumab: a Study of Efficacy (PAUSE), a parallel-design, double-blind, placebo-controlled randomized clinical trial, was conducted at 10 sites in the US and Canada. Participant enrollment opened on March 19, 2014, and concluded on April 11, 2016. Participants were adults with moderate to severe plaque psoriasis and received ustekinumab in a lead-in phase. Those who responded to ustekinumab at week 12 were randomized 1:1 to either the continued with ustekinumab group (ustekinumab group) or the switched to abatacept group (abatacept group). Treatment was discontinued at week 39, and participants were followed up for psoriasis relapse until week 88. Statistical analyses were performed in the intention-to-treat (ITT) and safety samples from May 3, 2018, to July 6, 2021. Interventions Participants received subcutaneous ustekinumab at weeks 0 and 4 (45 mg per dose for those ≤100 kg; 90 mg per dose for those >100 kg). Participants randomized to the abatacept group at week 12 received subcutaneous abatacept, 125 mg weekly, from weeks 12 to 39 and ustekinumab placebo at weeks 16 and 28. Participants randomized to the ustekinumab group received ustekinumab at weeks 16 and 28 and abatacept placebo weekly from weeks 12 to 39. Main Outcomes and Measures The primary end point was the proportion of participants with psoriasis relapse (loss of ≥50% of the initial Psoriasis Area and Severity Index improvement) between weeks 12 and 88. Secondary end points included time to psoriasis relapse, proportion of participants with psoriasis relapse between weeks 12 and 40, and adverse events. The psoriasis transcriptome and serum cytokines were evaluated. Results A total of 108 participants (mean [SD] age, 46.1 [12.1] years; 73 [67.6%] men) were treated with open-label ustekinumab; 91 were randomized to blinded treatment. Similar proportions of participants in the abatacept group and the ustekinumab group relapsed between weeks 12 and 88 (41 of 45 [91.1%] vs 40 of 46 [87.0%]; P = .41). Median time to relapse from the last dose of ustekinumab was similar between groups as well: 36 weeks (95% CI, 36-48 weeks) in the abatacept group vs 32 weeks (95% CI, 28-40 weeks) in the ustekinumab group. Similar numbers and rates of adverse events occurred. Abatacept did not maintain suppression of the pathogenic IL-23-mediated psoriasis molecular signature in lesions after ustekinumab withdrawal, and serum IL-19 levels increased. Conclusions and Relevance This parallel-design, double-blind randomized clinical trial found that abatacept did not prevent psoriasis relapse that occurred after ustekinumab withdrawal because it did not completely block the pathogenic psoriasis molecular pathways that led to relapse. Trial Registration ClinicalTrials.gov Identifier: NCT01999868
Collapse
Affiliation(s)
- Kristina M Harris
- Biomarker and Discovery Research, Immune Tolerance Network, University of California, San Francisco, San Francisco
| | - Dawn E Smilek
- Clinical Trials Group, Clinical and Translational Medicine, Immune Tolerance Network, University of California, San Francisco, San Francisco
| | | | - Noha Lim
- Biomarker and Discovery Research, Immune Tolerance Network, University of California, San Francisco, San Francisco
| | | | - James McNamara
- Autoimmunity and Mucosal Immunology Branch, Division of Allergy, Immunology, and Transplantation/National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | | | - Robert J Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | | | | | - Neil J Korman
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Steven R Feldman
- Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Erin E Boh
- Health Sciences Center, Tulane University School of Medicine, New Orleans, Louisiana
| | - Kirk Barber
- Department of Medicine (Dermatology), University of Calgary, Calgary, Alberta, Canada
| | - Anne E Laumann
- Department of Dermatology, Northwestern University, Colorado Springs, Colorado
| | | | - Gerald G Krueger
- Department of Dermatology, University of Utah School of Medicine, Salt Lake City
| | - Howard Sofen
- Dermatology, David Geffen UCLA (University of California, Los Angeles) School of Medicine, Los Angeles
| | | | | |
Collapse
|
44
|
Geyer CE, Newling M, Sritharan L, Griffith GR, Chen HJ, Baeten DLP, den Dunnen J. C-Reactive Protein Controls IL-23 Production by Human Monocytes. Int J Mol Sci 2021; 22:ijms222111638. [PMID: 34769069 PMCID: PMC8583945 DOI: 10.3390/ijms222111638] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023] Open
Abstract
C-reactive protein (CRP) is an acute-phase protein in humans that is produced in high quantities by the liver upon infection and under inflammatory conditions. Although CRP is commonly used as a marker of inflammation, CRP can also directly contribute to inflammation by eliciting pro-inflammatory cytokine production by immune cells. Since CRP is highly elevated in serum under inflammatory conditions, we have studied the CRP-induced cytokine profile of human monocytes, one of the main innate immune cell populations in blood. We identified that CRP is relatively unique in its capacity to induce production of the pro-inflammatory cytokine IL-23, which was in stark contrast to a wide panel of pattern recognition receptor (PRR) ligands. We show that CRP-induced IL-23 production was mediated at the level of gene transcription, since CRP particularly promoted gene transcription of IL23A (encoding IL-23p19) instead of IL12A (encoding IL-12p35), while PRR ligands induce the opposite response. Interestingly, when CRP stimulation was combined with PRR ligand stimulation, as for example, occurs in the context of sepsis, IL-23 production by monocytes was strongly reduced. Combined, these data identify CRP as a unique individual ligand to induce IL-23 production by monocytes, which may contribute to shaping systemic immune responses under inflammatory conditions.
Collapse
Affiliation(s)
- Chiara E. Geyer
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Melissa Newling
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (M.N.); (L.S.); (D.L.P.B.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Lathees Sritharan
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (M.N.); (L.S.); (D.L.P.B.)
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Guillermo R. Griffith
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (G.R.G.); (H.-J.C.)
| | - Hung-Jen Chen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (G.R.G.); (H.-J.C.)
| | - Dominique L. P. Baeten
- Department of Rheumatology & Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (M.N.); (L.S.); (D.L.P.B.)
| | - Jeroen den Dunnen
- Center for Experimental and Molecular Medicine, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Correspondence: ; Tel.: +31-205668043
| |
Collapse
|
45
|
Yadav K, Singh D, Singh MR. Novel archetype in psoriasis management bridging molecular dynamics in exploring novel therapies. Eur J Pharmacol 2021; 907:174254. [PMID: 34118225 DOI: 10.1016/j.ejphar.2021.174254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/21/2022]
Abstract
Psoriasis is an autoimmune chronic inflammatory condition of skin affecting 125 million populaces around the globe. It is implicated as a result of multifaceted phenomena involving various cell and subcell activities with the aid of numerous cellular and molecular components including signaling aisle and regulatory proteins owing to the development of such hyperproliferative dermatological conditions. This involves a deeply complex and conflicting pathology owing to genetic and immunological deviations resulting from the unusual presentation of different signaling pathways and regulatory proteins. Explorations of these biomarkers and intervention of molecular and cellular processes in psoriasis are yet to be investigated and could be an exceptional aspect for understanding pathology with successful targeting of disease. In the presented study, we have integrated molecular insights, including signaling molecules, pathways, and proteins implicated in pathogenesis, and we have attempted to link this knowledge to the targeting of these phenomena in order to manage the conditions precisely. Further, therapeutic delivery approaches for targeting distinct layers of skin have also been investigated based on the application of different nanocarriers for successful psoriasis treatment.
Collapse
Affiliation(s)
- Krishna Yadav
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492010, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492010, India
| | - Manju Rawat Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492010, India.
| |
Collapse
|
46
|
Eyerich K, Weisenseel P, Pinter A, Schäkel K, Asadullah K, Wegner S, Muñoz-Elias EJ, Bartz H, Taut FJH, Reich K. IL-23 blockade with guselkumab potentially modifies psoriasis pathogenesis: rationale and study protocol of a phase 3b, randomised, double-blind, multicentre study in participants with moderate-to-severe plaque-type psoriasis (GUIDE). BMJ Open 2021; 11:e049822. [PMID: 34518264 PMCID: PMC8438891 DOI: 10.1136/bmjopen-2021-049822] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Guselkumab is an interleukin (IL)-23 pathway blocker with proven efficacy in patients with moderate-to-severe plaque psoriasis. Early intervention with guselkumab may result in changes to the clinical disease course versus later intervention. METHODS AND ANALYSIS Here we present the rationale and design of a phase 3b, randomised, double-blind, multicentre study (GUIDE), comparing treatment effects of guselkumab in patients with short (≤2 years) or longer (>2 years) duration of plaque-type psoriasis, measured from first appearance of psoriatic plaques. Participants achieving skin clearance (Psoriasis Area and Severity Index (PASI)=0) by week 20 and maintaining complete clearance at week 28 visit ('super-responders' (SRe)) will be randomised to continue approved maintenance dosing every 8 weeks (q8w) versus an investigational maintenance dosing interval of 16 weeks (q16w) until week 68. Primary endpoint: proportion of participants in the q8w vs q16w arms with absolute PASI <3 at week 68. Participants with PASI <3 at week 68 will be withdrawn from guselkumab treatment for up to 48 weeks. Participants not achieving SRe criteria (non-SRe) will remain in the study with q8w guselkumab dosing through week 68. Additional to serum samples obtained from all patients, skin biopsies and whole-blood samples will be taken from SRe and non-SRe participants at various time points in optional substudies. Analyses include: genetics; immunophenotyping (fluorescence-activated cell sorting); gene and protein expression profiling; immunohistology. By merging clinical endpoints with mechanistic findings, this study aims to elucidate how IL-23 blockade with guselkumab can modify the disease course by altering molecular and cellular drivers that cause relapse after treatment withdrawal, particularly among SRe. ETHICS AND DISSEMINATION Approval obtained from ethics committee Medical Council Hamburg, Germany (PVN5925). GUIDE is compliant with the Declaration of Helsinki. TRIAL REGISTRATION NUMBER Registered at ClinicalTrials.gov (NCT03818035). All primary endpoint results (prespecified analyses) will be submitted to peer-reviewed, international journals within 18 months after primary completion date.
Collapse
Affiliation(s)
- Kilian Eyerich
- Department of Medicine, Division of Dermatology and Venereology, Karolinska Institute, Stockholm, Sweden
| | | | - Andreas Pinter
- University Hospital Frankfurt am Main, Frankfurt, Germany
| | - Knut Schäkel
- Department of Dermatology, and Interdisciplinary Center for Chronic Inflammatory Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | - Kristian Reich
- Institute for Health Services Research in Dermatology and Nursing, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
47
|
Chan TC, Lee MS, Huang WC, Chang WY, Krueger JG, Tsai TF. Capsaicin attenuates imiquimod-induced epidermal hyperplasia and cutaneous inflammation in a murine model of psoriasis. Biomed Pharmacother 2021; 141:111950. [PMID: 34328106 DOI: 10.1016/j.biopha.2021.111950] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/21/2022] Open
Abstract
Psoriasis is one of the most common chronic inflammatory diseases that is characterized by well-defined erythematous plaques, with typical histopathological findings of lymphocytic infiltration and epidermal hyperplasia. Topical treatments of psoriasis are either associated with limited response or with side effects. Up to date, topicals targeting neuroimmune axis in psoriasis or psoriasiform dermatitis have not been explored. Here, we investigated whether percutaneous delivery of capsaicin could attenuate the pathological change of psoriasiform inflammation. Imiquimod-induced psoriasis-like murine model was used to evaluate therapeutic effects from topical application of capsaicin. An additional model of psoriasiform dermatitis induced by direct IL-23 injection was used to identify the level of action from capsaicin in this neuroimmune axis. Cutaneous inflammation was assessed by erythema level and ear thickness change. Key cytokines, infiltrating cells in the skin, and draining lymph node cells were investigated. The results showed that capsaicin administration obstructed the activation of IL-23/IL-17 pathway induced by imiquimod, presenting with significantly reduced psoriasiform dermatitis both in gross appearance and microscopic features. Tissue gene expression of psoriatic core cytokines induced by imiquimod (including IL-23, IL-17A, IL-22, TNF-α, and IL-6) were greatly decreased by capsaicin application. This protective effect from capsaicin could be hampered by direct intradermal injection of IL-23. CONCLUSION: Epicutaneous delivery of capsaicin on imiquimod-treated murine skin could significantly decrease expression of multiple inflammatory cytokines and the severity of prototypic change of psoriasiform inflammation. The beneficial effect imposed by capsaicin reinforces the neuroimmune contribution towards psoriasiform inflammation and provides a potential non-steroidal therapeutic alternative for topical treatment of psoriasiform dermatitis.
Collapse
Affiliation(s)
- Tom C Chan
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Meng-Sui Lee
- Department of Dermatology, Taipei City Hospital, Taipei, Taiwan; Department of Dermatology, National Yang-Ming University, Taipei, Taiwan
| | - Wen-Chih Huang
- Department of Anatomic Pathology, Far Eastern Memorial Hospital, New Taipei, Taiwan; Department of Anatomical Pathology, Taipei Institute of Pathology, Taipei, Taiwan
| | - Wen-Yu Chang
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Department of Dermatology, E-Da Cancer Hospital, Kaohsiung, Taiwan
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
48
|
Elevated Serum Interleukin-23 Levels in Patients with Oral and Cutaneous Lichen Planus. Mediators Inflamm 2021; 2021:5578568. [PMID: 34335090 PMCID: PMC8289569 DOI: 10.1155/2021/5578568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/03/2021] [Indexed: 12/21/2022] Open
Abstract
Lichen planus is considered a chronic inflammatory disease which affects different sites, such as the skin, mucous membranes, hair, and nails. Based on the evidence, a complex cytokine network plays a crucial role in lichen planus pathogenesis. The study was aimed at assessing the serum IL-23 levels in the patients with cutaneous and oral lichen planus compared to healthy controls. Method. The study included 30 cutaneous lichen planus patients, 20 oral lichen planus patients, and 33 control subjects. Five milliliters of peripheral blood was obtained from each patient, and the serum was separated. IL-23 levels were determined using the ELISA kit, and the data were analyzed using the Mann–Whitney test. Results. IL-23 levels in the patient serum with oral lichen planus (P value ≤ 0.001) were significantly higher than in controls. Furthermore, there were significant differences in IL-23 serum levels in the patients with cutaneous lichen planus compared to the healthy controls (P value ≤ 0.001). Moreover, IL-23 serum levels were statistically different between patients with cutaneous lichen planus and patients with oral lichen planus (P value ≤ 0.001). Based on the mean concentration of interleukin-23, IL-23 levels were higher in the patients with oral lichen planus than in the patients with cutaneous lichen planus. Conclusions. Elevated serum IL-23 levels in the patients with oral lichen planus may indicate that IL-23 plays a crucial role in the pathogenesis of oral lichen planus. However, more research is needed with a larger sample size.
Collapse
|
49
|
Shi Z, Garcia-Melchor E, Wu X, Getschman AE, Nguyen M, Rowland DJ, Wilson M, Sunzini F, Akbar M, Huynh M, Law T, Kundu-Raychaudhuri SK, Raychaudhuri SP, Volkman BF, Millar NL, Hwang ST. Targeting the CCR6/CCL20 axis in entheseal and cutaneous inflammation. Arthritis Rheumatol 2021; 73:2271-2281. [PMID: 34081845 DOI: 10.1002/art.41882] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 05/18/2021] [Indexed: 11/12/2022]
Abstract
OBJECTIVES To assess the involvement of the CCR6/CCL20 axis in psoriatic arthritis (PsA) and psoriasis (PsO) and to evaluate its potential as a therapeutic target. METHODS First, we quantified CCL20 levels in peripheral blood and synovial fluid of PsA patients and the presence of CCR6+ cells in synovial and tendon tissue. Utilizing an IL-23 minicircle DNA (MC) mouse model exhibiting key features of both PsO and PsA, we investigated CCR6 and CCL20 expression and the preventive and therapeutical effect of CCL20 blockade. Healthy tendon stromal cells were stimulated in vitro with IL-1β to assess the production of CCL20 by qPCR and ELISA. The effect of conditioned media from stimulated tenocytes in inducing T cell migration was interrogated with a transwell system. RESULTS We observed an upregulation of both CCR6 and CCL20 in the enthesis of IL-23 MC-treated mice, which was confirmed in human biopsies. Specific targeting of the CCR6/CCL20 axis with a CCL20 locked dimer (CCL20LD) blocked entheseal inflammation, leading to profound reductions in clinical and proinflammatory markers in the joints and skin of IL-23 MC-treated mice. The stromal compartment in the tendon was the main source of CCL20 in this model and accordingly, in vitro activated human tendon cells were able to produce this chemokine and to induce CCR6+ T cell migration, the latter of which could be blocked by CCL20LD. CONCLUSIONS Our studies highlight the pathogenic role of CCR6-CCL20 axis in enthesitis and raise the prospect of a novel therapeutic approach for treating patients with PsO and PsA.
Collapse
Affiliation(s)
- Zhenrui Shi
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA.,Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China
| | - Emma Garcia-Melchor
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences, University of Glasgow, UK
| | - Xuesong Wu
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | | | - Mimi Nguyen
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Douglas J Rowland
- Center for Molecular and Genomic Imaging, University of California, Davis, Sacramento, CA, USA
| | - Machelle Wilson
- Division of Biostatistics, Clinical and Translational Science Center, University of California, Davis, Sacramento, CA, USA
| | - Flavia Sunzini
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences, University of Glasgow, UK
| | - Moeed Akbar
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences, University of Glasgow, UK
| | - Mindy Huynh
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Timothy Law
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Smriti K Kundu-Raychaudhuri
- Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, CA, USA
| | - Siba P Raychaudhuri
- Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, CA, USA
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, WI, USA
| | - Neal L Millar
- Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary and Life Sciences, University of Glasgow, UK
| | - Sam T Hwang
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
50
|
Single cell transcriptional zonation of human psoriasis skin identifies an alternative immunoregulatory axis conducted by skin resident cells. Cell Death Dis 2021; 12:450. [PMID: 33958582 PMCID: PMC8102483 DOI: 10.1038/s41419-021-03724-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 02/08/2023]
Abstract
Psoriasis is the most common skin disease in adults. Current experimental and clinical evidences suggested the infiltrating immune cells could target local skin cells and thus induce psoriatic phenotype. However, recent studies indicated the existence of a potential feedback signaling loop from local resident skin cells to infiltrating immune cells. Here, we deconstructed the full-thickness human skins of both healthy donors and patients with psoriasis vulgaris at single cell transcriptional level, and further built a neural-network classifier to evaluate the evolutional conservation of skin cell types between mouse and human. Last, we systematically evaluated the intrinsic and intercellular molecular alterations of each cell type between healthy and psoriatic skin. Cross-checking with psoriasis susceptibility gene loci, cell-type based differential expression, and ligand-receptor communication revealed that the resident psoriatic skin cells including mesenchymal and epidermis cell types, which specifically harbored the target genes of psoriasis susceptibility loci, intensively evoked the expression of major histocompatibility complex (MHC) genes, upregulated interferon (INF), tumor necrosis factor (TNF) signalling and increased cytokine gene expression for primarily aiming the neighboring dendritic cells in psoriasis. The comprehensive exploration and pathological observation of psoriasis patient biopsies proposed an uncovered immunoregulatory axis from skin local resident cells to immune cells, thus provided a novel insight for psoriasis treatment. In addition, we published a user-friendly website to exhibit the transcriptional change of each cell type between healthy and psoriatic human skin.
Collapse
|