1
|
Tobias R, Kumar S, Liu J, Lenci N, Gharabi A, Stibor D, Advincula R, Achacoso P, Huang ZM, Bowman C, Ricardo-Gonzalez R, Nakamura MC, Liao W, Malynn BA, Ma A, Razani B. Unrestrained MyD88 signaling in A20-deficient keratinocytes triggers T cell dependent Psoriatic Arthritis like disease. J Invest Dermatol 2025:S0022-202X(25)00451-8. [PMID: 40316204 DOI: 10.1016/j.jid.2025.03.043] [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: 11/25/2024] [Revised: 03/14/2025] [Accepted: 03/27/2025] [Indexed: 05/04/2025]
Abstract
Polymorphisms in A20 (TNFAIP3), a negative regulator of ubiquitin-mediated immune signaling, are strongly associated with Psoriasis and PsA. The tissue-specific roles of A20 in preventing these diseases are poorly understood. As cutaneous psoriasis typically precedes PsA by several years, skin inflammation may represent a key driver of joint disease. We now find that keratinocyte-specific deletion of A20 in normally developed adult mice spontaneously triggers both psoriasiform skin and joint disease, demonstrating a crucial role for epidermal A20 in restricting PsA-like pathology. Mice with A20-deficient keratinocytes that lack T cells were protected from PsA-like disease, showing a key role for epidermally-triggered lymphocytes in driving joint inflammation. Early gene expression analysis following keratinocyte A20 deletion identified activation of MyD88 and antiviral signaling, reflecting spatial transcriptomic changes of human psoriatic epidermis. Keratinocyte-specific loss of A20 together with MyD88, but not germline disruption of interferon receptors, in vivo protected mice from skin and joint pathology. A20-deficient primary keratinocytes from both mice and Crispr-edited human cells spontaneously produced inflammatory cytokines and chemokines in vitro in a MyD88-dependent manner. A20-deficient murine keratinocytes also directly triggered IL17A-secretion from wildtype T cells. Together, our data demonstrate that keratinocyte A20 is critical for preventing T cell dependent PsA-like disease.
Collapse
Affiliation(s)
- Ryan Tobias
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Sugandh Kumar
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Jared Liu
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Nika Lenci
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ameneh Gharabi
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Dorothea Stibor
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Rommel Advincula
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Philip Achacoso
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Zhi-Ming Huang
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher Bowman
- Dept. of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Mary C Nakamura
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; San Francisco VA Health Care System, San Francisco, CA, USA
| | - Wilson Liao
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Barbara A Malynn
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| | - Bahram Razani
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA; San Francisco VA Health Care System, San Francisco, CA, USA.
| |
Collapse
|
2
|
Garrido AN, Machhar R, Cruz-Correa OF, Ganatra D, Crome SQ, Wither J, Jurisica I, Gladman DD. Single-cell RNA sequencing of circulating immune cells supports inhibition of TNFAIP3 and NFKBIA translation as psoriatic arthritis biomarkers. Front Immunol 2025; 16:1483393. [PMID: 39991156 PMCID: PMC11842318 DOI: 10.3389/fimmu.2025.1483393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
Objective To identify biomarkers that distinguish psoriatic arthritis (PsA) from cutaneous psoriasis without arthritis (PsC) and healthy controls (HC) using single cell RNA sequencing (scRNA-seq). Method Peripheral blood mononuclear cell samples from three patients with PsA fulfilling CASPAR criteria, three patients with PsC and two HC were profiled using scRNA-seq. Differentially expressed genes (DEGs) identified through scRNA-seq were validated on classical monocytes, and CD4+ and CD8+ T cell subsets derived from an independent cohort of patients using the NanoString nCounter® platform. Protein expression was measured in CD4+ and CD8+ T cells by immunoblotting. Results A total of 18 immune cell population clusters were identified. Across 18 cell clusters, we identified 234 DEGs. NFKBIA and TNFAIP3 were overexpressed in PsA vs HC and PsC patients. Immunoblotting of the proteins encoded in these genes (IκBα and A20, respectively) showed higher levels in PsA CD4+ T cells compared to HC. Conversely, lower levels were observed in PsA CD8+ T cell lysates compared to HC for both proteins. Conclusion These results suggest that translation of TNFAIP3 and NFKBIA may be inhibited in PsA CD8+ T cells. This study provides insight into the cellular heterogeneity of PsA, showing that non-cell type specific expression of genes associated with the disease can be dysregulated through different mechanisms in distinct cell types.
Collapse
Affiliation(s)
- Ameth N. Garrido
- Gladman-Krembil PsA Research Program, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Rohan Machhar
- Gladman-Krembil PsA Research Program, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Omar F. Cruz-Correa
- Gladman-Krembil PsA Research Program, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Darshini Ganatra
- Gladman-Krembil PsA Research Program, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Sarah Q. Crome
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Joan Wither
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Division of Rheumatology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Departments of Medical Biophysics and Computer Science, and Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Dafna D. Gladman
- Gladman-Krembil PsA Research Program, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Division of Rheumatology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Saw PE, Song E. The 'inflammazone' in chronic inflammatory diseases: psoriasis and sarcoidosis. Trends Immunol 2025; 46:121-137. [PMID: 39875239 DOI: 10.1016/j.it.2025.01.002] [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: 11/12/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
Chronic inflammatory diseases show significant heterogeneity in their phenotypes, with diverse immune cells and mediators interacting in response to various stimuli. This review proposes the concept of the 'inflammazone' framework - which maps the distribution of immune components driving disease pathogenesis - using sarcoidosis and psoriasis as examples. Sarcoidosis features granulomatous inflammation with macrophages and CD4+ T cells, which can spread to lymph nodes and other organs. Psoriasis, affecting primarily the skin, involves Th1, Th17, and Th22 pathways with CD8+ T cells and dendritic cells. Human sarcoidosis exhibits geographic and racial variability, while psoriasis shows varying morphologies and disease courses. Sarcoidosis has more extensive distal immune signaling, whereas psoriasis remains more localized. Understanding the inflammazone is crucial for advancing personalized treatments for inflammatory diseases.
Collapse
Affiliation(s)
- Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Nanhai Clinical Translational Center, Sun Yat-sen Memorial Hospital, Foshan, China; Department of General Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Nanhai Clinical Translational Center, Sun Yat-sen Memorial Hospital, Foshan, China; Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Zenith Institute of Medical Sciences, Guangzhou 510120, China.
| |
Collapse
|
4
|
Sugumaran D, Yong ACH, Stanslas J. Advances in psoriasis research: From pathogenesis to therapeutics. Life Sci 2024; 355:122991. [PMID: 39153596 DOI: 10.1016/j.lfs.2024.122991] [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: 06/04/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Psoriasis is a chronic inflammatory condition affecting approximately 2 % to 3 % of the global population. The pathogenesis of psoriasis is complex, involving immune dysregulation, hyperproliferation and angiogenesis. It is a multifactorial disease which is influenced by genetic and environmental factors. The development of various therapeutic agents, such as JAK inhibitors, small molecules, and biologics with potential anti-psoriatic properties was possible with the vast understanding of the pathogenesis of psoriasis. Various signalling pathways, including NF-κB, JAK-STAT, S1P, PDE-4, and A3AR that are involved in the pathogenesis of psoriasis as well as the preclinical models utilised in the research of psoriasis have been highlighted in this review. The review also focuses on technological advancements that have contributed to a better understanding of psoriasis. Then, the molecules targeting the respective signalling pathways that are still under clinical trials or recently approved as well as the latest breakthroughs in therapeutic and drug delivery approaches that can contribute to the improvement in the management of psoriasis are highlighted in this review. This review provides an extensive understanding of the current state of research in psoriasis, giving rise to opportunities for researchers to discover future therapeutic breakthroughs and personalised interventions. Efficient treatment options for individuals with psoriasis can be achieved by an extensive understanding of pathogenesis, therapeutic agents, and novel drug delivery strategies.
Collapse
Affiliation(s)
- Dineshwar Sugumaran
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Audrey Chee Hui Yong
- Faculty of Pharmacy, Mahsa University, Bandar Saujana Putra, Jenjarom, Selangor, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| |
Collapse
|
5
|
Dong J, Ji B, Jiang Y, Fei F, Guo L, Liu K, Cui L, Meng X, Li J, Wang H. A20 Alleviates the Inflammatory Response in Bovine Endometrial Epithelial Cells by Promoting Autophagy. Animals (Basel) 2024; 14:2876. [PMID: 39409825 PMCID: PMC11475781 DOI: 10.3390/ani14192876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Endometritis represents a prevalent condition in perinatal dairy cows. Bovine endometrial epithelial cells (BEECs), as the primary interface between cavity and the external environment, are particularly vulnerable to infection by pathogenic bacteria following parturition. A20 is essential for regulating inflammation and modulating immune responses. Nevertheless, the exact role of A20 in the BEECs in response to inflammatory response is not fully understood. An endometritis model infected by Escherichia coli (E. coli) in vivo and a BEECs inflammation model induced with lipopolysaccharide (LPS) in vitro were built to investigate the function and governing mechanisms of A20 in endometritis. The results showed that infection with E. coli resulted in endometrial damage, inflammatory cell infiltration, and upregulation of inflammatory factors in dairy cows. Furthermore, A20 expression was upregulated in the endometrium of cows with endometritis and in BEECs following LPS stimulation. A20 overexpression attenuated the level of proinflammatory cytokines in LPS-stimulated BEECs; conversely, A20 knockdown lead to an exacerbated response to LPS stimulation. The overexpression of A20 was shown to activate autophagy and suppress the NF-κB signaling pathway in LPS-stimulated BEECs. However, blocking autophagy with chloroquine notably attenuated the anti-inflammatory effect of A20, leading to the activation of the NF-κB signaling pathway. In summary, the study demonstrated that A20's suppression of inflammation in LPS-stimulated BEECs is associated with the activation of autophagy. Therefore, the A20 protein showed potential as a novel treatment focus for managing endometritis in dairy cows.
Collapse
Affiliation(s)
- Junsheng Dong
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; (B.J.); (Y.J.); (F.F.); (L.G.); (K.L.); (L.C.); (X.M.); (J.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Bowen Ji
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; (B.J.); (Y.J.); (F.F.); (L.G.); (K.L.); (L.C.); (X.M.); (J.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Yeqi Jiang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; (B.J.); (Y.J.); (F.F.); (L.G.); (K.L.); (L.C.); (X.M.); (J.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Fan Fei
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; (B.J.); (Y.J.); (F.F.); (L.G.); (K.L.); (L.C.); (X.M.); (J.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Long Guo
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; (B.J.); (Y.J.); (F.F.); (L.G.); (K.L.); (L.C.); (X.M.); (J.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Kangjun Liu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; (B.J.); (Y.J.); (F.F.); (L.G.); (K.L.); (L.C.); (X.M.); (J.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Luying Cui
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; (B.J.); (Y.J.); (F.F.); (L.G.); (K.L.); (L.C.); (X.M.); (J.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Xia Meng
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; (B.J.); (Y.J.); (F.F.); (L.G.); (K.L.); (L.C.); (X.M.); (J.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Jianji Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; (B.J.); (Y.J.); (F.F.); (L.G.); (K.L.); (L.C.); (X.M.); (J.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Heng Wang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; (B.J.); (Y.J.); (F.F.); (L.G.); (K.L.); (L.C.); (X.M.); (J.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
6
|
do Amaral SR, Amantino CF, Atanasov A, Sousa SO, Moakes R, Oliani SM, Grover LM, Primo FL. Photodynamic Therapy as a Novel Therapeutic Modality Applying Quinizarin-Loaded Nanocapsules and 3D Bioprinting Skin Permeation for Inflammation Treatment. Pharmaceuticals (Basel) 2024; 17:1169. [PMID: 39338332 PMCID: PMC11434822 DOI: 10.3390/ph17091169] [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: 07/30/2024] [Revised: 08/27/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Skin inflammation associated with chronic diseases involves a direct role of keratinocytes in its immunopathogenesis, triggering a cascade of immune responses. Despite this, highly targeted treatments remain elusive, highlighting the need for more specific therapeutic strategies. In this study, nanocapsules containing quinizarin (QZ/NC) were developed and evaluated in an in vitro model of keratinocyte-mediated inflammation, incorporating the action of photodynamic therapy (PDT) and analyzing permeation in a 3D skin model. Comprehensive physicochemical, stability, cytotoxicity, and permeation analyses of the nanomaterials were conducted. The nanocapsules demonstrated desirable physicochemical properties, remained stable throughout the analysis period, and exhibited no spectroscopic alterations. Cytotoxicity tests revealed no toxicity at the lowest concentrations of QZ/NC. Permeation and cellular uptake studies confirmed QZ/NC permeation in 3D skin models, along with intracellular incorporation and internalization of the drug, thereby enhancing its efficacy in drug delivery. The developed model for inducing the inflammatory process in vitro yielded promising results, particularly when the synthesized nanomaterial was combined with PDT, showing a reduction in cytokine levels. These findings suggest a potential new therapeutic approach for treating inflammatory skin diseases.
Collapse
Affiliation(s)
- Stéphanie R. do Amaral
- Department of Bioprocess and Biotechnology Engineering, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (S.R.d.A.); (C.F.A.)
| | - Camila F. Amantino
- Department of Bioprocess and Biotechnology Engineering, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (S.R.d.A.); (C.F.A.)
- São Paulo Federal Institute of Education, Science and Technology (IFSP), Matão 15991-502, SP, Brazil
| | - Aleksandar Atanasov
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK; (A.A.); (R.M.); (L.M.G.)
| | - Stefanie Oliveira Sousa
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto 15054-000, SP, Brazil; (S.O.S.); (S.M.O.)
| | - Richard Moakes
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK; (A.A.); (R.M.); (L.M.G.)
| | - Sonia Maria Oliani
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto 15054-000, SP, Brazil; (S.O.S.); (S.M.O.)
| | - Liam M. Grover
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK; (A.A.); (R.M.); (L.M.G.)
| | - Fernando L. Primo
- Department of Bioprocess and Biotechnology Engineering, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (S.R.d.A.); (C.F.A.)
| |
Collapse
|
7
|
Du L, Pan D, Huang H, Liu Q, Yang Y, Jiang G. Shoutai Wan treatment upregulates the expression of TNFAIP3 and improves T cell immune tolerance at maternal-fetal interface. J Reprod Immunol 2024; 165:104301. [PMID: 39146884 DOI: 10.1016/j.jri.2024.104301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 08/17/2024]
Abstract
Shoutai Wan (STW) is a traditional Chinese medicine formula used to treat various conditions. The objective of this study was to evaluate the impact of STW on the abortion rate in the URSA mouse model and elucidate its underlying molecular mechanisms. Female CBA/J mice were mated with male DBA/2 mice to establish the URSA model. Network pharmacological analysis was employed to investigate the potential molecular mechanisms of STW. Hematoxylin-eosin staining, immunofluorescence, and ELISA were performed to examine placental microenvironmental changes, protein expression related to TNFAIP3 and the NF-κB signaling pathway. Treatment with STW reduced the abortion rate in URSA model mice and improved trophoblast development. TNFAIP3 was identified as a potential target of STW for treating URSA, as STW enhanced TNFAIP3 protein expression while decreasing IL-6 and TNF-α secretion in the placenta. Moreover, STW upregulated TNFAIP3 protein expression and Foxp3 mRNA levels, increased the production of anti-inflammatory cytokines such as IL-10 and TGF-β1, and decreased p-NF-κB expression in CD4+ cells at the placenta. The findings of this study indicate that STW treatment reduces the abortion rate in the URSA mouse model. These effects are likely mediated by increased TNFAIP3 expression and decreased NF-κB signaling pathway activity at the maternal-fetal interface. These molecular changes may contribute to the regulation of T cell immunity and immune tolerance during pregnancy.
Collapse
Affiliation(s)
- Le Du
- Department of traditional Chinese Medicine, Pizhou people's Hospital affiliated to Xuzhou Medical University, Jiangsu 221000, China
| | - Dingchen Pan
- Department of Obstetrics and Gynecology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200000, China
| | - He Huang
- ShuGuang Clinical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Liu
- ShuGuang Clinical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Yang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 200000, China.
| | - Guojing Jiang
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200092, China.
| |
Collapse
|
8
|
Sen’kova AV, Savin IA, Chernolovskaya EL, Davydova AS, Meschaninova MI, Bishani A, Vorobyeva MA, Zenkova MA. LPS-Induced Acute Lung Injury: Analysis of the Development and Suppression by the TNF-α-Targeting Aptamer. Acta Naturae 2024; 16:61-71. [PMID: 39188267 PMCID: PMC11345095 DOI: 10.32607/actanaturae.27393] [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: 03/22/2024] [Accepted: 04/15/2024] [Indexed: 08/28/2024] Open
Abstract
Acute lung injury (ALI) is a specific form of lung inflammation characterized by diffuse alveolar damage, noncardiogenic pulmonary edema, as well as a pulmonary and systemic inflammation. The pathogenesis of ALI involves a cascade inflammatory response accompanied by an increase in the local and systemic levels of proinflammatory cytokines and chemokines. The development of molecular tools targeting key components of cytokine signaling appears to be a promising approach in ALI treatment. The development of lipopolysaccharide (LPS)-induced ALI, as well as the feasibility of suppressing it by an aptamer targeting the proinflammatory cytokine TNF-α, was studied in a mouse model. The TNF-α level was shown to increase significantly and remain steadily high during the development of ALI. LPS-induced morphological signs of inflammation in the respiratory system become most pronounced 24 h after induction. Intranasal administration of TNF-α-targeting aptamers conjugated with polyethylene glycol (PEG-aptTNF-α) to mice with ALI reduced the intensity of inflammatory changes in lung tissue. Assessment of the levels of potential TNF-α target genes (Usp18, Traf1, and Tnfaip3) showed that their expression levels in the lungs increase during ALI development, while declining after the application of PEG-aptTNF-α. Therefore, topical use of TNF-α- targeting aptamers may be an efficient tool for treating ALI and other inflammatory lung diseases.
Collapse
Affiliation(s)
- A. V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russian Federation
| | - I. A. Savin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russian Federation
| | - E. L. Chernolovskaya
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russian Federation
| | - A. S. Davydova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russian Federation
| | - M. I. Meschaninova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russian Federation
| | - A. Bishani
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russian Federation
| | - M. A. Vorobyeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russian Federation
| | - M. A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russian Federation
| |
Collapse
|
9
|
Liu Y, Li H, Ouyang Y, Zhang Y, Pan P. Exploration of the role of oxidative stress-related genes in LPS-induced acute lung injury via bioinformatics and experimental studies. Sci Rep 2023; 13:21804. [PMID: 38071255 PMCID: PMC10710410 DOI: 10.1038/s41598-023-49165-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
During the progression of acute lung injury (ALI), oxidative stress and inflammatory responses always promote each other. The datasets analyzed in this research were acquired from the Gene Expression Omnibus (GEO) database. The Weighted Gene Co-expression Network Analysis (WGCNA) and limma package were used to obtain the ALI-related genes (ALIRGs) and differentially expressed genes (DEGs), respectively. In total, two biological markers (Gch1 and Tnfaip3) related to oxidative stress were identified by machine learning algorithms, Receiver Operator Characteristic (ROC), and differential expression analyses. The area under the curve (AUC) value of biological markers was greater than 0.9, indicating an excellent power to distinguish between ALI and control groups. Moreover, 15 differential immune cells were selected between the ALI and control samples, and they were correlated to biological markers. The transcription factor (TF)-microRNA (miRNA)-Target network was constructed to explore the potential regulatory mechanisms. Finally, based on the quantitative reverse transcription polymerase chain reaction (qRT-PCR), the expression of Gch1 and Tnfaip3 was significantly higher in ALI lung tissue than in healthy controls. In conclusion, the differences in expression profiles between ALI and normal controls were found, and two biological markers were identified, providing a research basis for further understanding the pathogenesis of ALI.
Collapse
Affiliation(s)
- Yuanshui Liu
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, People's Republic of China.
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.
| | - Huamei Li
- Department of Ultrasound, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, People's Republic of China.
| | - Yanhong Ouyang
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, People's Republic of China
| | - Yan Zhang
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.
| | - Pinhua Pan
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.
| |
Collapse
|
10
|
Wang M, Huang X, Ouyang M, Lan J, Huang J, Li H, Lai W, Gao Y, Xu Q. A20 ameliorates advanced glycation end products-induced melanogenesis by inhibiting NLRP3 inflammasome activation in human dermal fibroblasts. J Dermatol Sci 2023; 112:71-82. [PMID: 37741724 DOI: 10.1016/j.jdermsci.2023.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/03/2023] [Accepted: 09/05/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND Advanced glycation end products (AGEs) promote melanogenesis through activating NLRP3 inflammasome in fibroblasts. Although A20 has been highlighted to inhibit NLRP3 inflammasome activation, its roles and mechanisms remain elusive in photoaging-associated pigmentation. OBJECTIVES To determine the significance of fibroblast A20 in AGEs-induced NLRP3 inflammasome activation and pigmentation. METHODS The correlation between A20 and AGEs or melanin was studied in sun-exposed skin and lesions of melasma and solar lentigo. We then investigated A20 level in AGEs-treated fibroblast and the effect of fibroblast A20 overexpression or knockdown on AGEs-BSA-induced NLRP3 inflammasome activation and pigmentation, respectively. Finally, the severity of NLRP3 inflammasome activation and pigmentation was evaluated after mice were injected intradermally with A20-overexpression adeno-associated virus and AGEs-BSA. RESULTS Dermal A20 expression was decreased and exhibited negative correlation with either dermal AGEs deposition or epidermal melanin level in sun-exposed skin and pigmentary lesions. Moreover, both AGEs-BSA and AGEs-collagen robustly decreased A20 expression via binding to RAGE in fibroblasts. Further, A20 overexpression or depletion significantly decreased or augmented AGEs-BSA-induced activation of NF-κB pathway and NLRP3 inflammasome and IL-18 production and secretion in fibroblasts, respectively. Importantly, fibroblast A20 potently repressed AGEs-BSA-stimulated melanin content,tyrosinase activity,and expression of microphthalmia-associated transcription factor and tyrosinase in melanocytes. Particularly, fibroblast A20 significantly abrogated AGEs-BSA-promoted melanogenesis in ex vivo skin and mouse models. Additionally, fibroblast A20 inhibited AGEs-BSA-activated MAPKs in melanocytes and the epidermis of ex vivo skin. CONCLUSIONS Fibroblast A20 suppresses AGEs-stimulate melanogenesis in photoaging-associated hyperpigmentation disorders by inhibiting NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Mengyao Wang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Xianyin Huang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Mengting Ouyang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Jingjing Lan
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Jingqian Huang
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Hongpeng Li
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Wei Lai
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China.
| | - Yifeng Gao
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China.
| | - Qingfang Xu
- Department of Dermato-Venereology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
11
|
Carman LE, Samulevich ML, Aneskievich BJ. Repressive Control of Keratinocyte Cytoplasmic Inflammatory Signaling. Int J Mol Sci 2023; 24:11943. [PMID: 37569318 PMCID: PMC10419196 DOI: 10.3390/ijms241511943] [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: 06/23/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
The overactivity of keratinocyte cytoplasmic signaling contributes to several cutaneous inflammatory and immune pathologies. An important emerging complement to proteins responsible for this overactivity is signal repression brought about by several proteins and protein complexes with the native role of limiting inflammation. The signaling repression by these proteins distinguishes them from transmembrane receptors, kinases, and inflammasomes, which drive inflammation. For these proteins, defects or deficiencies, whether naturally arising or in experimentally engineered skin inflammation models, have clearly linked them to maintaining keratinocytes in a non-activated state or returning cells to a post-inflamed state after a signaling event. Thus, together, these proteins help to resolve acute inflammatory responses or limit the development of chronic cutaneous inflammatory disease. We present here an integrated set of demonstrated or potentially inflammation-repressive proteins or protein complexes (linear ubiquitin chain assembly complex [LUBAC], cylindromatosis lysine 63 deubiquitinase [CYLD], tumor necrosis factor alpha-induced protein 3-interacting protein 1 [TNIP1], A20, and OTULIN) for a comprehensive view of cytoplasmic signaling highlighting protein players repressing inflammation as the needed counterpoints to signal activators and amplifiers. Ebb and flow of players on both sides of this inflammation equation would be of physiological advantage to allow acute response to damage or pathogens and yet guard against chronic inflammatory disease. Further investigation of the players responsible for repressing cytoplasmic signaling would be foundational to developing new chemical-entity pharmacologics to stabilize or enhance their function when clinical intervention is needed to restore balance.
Collapse
Affiliation(s)
- Liam E. Carman
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (L.E.C.); (M.L.S.)
| | - Michael L. Samulevich
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (L.E.C.); (M.L.S.)
| | - Brian J. Aneskievich
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
12
|
Jia Y, Yin C, Ke W, Liu J, Guo B, Wang X, Zhao P, Hu S, Zhang C, Li X, Liu R, Zheng X, Wang Y, Wang G, Pan H, Hu W, Song Z. Alpha-ketoglutarate alleviates cadmium-induced inflammation by inhibiting the HIF1A-TNFAIP3 pathway in hepatocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 878:163069. [PMID: 36996991 DOI: 10.1016/j.scitotenv.2023.163069] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 05/13/2023]
Abstract
The threat to public health posed by rapidly increasing levels of cadmium (Cd) in the environment is receiving worldwide attention. Although, Cd is known to be absorbed into the body and causes non-negligible damage to the liver, the detailed mechanisms underlying its hepatoxicity are incompletely understood. In the present study, investigated the effect of TNFAIP3 and α-ketoglutarate (AKG) on Cd-induced liver inflammation and hepatocyte death. Male C57BL/6 mice were exposed to cadmium chloride (1.0 mg/kg) while being fed a diet with 2 % AKG for two weeks. We found that Cd induced hepatocyte injury and inflammatory infiltration. In addition, TNFAIP3 expression was inhibited in the liver tissues and cells of CdCl2-treated mice. Mouse hepatocyte-specific TNFAIP3 overexpression by tail vein injection of an adeno-associated virus (AAV) vector effectively alleviated Cd-induced hepatic necrosis and inflammation, which was mediated by the NF-κB signaling pathway. Notably, this inhibitory effect of TNFAIP3 on Cd-induced liver injury was dependent on AKG. Exogenous addition of AKG prevented Cd exposure-induced increases in serum ALT, AST and LDH levels, production of pro-inflammatory cytokines, activation of the NF-κB signaling pathway, and even significantly reduced Cd-induced oxidative stress and hepatocyte death. Mechanistically, AKG exerted its anti-inflammatory effect by promoting the hydroxylation and degradation of HIF1A to reduce its Cd-induced overexpression in vivo and in vitro, avoiding the inhibition of the TNFAIP3 promoter by HIF1A. Moreover, the protective effect of AKG was significantly weaker in Cd-treated primary hepatocytes transfected with HIF1A pcDNA. Overall, our results reveal a novel mechanism of Cd-induced hepatotoxicity.
Collapse
Affiliation(s)
- Yinzhao Jia
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Chuanzheng Yin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Wenbo Ke
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jing Liu
- Key Laboratory of Coal Science and Technology of Ministry of Education, College of Chemistry and Chemical Engineering, Taiyuan University of Technology, Taiyuan 030000, China
| | - Bing Guo
- Insitute for Genome Sciences, University of Maryland School of Medical, Baltimore, MD 21201, United States
| | - Xiaofei Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Peng Zhao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Shaobo Hu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xuan Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Ran Liu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xichuan Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yaofeng Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Gengqiao Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hao Pan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Wenjun Hu
- School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| |
Collapse
|
13
|
Shahine Y, El-Aal SAA, Reda AM, Sheta E, Atia NM, Abdallah OY, Ibrahim SSA. Diosmin nanocrystal gel alleviates imiquimod-induced psoriasis in rats via modulating TLR7,8/NF-κB/micro RNA-31, AKT/mTOR/P70S6K milieu, and Tregs/Th17 balance. Inflammopharmacology 2023; 31:1341-1359. [PMID: 37010718 DOI: 10.1007/s10787-023-01198-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/17/2023] [Indexed: 04/04/2023]
Abstract
Diosmin is a flavonoid with promising anti-inflammatory and antioxidant properties. However, it has difficult physicochemical characteristics since its solubility demands a pH level of 12, which has an impact on the drug's bioavailability. The aim of this work is the development and characterization of diosmin nanocrystals using anti-solvent precipitation technique to be used for topical treatment of psoriasis. Results revealed that diosmin nanocrystals stabilized with hydroxypropyl methylcellulose (HPMC E15) in ratio (diosmin:polymer; 1:1) reached the desired particle size (276.9 ± 16.49 nm); provided promising colloidal properties and possessed high drug release profile. Additionally, in-vivo assessment was carried out to evaluate and compare the activities of diosmin nanocrystal gel using three different doses and diosmin powder gel in alleviating imiquimod-induced psoriasis in rats and investigating their possible anti-inflammatory mechanisms. Herein, 125 mg of 5% imiquimod cream (IMQ) was applied topically for 5 consecutive days on the shaved backs of rats to induce psoriasis. Diosmin nanocrystal gel especially in the highest dose used offered the best anti-inflammatory effect. This was confirmed by causing the most statistically significant reduction in the psoriasis area severity index (PASI) score and the serum inflammatory cytokines levels. Furthermore, it was capable of maintaining the balance between T helper (Th17) and T regulatory (Treg) cells. Moreover, it tackled TLR7/8/NF-κB, miRNA-31, AKT/mTOR/P70S6K and elevated the TNFAIP3/A20 (a negative regulator of NF-κB) expression in psoriatic skin tissues. This highlights the role of diosmin nanocrystal gel in tackling imiquimod-induced psoriasis in rats, and thus it could be a novel promising therapy for psoriasis.
Collapse
Affiliation(s)
- Yasmine Shahine
- Department of Microbiology & Immunology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Sarah A Abd El-Aal
- Department of Pharmacy, Kut University College, Al Kut, Wasit, 52001, Iraq
| | - Ahmed M Reda
- Department of Pharmacy, Kut University College, Al Kut, Wasit, 52001, Iraq
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, Egypt
| | - Eman Sheta
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nouran M Atia
- Department of Pharmaceutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Sherihan Salaheldin Abdelhamid Ibrahim
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria (PUA), Canal El- Mahmoudia Street, Smouha, Alexandria, Egypt.
| |
Collapse
|
14
|
Trompette A, Ubags ND. Skin barrier immunology from early life to adulthood. Mucosal Immunol 2023; 16:194-207. [PMID: 36868478 DOI: 10.1016/j.mucimm.2023.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/15/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023]
Abstract
Our skin has a unique barrier function, which is imperative for the body's protection against external pathogens and environmental insults. Although interacting closely and sharing many similarities with key mucosal barrier sites, such as the gut and the lung, the skin also provides protection for internal tissues and organs and has a distinct lipid and chemical composition. Skin immunity develops over time and is influenced by a multiplicity of different factors, including lifestyle, genetics, and environmental exposures. Alterations in early life skin immune and structural development may have long-term consequences for skin health. In this review, we summarize the current knowledge on cutaneous barrier and immune development from early life to adulthood, with an overview of skin physiology and immune responses. We specifically highlight the influence of the skin microenvironment and other host intrinsic, host extrinsic (e.g. skin microbiome), and environmental factors on early life cutaneous immunity.
Collapse
Affiliation(s)
- Aurélien Trompette
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Niki D Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
15
|
Zhang Y, Xu X, Cheng H, Zhou F. AIM2 and Psoriasis. Front Immunol 2023; 14:1085448. [PMID: 36742336 PMCID: PMC9889639 DOI: 10.3389/fimmu.2023.1085448] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease occurring worldwide, with multiple systemic complications, which seriously affect the quality of life and physical and mental health of patients. The pathogenesis of psoriasis is related to the environment, genetics, epigenetics, and dysregulation of immune cells such as T cells, dendritic cells (DCs), and nonimmune cells such as keratinocytes. Absent in melanoma 2 (AIM2), a susceptibility gene locus for psoriasis, has been strongly linked to the genetic and epigenetic aspects of psoriasis and increased in expression in psoriatic keratinocytes. AIM2 was found to be activated in an inflammasome-dependent way to release IL-1β and IL-18 to mediate inflammation, and to participate in immune regulation in psoriasis, or in an inflammasome-independent way by regulating the function of regulatory T(Treg) cells or programming cell death in keratinocytes as well as controlling the proliferative state of different cells. AIM2 may also play a role in the recurrence of psoriasis by trained immunity. In this review, we will elaborate on the characteristics of AIM2 and how AIM2 mediates the development of psoriasis.
Collapse
Affiliation(s)
- Yuxi Zhang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China.,Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xiaoqing Xu
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China.,Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Hui Cheng
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China.,Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Fusheng Zhou
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China.,Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| |
Collapse
|
16
|
Dombrowicz D. Identification of major human IgE-inducing parasite antigens: A path to therapeutic approaches? J Allergy Clin Immunol 2022; 150:1412-1414. [PMID: 36270491 DOI: 10.1016/j.jaci.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Affiliation(s)
- David Dombrowicz
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France.
| |
Collapse
|
17
|
Shi L, Liu C, Xiong H, Shi D. Elevation of IgE in patients with psoriasis: Is it a paradoxical phenomenon? Front Med (Lausanne) 2022; 9:1007892. [PMID: 36314037 PMCID: PMC9606585 DOI: 10.3389/fmed.2022.1007892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022] Open
Abstract
Immunoglobulin E (IgE) elevation is a hallmark of allergic conditions such as atopic dermatitis (AD). The pathogenesis of AD is typically associated with high levels of IL-4 and IL-13 produced by activated T helper 2 (Th2) cells. Psoriasis, on the other hand, is an inflammatory skin disease mainly driven by Th17 cells and their related cytokines. Although the immunopathologic reactions and clinical manifestations are often easily distinguished in the two skin conditions, patients with psoriasis may sometimes exhibit AD-like manifestations, such as elevated IgE and persistent pruritic lesions. Given the fact that the effective T cells have great plasticity to re-differentiate in response to innate and environmental factors, this unusual skin condition could be a consequence of a cross-reaction between distinct arms of T-cell and humoral immunity. Here we review the literature concerning the roles of IgE in the development of AD and psoriasis, showing that elevated IgE seems to be an important indicator for this non-typical psoriasis.
Collapse
Affiliation(s)
- Leyao Shi
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China,The Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, China
| | - Chen Liu
- The Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, China
| | - Huabao Xiong
- Basic Medical School, Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China,Huabao Xiong
| | - Dongmei Shi
- The Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, China,Department of Dermatology, Jining No.1 People's Hospital, Jining, China,*Correspondence: Dongmei Shi
| |
Collapse
|
18
|
Ma X, Ru Y, Luo Y, Kuai L, Chen QL, Bai Y, Liu YQ, Chen J, Luo Y, Song JK, Zhou M, Li B. Post-Translational Modifications in Atopic Dermatitis: Current Research and Clinical Relevance. Front Cell Dev Biol 2022; 10:942838. [PMID: 35874824 PMCID: PMC9301047 DOI: 10.3389/fcell.2022.942838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/16/2022] [Indexed: 11/20/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic and relapsing cutaneous disorder characterized by compromised immune system, excessive inflammation, and skin barrier disruption. Post-translational modifications (PTMs) are covalent and enzymatic modifications of proteins after their translation, which have been reported to play roles in inflammatory and allergic diseases. However, less attention has been paid to the effect of PTMs on AD. This review summarized the knowledge of six major classes (including phosphorylation, acetylation, ubiquitination, SUMOylation, glycosylation, o-glycosylation, and glycation) of PTMs in AD pathogenesis and discussed the opportunities for disease management.
Collapse
Affiliation(s)
- Xin Ma
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Yi Ru
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Qi-Long Chen
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Yun Bai
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Ye-Qiang Liu
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Jia Chen
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Yue Luo
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Jian-Kun Song
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Mi Zhou
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Mi Zhou, ; Bin Li,
| | - Bin Li
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Mi Zhou, ; Bin Li,
| |
Collapse
|
19
|
Sylvester M, Son A, Schwartz DM. The Interactions Between Autoinflammation and Type 2 Immunity: From Mechanistic Studies to Epidemiologic Associations. Front Immunol 2022; 13:818039. [PMID: 35281022 PMCID: PMC8907424 DOI: 10.3389/fimmu.2022.818039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/02/2022] [Indexed: 12/30/2022] Open
Abstract
Autoinflammatory diseases are a group of clinical syndromes characterized by constitutive overactivation of innate immune pathways. This results in increased production of or responses to monocyte- and neutrophil-derived cytokines such as interleukin-1β (IL-1β), Tumor Necrosis Factor-α (TNF-α), and Type 1 interferon (IFN). By contrast, clinical allergy is caused by dysregulated type 2 immunity, which is characterized by expansion of T helper 2 (Th2) cells and eosinophils, as well as overproduction of the associated cytokines IL-4, IL-5, IL-9, and IL-13. Traditionally, type 2 immune cells and autoinflammatory effectors were thought to counter-regulate each other. However, an expanding body of evidence suggests that, in some contexts, autoinflammatory pathways and cytokines may potentiate type 2 immune responses. Conversely, type 2 immune cells and cytokines can regulate autoinflammatory responses in complex and context-dependent manners. Here, we introduce the concepts of autoinflammation and type 2 immunity. We proceed to review the mechanisms by which autoinflammatory and type 2 immune responses can modulate each other. Finally, we discuss the epidemiology of type 2 immunity and clinical allergy in several monogenic and complex autoinflammatory diseases. In the future, these interactions between type 2 immunity and autoinflammation may help to expand the spectrum of autoinflammation and to guide the management of patients with various autoinflammatory and allergic diseases.
Collapse
Affiliation(s)
- McKella Sylvester
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Aran Son
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniella M Schwartz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
20
|
Xu X, Tang X, Zhang Y, Pan Z, Wang Q, Tang L, Zhu C, Cheng H, Zhou F. Chromatin accessibility and transcriptome integrative analysis revealed AP-1-mediated genes potentially modulate histopathology features in psoriasis. Clin Epigenetics 2022; 14:38. [PMID: 35277199 PMCID: PMC8917665 DOI: 10.1186/s13148-022-01250-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/16/2022] [Indexed: 12/19/2022] Open
Abstract
Abstract
Background
Psoriasis is a chronic and hyperproliferative skin disease featured by hyperkeratosis with parakeratosis, Munro micro-abscess, elongation of rete pegs, granulosa thinning, and lymphocyte infiltration. We previously profiled gene expression and chromatin accessibility of psoriatic skins by transcriptome sequencing and ATAC-seq. However, integrating both of these datasets to unravel gene expression regulation is lacking. Here, we integrated transcriptome and ATAC-seq of the same psoriatic and normal skin tissues, trying to leverage the potential role of chromatin accessibility and their function in histopathology features.
Results
By inducing binding and expression target analysis (BETA) algorithms, we explored the target prediction of transcription factors binding in 15 psoriatic and 19 control skins. BETA identified 408 upregulated genes (rank product < 0.01) and 133 downregulated genes linked with chromatin accessibility. We noticed that cumulative fraction of genes in upregulation group was statistically higher than background, while that of genes in downregulation group was not significant. KEGG pathway analysis showed that the upregulated 408 genes were enriched in TNF, NOD, and IL-17 signaling pathways. In addition, the motif module in BETA suggested the 57 upregulated genes are targeted by transcription factor AP-1, indicating that increased chromatin accessibility facilitated the binding of AP-1 to the target regions and further induced expression of relevant genes. Among these genes, SQLE, STRN, EIF4, and MYO1B expression was increased in patients with hyperkeratosis, parakeratosis, and acanthosis thickening.
Conclusions
In summary, with the advantage of BETA, we identified a series of genes that contribute to the disease pathogenesis, especially in modulating histopathology features, providing us with new clues in treating psoriasis.
Collapse
|
21
|
Xia P, Pasquali L, Gao C, Srivastava A, Khera N, Freisenhausen JC, Luo L, Rosén E, van Lierop A, Homey B, Pivarcsi A, Sonkoly E. miR-378a regulates keratinocyte responsiveness to IL-17A in psoriasis. Br J Dermatol 2022; 187:211-222. [PMID: 35257359 PMCID: PMC9545829 DOI: 10.1111/bjd.21232] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/15/2022] [Accepted: 02/28/2022] [Indexed: 12/04/2022]
Abstract
Background Psoriasis is an immune‐mediated inflammatory skin disease, in which an interplay between infiltrating immune cells and keratinocytes sustains chronic skin inflammation. Interleukin (IL)‐17A is a key inflammatory cytokine in psoriasis and its main cellular targets are keratinocytes. Objectives To explore the role of miR‐378a in psoriasis. Methods Keratinocytes obtained from psoriatic skin and healthy epidermis were separated by magnetic sorting, and the expression of miR‐378a was analysed by quantitative polymerase chain reaction. The regulation and function of miR‐378a was studied using primary human keratinocytes. The expression of miR‐378a was modulated by synthetic mimics, and nuclear factor kappa B (NF‐κB) activity and transcriptomic changes were studied. Synthetic miR‐378a was delivered to mouse skin in conjunction with induction of psoriasiform skin inflammation by imiquimod. Results We show that miR‐378a is induced by IL‐17A in keratinocytes through NF‐κB, C/EBP‐β and IκBζ and that it is overexpressed in psoriatic epidermis. In cultured keratinocytes, ectopic expression of miR‐378a resulted in the nuclear translocation of p65 and enhanced NF‐κB‐driven promoter activity even in the absence of inflammatory stimuli. Moreover, miR‐378a potentiated the effect of IL‐17A on NF‐κB nuclear translocation and downstream activation of the NF‐κB pathway. Finally, injection of miR‐378a into mouse skin augmented psoriasis‐like skin inflammation with increased epidermal proliferation and induction of inflammatory mediators. Mechanistically, miR‐378a acts as a suppressor of NFKBIA/IκBζ, an important negative regulator of the NF‐κB pathway in keratinocytes. Conclusions Collectively, our findings identify miR‐378a as an amplifier of IL‐17A‐induced NF‐κB signalling in keratinocytes and suggest that increased miR‐378a levels contribute to the amplification of IL‐17A‐driven skin inflammation in psoriasis.
Collapse
Affiliation(s)
- Ping Xia
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine (CMM), Karolinska University Hospital, Stockholm, Sweden.,Dermatology Unit, Wuhan No.1 Hospital, Wuhan, China
| | - Lorenzo Pasquali
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine (CMM), Karolinska University Hospital, Stockholm, Sweden
| | - Chenying Gao
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine (CMM), Karolinska University Hospital, Stockholm, Sweden.,Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Ankit Srivastava
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine (CMM), Karolinska University Hospital, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden.,The Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Nupur Khera
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine (CMM), Karolinska University Hospital, Stockholm, Sweden
| | - Jan Cedric Freisenhausen
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine (CMM), Karolinska University Hospital, Stockholm, Sweden
| | - Longlong Luo
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine (CMM), Karolinska University Hospital, Stockholm, Sweden
| | - Einar Rosén
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Anke van Lierop
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Bernhard Homey
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Andor Pivarcsi
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine (CMM), Karolinska University Hospital, Stockholm, Sweden.,Department of Medical Biochemistry and Microbiology (IMBIM), Uppsala University, Uppsala, Sweden
| | - Enikö Sonkoly
- Dermatology and Venereology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine (CMM), Karolinska University Hospital, Stockholm, Sweden.,Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
DeVore SB, Stevens ML, He H, Biagini JM, Kroner JW, Martin LJ, Hershey GKK. Novel role for caspase recruitment domain family member 14 and its genetic variant rs11652075 in skin filaggrin homeostasis. J Allergy Clin Immunol 2022; 149:708-717. [PMID: 34271060 PMCID: PMC9119145 DOI: 10.1016/j.jaci.2021.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Low epidermal filaggrin (FLG) is a risk factor for atopic dermatitis (AD) and allergic comorbidity. FLG mutations do not fully explain the variation in epidermal FLG levels, highlighting that other genetic loci may also regulate FLG expression. OBJECTIVE We sought to identify genetic loci that regulate FLG expression and elucidate their functional and mechanistic consequences. METHODS A genome-wide association study of quantified skin FLG expression in lesional and baseline non(never)-lesional skin of children with AD in the Mechanisms of Progression of Atopic Dermatitis to Asthma in Children cohort was conducted. Clustered regularly interspaced short palindromic repeat approaches were used to create isogenic human keratinocytes differing only at the identified variant rs11652075, and caspase recruitment domain family member 14 (CARD14)-deficient keratinocytes for subsequent mechanistic studies. RESULTS The genome-wide association study identified the CARD14 rs11652075 variant to be associated with FLG expression in non(never)-lesional skin of children with AD. Rs11652075 is a CARD14 expression quantitative trait locus in human skin and primary human keratinocytes. The T variant destroys a functional cytosine-phosphate-guanine site, resulting in reduced cytosine-phosphate-guanine methylation at this site (but not neighboring sites) in TT and CT compared with CC primary human keratinocytes and Mechanisms of Progression of Atopic Dermatitis to Asthma in Children children's skin samples, and rs11652075 increases CARD14 expression in an allele-specific fashion. Furthermore, studies in clustered regularly interspaced short palindromic repeat-generated CC and TT isogenic keratinocytes, as well as CARD14-haplosufficient and deficient keratinocytes, reveal that IL-17A regulates FLG expression via CARD14, and that the underlying mechanisms are dependent on the rs11652075 genotype. CONCLUSIONS Our study identifies CARD14 as a novel regulator of FLG expression in the skin of children with AD. Furthermore, CARD14 regulates skin FLG homeostasis in an rs11652075-dependent fashion.
Collapse
Affiliation(s)
- Stanley B. DeVore
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, Ohio 45267, USA.,Division of Asthma Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Mariana L. Stevens
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Hua He
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Jocelyn M. Biagini
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, Ohio 45267, USA.,Division of Asthma Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - John W. Kroner
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Lisa J. Martin
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, Ohio 45267, USA.,Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Gurjit K. Khurana Hershey
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, Ohio 45267, USA.,Division of Asthma Research, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA.,Corresponding Author Information Gurjit Khurana Hershey, MD, PhD, 3333 Burnet Avenue, MLC 7037, Cincinnati, OH 45229, USA, Phone 513-636-7054, Fax 513-636-1657,
| |
Collapse
|
23
|
Yan H, Jin M, Li Y, Gao Y, Ding Q, Wang X, Zeng W, Chen Y. miR-1 Regulates Differentiation and Proliferation of Goat Hair Follicle Stem Cells by Targeting IGF1R and LEF1 Genes. DNA Cell Biol 2022; 41:190-201. [PMID: 35007429 DOI: 10.1089/dna.2021.0288] [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] [Indexed: 11/12/2022] Open
Abstract
Hair follicle stem cells (HFSCs) play a significant role in hair development. miR-1 has been reported as an important regulatory factor that affects hair follicle growth and development, but its regulatory mechanism on HFSC development remains unknown. In this study, the molecular mechanism of miR-1 in regulating HFSC proliferation and differentiation was investigated. High-throughput RNA-seq and integrated analysis were performed to identify differentially transcribed mRNAs and microRNAs (miRNAs) in HFSCs co-cultured with dermal papilla cells (named dHFSCs) and control HFSCs. We then determined the molecular function of miR-1 in HFSCs. Compared with HFSCs, 13 differentially transcribed miRNAs were identified in dHFSCs. The in vitro results indicated that the overtranscription of miR-1 inhibited HFSC proliferation, but enhanced HFSC differentiation by targeting IGF1R and LEF1 genes. This study provides new insights into the molecular mechanisms of HFSC development. Approval ID (2014ZX08008-002).
Collapse
Affiliation(s)
- Hailong Yan
- Department of Neurology, Institute of Brain Science, Medical School, Shanxi Datong University, Datong, China
- Shanxi key Laboratory of Inflammatory Neurodegenerative Disease, Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Miaohan Jin
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yan Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Ye Gao
- Department of Neurology, Institute of Brain Science, Medical School, Shanxi Datong University, Datong, China
- Shanxi key Laboratory of Inflammatory Neurodegenerative Disease, Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Qiang Ding
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiaolong Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Wenxian Zeng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yulin Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
24
|
Zhou X, Chen Y, Cui L, Shi Y, Guo C. Advances in the pathogenesis of psoriasis: from keratinocyte perspective. Cell Death Dis 2022; 13:81. [PMID: 35075118 PMCID: PMC8786887 DOI: 10.1038/s41419-022-04523-3] [Citation(s) in RCA: 282] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 12/14/2021] [Accepted: 01/11/2022] [Indexed: 02/08/2023]
Abstract
Psoriasis is a complex long-lasting inflammatory skin disease with high prevalence and associated comorbidity. It is characterized by epidermal hyperplasia and dermal infiltration of immune cells. Here, we review the role of keratinocytes in the pathogenesis of psoriasis, focusing on factors relevant to genetics, cytokines and receptors, metabolism, cell signaling, transcription factors, non-coding RNAs, antimicrobial peptides, and proteins with other different functions. The critical role of keratinocytes in initiating and maintaining the inflammatory state suggests the great significance of targeting keratinocytes for the treatment of psoriasis.
Collapse
Affiliation(s)
- Xue Zhou
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China
| | - Youdong Chen
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China
| | - Lian Cui
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China
| | - Yuling Shi
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China.
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China.
| | - Chunyuan Guo
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, 200443, Shanghai, China.
- Institute of Psoriasis, Tongji University School of Medicine, 200443, Shanghai, China.
| |
Collapse
|
25
|
Kurgyis Z, Vornholz L, Pechloff K, Kemény LV, Wartewig T, Muschaweckh A, Joshi A, Kranen K, Hartjes L, Möckel S, Steiger K, Hameister E, Volz T, Mellett M, French LE, Biedermann T, Korn T, Ruland J. Keratinocyte-intrinsic BCL10/MALT1 activity initiates and amplifies psoriasiform skin inflammation. Sci Immunol 2021; 6:eabi4425. [PMID: 34826258 DOI: 10.1126/sciimmunol.abi4425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Zsuzsanna Kurgyis
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany.,Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | - Larsen Vornholz
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Konstanze Pechloff
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Lajos V Kemény
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Dermatology, Venereology, and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Tim Wartewig
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Andreas Muschaweckh
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Abhinav Joshi
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Katja Kranen
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | - Lara Hartjes
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Sigrid Möckel
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany.,Institute of Pathology, Universität Würzburg, Würzburg, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Erik Hameister
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Thomas Volz
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | - Mark Mellett
- Department of Dermatology, University Hospital of Zürich, University of Zurich (UZH), Zürich, Switzerland
| | - Lars E French
- Department of Dermatology, University Hospital of Zürich, University of Zurich (UZH), Zürich, Switzerland.,Department of Dermatology and Allergy, University Hospital, LMU Munich Munich, Germany.,Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tilo Biedermann
- Department of Dermatology and Allergy, Technical University of Munich, Munich, Germany
| | - Thomas Korn
- Department of Experimental Neuroimmunology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Center for Infection Research (DZIF), Munich partner site, Munich Germany
| |
Collapse
|
26
|
Merlin J, Ivanov S, Dumont A, Sergushichev A, Gall J, Stunault M, Ayrault M, Vaillant N, Castiglione A, Swain A, Orange F, Gallerand A, Berton T, Martin JC, Carobbio S, Masson J, Gaisler-Salomon I, Maechler P, Rayport S, Sluimer JC, Biessen EAL, Guinamard RR, Gautier EL, Thorp EB, Artyomov MN, Yvan-Charvet L. Non-canonical glutamine transamination sustains efferocytosis by coupling redox buffering to oxidative phosphorylation. Nat Metab 2021; 3:1313-1326. [PMID: 34650273 PMCID: PMC7611882 DOI: 10.1038/s42255-021-00471-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 09/06/2021] [Indexed: 12/04/2022]
Abstract
Macrophages rely on tightly integrated metabolic rewiring to clear dying neighboring cells by efferocytosis during homeostasis and disease. Here we reveal that glutaminase-1-mediated glutaminolysis is critical to promote apoptotic cell clearance by macrophages during homeostasis in mice. In addition, impaired macrophage glutaminolysis exacerbates atherosclerosis, a condition during which, efficient apoptotic cell debris clearance is critical to limit disease progression. Glutaminase-1 expression strongly correlates with atherosclerotic plaque necrosis in patients with cardiovascular diseases. High-throughput transcriptional and metabolic profiling reveals that macrophage efferocytic capacity relies on a non-canonical transaminase pathway, independent from the traditional requirement of glutamate dehydrogenase to fuel ɑ-ketoglutarate-dependent immunometabolism. This pathway is necessary to meet the unique requirements of efferocytosis for cellular detoxification and high-energy cytoskeletal rearrangements. Thus, we uncover a role for non-canonical glutamine metabolism for efficient clearance of dying cells and maintenance of tissue homeostasis during health and disease in mouse and humans.
Collapse
Affiliation(s)
- Johanna Merlin
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Stoyan Ivanov
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Adélie Dumont
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | | | - Julie Gall
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Marion Stunault
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Marion Ayrault
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Nathalie Vaillant
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Alexia Castiglione
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Amanda Swain
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Francois Orange
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée (CCMA), Nice, France
| | - Alexandre Gallerand
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Thierry Berton
- Centre de Recherche Cardiovasculaire et Nutritionnelle (C2VN), INSERM, Institut National de la Recherche Agricole (INRA), BioMet, Aix-Marseille University, Marseille, France
| | - Jean-Charles Martin
- Centre de Recherche Cardiovasculaire et Nutritionnelle (C2VN), INSERM, Institut National de la Recherche Agricole (INRA), BioMet, Aix-Marseille University, Marseille, France
| | - Stefania Carobbio
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
- Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Justine Masson
- Inserm UMR-S1270, Institut du Fer à Moulin, Sorbonne Université, Paris, France
- Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY, USA
| | - Inna Gaisler-Salomon
- Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
- SPC-IBBR, University of Haifa, Haifa, Israel
| | - Pierre Maechler
- Department of Cell Physiology and Metabolism, University of Geneva Medical Centre, Geneva, Switzerland
| | - Stephen Rayport
- Department of Molecular Therapeutics, NYS Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
- Institute for Molecular Cardiovascular Research, RWTH Klinikum Aachen, Aachen, Germany
| | - Rodolphe R Guinamard
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | | | - Edward B Thorp
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Centre National de la Recherche Scientifique (CNRS) (R.G.), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France.
| |
Collapse
|
27
|
Cui SB, Wang TX, Liu ZW, Yan JY, Zhang K. Zinc finger protein A20 regulates the development and progression of osteoarthritis by affecting the activity of NF-κB p65. Immunopharmacol Immunotoxicol 2021; 43:713-723. [PMID: 34463587 DOI: 10.1080/08923973.2021.1970764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
OBJECTIVE To investigate the role of Zinc finger protein A20 in osteoarthritis (OA) by regulating NF-κB p65. METHODS A20, MMP1, MMP13 and IL-1β expressions in human OA cartilage samples were detected by qRT-PCR. IL-1β-induced chondrocyte was treated with A20 lentivirus activation particle, pyrrolidine dithiocarbamate (PDTC, a NF-κB inhibitor) with/without A20 siRNA. IL-6, TNF-α, and PGE2 levels were measured by ELISA, and NO production by Greiss reaction. Destabilization of the medial meniscus (DMM) surgery was used to construct the OA models, followed by injection of A20 adenovirus. MMP1 and MMP13 expression was measured by immunohistochemistry. The mRNA and protein expression were performed by qRT-PCR and western blotting, respectively. RESULTS A20 was down-regulated in human OA cartilage samples, and negatively correlated with the expressions of MMP1, MMP13 and IL-1β. The IL-1β-induced chondrocyte manifested decreased A20 with increased NF-κB p65 activity. A20 overexpression suppressed the NF-κB p65 activity in IL-1β-induced chondrocyte. Furthermore, PDTC decreased IL-1β-induced chondrocyte apoptosis with the upregulated COL1A1, COL2A1, COL10A1 and ACAN, as well as the down-regulated MMP1, MMP13, COX2, iNOS, IL-6, TNF-α, NO and PGE2, which was reversed by A20 siRNA. In vivo, OA mice gained higher OARSI score and Mankin's score, exhibited up-regulations of MMP1 and MMP13, and decreased NF-κB p65 activity, which was improved after injection of A20 adenovirus. CONCLUSION A20 was reduced in OA cartilage samples, and its overexpression, by suppressing the activity of NF-κB p65, could improve IL-1β-induced chondrocyte degradation and apoptosis in vitro, as well as mitigate the inflammation in OA mice.
Collapse
Affiliation(s)
- Shu-Bei Cui
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| | - Tao-Xia Wang
- Department of Nephrology, Affiliated Hospital of Hebei University of Technology, Handan, China
| | - Zhen-Wu Liu
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| | - Ji-Ying Yan
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| | - Kai Zhang
- The First Department of Orthopedics, Handan Central Hospital, Handan, China
| |
Collapse
|
28
|
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: 65] [Impact Index Per Article: 16.3] [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
|
29
|
Ito Y, Sasaki T, Li Y, Tanoue T, Sugiura Y, Skelly AN, Suda W, Kawashima Y, Okahashi N, Watanabe E, Horikawa H, Shiohama A, Kurokawa R, Kawakami E, Iseki H, Kawasaki H, Iwakura Y, Shiota A, Yu L, Hisatsune J, Koseki H, Sugai M, Arita M, Ohara O, Matsui T, Suematsu M, Hattori M, Atarashi K, Amagai M, Honda K. Staphylococcus cohnii is a potentially biotherapeutic skin commensal alleviating skin inflammation. Cell Rep 2021; 35:109052. [PMID: 33910010 DOI: 10.1016/j.celrep.2021.109052] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 03/09/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022] Open
Abstract
Host-microbe interactions orchestrate skin homeostasis, the dysregulation of which has been implicated in chronic inflammatory conditions such as atopic dermatitis and psoriasis. Here, we show that Staphylococcus cohnii is a skin commensal capable of beneficially inhibiting skin inflammation. We find that Tmem79-/- mice spontaneously develop interleukin-17 (IL-17)-producing T-cell-driven skin inflammation. Comparative skin microbiome analysis reveals that the disease activity index is negatively associated with S. cohnii. Inoculation with S. cohnii strains isolated from either mouse or human skin microbiota significantly prevents and ameliorates dermatitis in Tmem79-/- mice without affecting pathobiont burden. S. cohnii colonization is accompanied by activation of host glucocorticoid-related pathways and induction of anti-inflammatory genes in the skin and is therefore effective at suppressing inflammation in diverse pathobiont-independent dermatitis models, including chemically induced, type 17, and type 2 immune-driven models. As such, S. cohnii strains have great potential as effective live biotherapeutics for skin inflammation.
Collapse
Affiliation(s)
- Yoshihiro Ito
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Dermatology, Keio University School of Medicine, Tokyo 160-8582, Japan; JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo 160-8582, Japan; Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Takashi Sasaki
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Youxian Li
- Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Takeshi Tanoue
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan; JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo 160-8582, Japan; Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ashwin N Skelly
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Wataru Suda
- Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan; Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Yusuke Kawashima
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba 292-0818, Japan
| | - Nobuyuki Okahashi
- Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Eiichiro Watanabe
- Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Hiroto Horikawa
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Dermatology, Keio University School of Medicine, Tokyo 160-8582, Japan; Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Aiko Shiohama
- Department of Dermatology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Rina Kurokawa
- Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan; Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Eiryo Kawakami
- Medical Sciences Innovation Hub Program (MIH), RIKEN, Kanagawa 230-0045, Japan
| | - Hachiro Iseki
- Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Hiroshi Kawasaki
- Department of Dermatology, Keio University School of Medicine, Tokyo 160-8582, Japan; JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo 160-8582, Japan; Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan; Medical Sciences Innovation Hub Program (MIH), RIKEN, Kanagawa 230-0045, Japan
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
| | - Atsushi Shiota
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan; JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Liansheng Yu
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo 189-0002, Japan
| | - Junzo Hisatsune
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo 189-0002, Japan
| | - Haruhiko Koseki
- Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan; Medical Sciences Innovation Hub Program (MIH), RIKEN, Kanagawa 230-0045, Japan
| | - Motoyuki Sugai
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo 189-0002, Japan
| | - Makoto Arita
- Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Osamu Ohara
- Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan; Department of Applied Genomics, Kazusa DNA Research Institute, Chiba 292-0818, Japan
| | - Takeshi Matsui
- JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo 160-8582, Japan; Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masahira Hattori
- Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan; Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Koji Atarashi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan; JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo 160-8582, Japan; Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo 160-8582, Japan; JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo 160-8582, Japan; Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan
| | - Kenya Honda
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan; JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo 160-8582, Japan; Center for Integrative Medical Science (IMS), RIKEN, Kanagawa 230-0045, Japan.
| |
Collapse
|
30
|
Yuan W, Chen Y, Zhou Y, Bao K, Yu X, Xu Y, Zhang Y, Zheng J, Jiang G, Hong M. Formononetin attenuates atopic dermatitis by upregulating A20 expression via activation of G protein-coupled estrogen receptor. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113397. [PMID: 32971159 DOI: 10.1016/j.jep.2020.113397] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atopic dermatitis (AD) is a complex skin disease with highly heterogeneous inflammation, which ranks among the largest component of the nonfatal diseases worldwide. The medications currently used to treat AD primarily include antihistamines, vitamin D and anti-inflammatory drugs, etc. But, the usage of these drugs is usually accompanied by various side-effects. Formononetin (FMN), a natural active ingredient of Astragalus membranaceus (Fisch.) Bunge, decreases the AD relapse rate, reduces recurring severity incidence and resists the inflammation in the initial stage of AD. However, the underlying mechanism of FMN on repressing the development of AD is still unknown. AIM OF THE STUDY To investigate the potential mechanism of FMN on relieving the initial responses of AD and elucidate its possible therapeutic targets in vivo and in vitro. MATERIALS AND METHODS A fluorescein isothiocyanate (FITC)-induced mouse model of the initial stage of AD was established in vivo. Human keratinocytes (HaCaT) cells were co-stimulated with tumor necrosis factor alpha (TNF-α) and polyinosinic-polycytidylic acid (Poly(I:C)) in vitro. The production of thymic stromal lymphopoietin (TSLP) and immunoglobulin E (IgE) were detected by enzyme-linked immunosorbnent assay (ELISA). The protein expression was measured through immunohistochemistry and western blotting. The mRNA expression was examined by real-time quantitative polymerase chain reaction (RT-qPCR). The impact of TNF-α-induced protein 3 (TNFAIP3/A20) was reflected using its small interfering RNA (siRNA). The role of G protein-coupled estrogen receptor (GPER) was explored using its agonist (G1), antagonist (G15) or siRNA (siGPER) in vitro. RESULTS We found that FMN upregulated the expression of A20 protein and mRNA in the initial stage of AD model, especially in the epithelial region of ear tissue, and inhibited the production of TSLP simultaneously. Consistently, FMN significantly upregulated A20 protein and its mRNA expression while reduced TSLP protein and its mRNA expression in vitro, and this effect could be antagonized by A20 siRNA (siA20). Moreover, compared with PPT (ERα agonist) and DPN (ERβ agonist), G1 could significantly increase the expression of A20. In addition, compared with MPP (ERα antagonist) and PHTPP (ERβ antagonist), G15 could markedly reduce the expression of A20. Furthermore, the effects of FMN on A20 were interfered by siGPER and G15 in vitro and in vivo. CONCLUSIONS These results demonstrated that FMN attenuated AD by upregulating A20 expression via activation of GPER. This new strategy might have effective therapeutic potential for AD and other inflammatory disorders.
Collapse
Affiliation(s)
- Weiyuan Yuan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, 215003, China.
| | - Yanyan Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yijing Zhou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Kaifan Bao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xuerui Yu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yifan Xu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yuheng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Jie Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Department of Pharmacology, School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Guorong Jiang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, 215003, China.
| | - Min Hong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
31
|
Abstract
Inflammation is triggered by stimulation of innate sensors that recognize pathogens, chemical and physical irritants, and damaged cells subsequently initiating a well-orchestrated adaptive immune response. Immune cell activation is a strictly regulated and self-resolving process supported by an array of negative feedback mechanisms to sustain tissue homeostasis. The disruption of these regulatory pathways forms the basis of chronic inflammatory diseases, including periodontitis. Ubiquitination, a covalent posttranslational modification of target proteins with ubiquitin, has a profound effect on the stability and activity of its substrates, thereby regulating the immune system at molecular and cellular levels. Through the cooperative actions of E3 ubiquitin ligases and deubiquitinases, ubiquitin modifications are implicated in several biological processes, including proteasomal degradation, transcriptional regulation, regulation of protein-protein interactions, endocytosis, autophagy, DNA repair, and cell cycle regulation. A20 (tumor necrosis factor α-induced protein 3 or TNFAIP3) is a ubiquitin-editing enzyme that mainly functions as an endogenous regulator of inflammation through termination of nuclear factor (NF)-κB activation as part of a negative feedback loop. A20 interacts with substrates that reside downstream of immune sensors, including Toll-like receptors, nucleotide-binding oligomerization domain-containing receptors, lymphocyte receptors, and cytokine receptors. Due to its pleiotropic functions as a ubiquitin binding protein, deubiquitinase and ubiquitin ligase, and its versatile role in various signaling pathways, aberrant A20 levels are associated with numerous conditions such as rheumatoid arthritis, diabetes, systemic lupus erythematosus, inflammatory bowel disease, psoriasis, Sjögren syndrome, coronary artery disease, multiple sclerosis, cystic fibrosis, asthma, cancer, neurological disorders, and aging-related sequelae. Similarly, A20 has recently been implicated as an essential regulator of inflammation in the oral cavity. This review presents information on the ubiquitin system and regulation of NF-κB by ubiquitination using A20 as a representative molecule and highlights how the dysregulation of this system can lead to several immune pathologies, including oral cavity-related disorders mainly focusing on periodontitis.
Collapse
Affiliation(s)
- E.C. Mooney
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Philips Institute for Oral Health Research, Virginia Commonwealth University, School of Dentistry, Richmond, VA, USA
| | - S.E. Sahingur
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
32
|
Abstract
A20/TNFAIP3 is a TNF induced gene that plays a profound role in preserving cellular and organismal homeostasis (Lee, et al., 2000; Opipari etal., 1990). This protein has been linked to multiple human diseases via genetic, epigenetic, and an emerging series of patients with mono-allelic coding mutations. Diverse cellular functions of this pleiotropically expressed protein include immune-suppressive, anti-inflammatory, and cell protective functions. The A20 protein regulates ubiquitin dependent cell signals; however, the biochemical mechanisms by which it performs these functions is surprisingly complex. Deciphering these cellular and biochemical facets of A20 dependent biology should greatly improve our understanding of murine and human disease pathophysiology as well as unveil new mechanisms of cell and tissue biology.
Collapse
Affiliation(s)
- Bahram Razani
- Department of Dermatology, University of California, San Francisco, CA, United States
| | - Barbara A Malynn
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States.
| |
Collapse
|
33
|
Transcriptome Profiling Analyses in Psoriasis: A Dynamic Contribution of Keratinocytes to the Pathogenesis. Genes (Basel) 2020; 11:genes11101155. [PMID: 33007857 PMCID: PMC7600703 DOI: 10.3390/genes11101155] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
Psoriasis is an immune-mediated inflammatory skin disease with a complex etiology involving environmental and genetic factors. A better insight into related genomic alteration helps design precise therapies leading to better treatment outcome. Gene expression in psoriasis can provide relevant information about the altered expression of mRNA transcripts, thus giving new insights into the disease onset. Techniques for transcriptome analyses, such as microarray and RNA sequencing (RNA-seq), are relevant tools for the discovery of new biomarkers as well as new therapeutic targets. This review summarizes the findings related to the contribution of keratinocytes in the pathogenesis of psoriasis by an in-depth review of studies that have examined psoriatic transcriptomes in the past years. It also provides valuable information on reconstructed 3D psoriatic skin models using cells isolated from psoriatic patients for transcriptomic studies.
Collapse
|
34
|
Mifflin L, Ofengeim D, Yuan J. Receptor-interacting protein kinase 1 (RIPK1) as a therapeutic target. Nat Rev Drug Discov 2020; 19:553-571. [PMID: 32669658 PMCID: PMC7362612 DOI: 10.1038/s41573-020-0071-y] [Citation(s) in RCA: 302] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2020] [Indexed: 02/08/2023]
Abstract
Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a key mediator of cell death and inflammation. The unique hydrophobic pocket in the allosteric regulatory domain of RIPK1 has enabled the development of highly selective small-molecule inhibitors of its kinase activity, which have demonstrated safety in preclinical models and clinical trials. Potential applications of these RIPK1 inhibitors for the treatment of monogenic and polygenic autoimmune, inflammatory, neurodegenerative, ischaemic and acute conditions, such as sepsis, are emerging. This article reviews RIPK1 biology and disease-associated mutations in RIPK1 signalling pathways, highlighting clinical trials of RIPK1 inhibitors and potential strategies to mitigate development challenges. Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) — a key mediator of cell death and inflammation — is activated in human diseases. Here, Yuan and colleagues discuss current understanding of RIPK1 biology and its association with diseases including inflammatory and autoimmune disorders, neurodegenerative diseases and sepsis. The clinical development of small-molecule RIPK1 inhibitors and associated challenges are discussed.
Collapse
Affiliation(s)
- Lauren Mifflin
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Dimitry Ofengeim
- Rare and Neurologic Disease Research, Sanofi, Framingham, MA, USA
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
35
|
Billi AC, Gudjonsson JE, Voorhees JJ. Psoriasis: Past, Present, and Future. J Invest Dermatol 2020; 139:e133-e142. [PMID: 31648690 DOI: 10.1016/j.jid.2019.08.437] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/09/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Allison C Billi
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| | | | - John J Voorhees
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
36
|
Immune Regulation of TNFAIP3 in Psoriasis through Its Association with Th1 and Th17 Cell Differentiation and p38 Activation. J Immunol Res 2020; 2020:5980190. [PMID: 32280718 PMCID: PMC7114769 DOI: 10.1155/2020/5980190] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/14/2020] [Accepted: 02/24/2020] [Indexed: 12/26/2022] Open
Abstract
Background Psoriasis is an immune-mediated chronic inflammatory skin disorder in which the dysregulation of immune cells plays an important role in its development. Tumor necrosis factor- (TNF-) α antagonists affect the immune repertoire, while TNF-α-induced protein 3 (TNFAIP3) has a protective role against the deleterious effects of inflammation and participates in immune regulation. Objective We investigated the immune regulation of TNFAIP3 in the pathogenesis of psoriasis and determined whether it is involved in the antipsoriatic effect of TNF-α antagonists. Methods mRNA levels were evaluated in blood from patients with moderate-to-severe psoriasis. The effects of TNF-α antagonists were examined in a mouse imiquimod- (IMQ-) induced psoriasis-like dermatitis model. In the mouse model, TNFAIP3 mRNA expression was determined using RT-PCR. Serum levels of IL-17A, IL-23, IFN-γ, TNF-α, phosphorylated ERK1/2, p38, and JNK were measured using ELISA. The proportion of Th1 and Th17 cells in mouse spleens was analyzed using flow cytometry. Results mRNA expression levels of TNFAIP3 in the blood were significantly lower in patients with moderate and severe psoriasis (mean ± SD = 0.44 ± 0.25) compared with normal subjects (mean ± SD = 1.00 ± 0.82) (P < 0.01). In the mouse model, IMQ downregulated TNFAIP3 expression levels, which were increased after TNF-α antagonist treatment (P < 0.05). Serum levels of Th17 cytokines (IL-17A and IL-23) and Th1 cytokines (IFN-γ and TNF-α) were significantly higher in the IMQ and IMQ/rat IgG1 groups compared with the control group, and the application of TNF-α antagonists significantly decreased the levels of inflammatory cytokines (P < 0.01). Notably, phosphorylated p38 levels were increased in the IMQ and IMQ/rat IgG1 groups compared with the control group but were downregulated by treatment with TNF-α antagonists (P < 0.05). Th1 and Th17 cells were significantly increased in the IMQ group compared with the control group (P < 0.01). Conclusion TNFAIP3 downregulation associated with Th1 and Th17 cell differentiation and p38 activation might contribute in part to the mechanism of immune dysfunction in psoriasis. TNF-α antagonists might partly exert their effects on psoriasis via this pathway.
Collapse
|
37
|
Li Y, Mooney EC, Xia XJ, Gupta N, Sahingur SE. A20 Restricts Inflammatory Response and Desensitizes Gingival Keratinocytes to Apoptosis. Front Immunol 2020; 11:365. [PMID: 32218782 PMCID: PMC7078700 DOI: 10.3389/fimmu.2020.00365] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 02/14/2020] [Indexed: 12/16/2022] Open
Abstract
The pathophysiology of periodontal disease involves a perturbed immune system to a dysbiotic microflora leading to unrestrained inflammation, collateral tissue damage, and various systemic complications. Gingival epithelial cells function as an important part of immunity to restrict microbial invasion and orchestrate the subsequent innate responses. A20 (TNFAIP3), an ubiquitin-editing enzyme, is one of the key regulators of inflammation and cell death in numerous tissues including gastrointestinal tract, skin, and lungs. Emerging evidence indicates A20 as an essential molecule in the oral mucosa as well. In this study, we characterized the role of A20 in human telomerase immortalized gingival keratinocytes (TIGKs) through loss and gain of function assays in preclinical models of periodontitis. Depletion of A20 through gene editing in TIGKs significantly increased IL-6 and IL-8 secretion in response to Porphyromonas gingivalis infection while A20 over-expression dampened the cytokine production compared to A20 competent cells through modulating NF-κB signaling pathway. In the subsequent experiments which assessed apoptosis, A20 depleted TIGKs displayed increased levels of cleaved caspase 3 and DNA fragmentation following P. gingivalis infection and TNF/CHX challenge compared to A20 competent cells. Consistently, there was reduced apoptosis in the cells overexpressing A20 compared to the control cells expressing GFP further substantiating the role of A20 in regulating gingival epithelial cell fate in response to exogenous insult. Collectively, our findings reveal first systematic evidence and demonstrate that A20 acts as a regulator of inflammatory response in gingival keratinocytes through its effect on NF-κB signaling and desensitizes cells to bacteria and cytokine induced apoptosis in the oral mucosa. As altered A20 levels can have profound effect on different cellular responses, future studies will determine whether A20-targeted therapies can be exploited to restrain periodontal inflammation and maintain oral mucosa tissue homeostasis.
Collapse
Affiliation(s)
- Yajie Li
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Erin C Mooney
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,School of Dentistry, Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, VA, United States
| | - Xia-Juan Xia
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nitika Gupta
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sinem Esra Sahingur
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
38
|
Martens A, van Loo G. A20 at the Crossroads of Cell Death, Inflammation, and Autoimmunity. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a036418. [PMID: 31427375 DOI: 10.1101/cshperspect.a036418] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A20 is a potent anti-inflammatory protein, acting by inhibiting nuclear factor κB (NF-κB) signaling and inflammatory gene expression and/or by preventing cell death. Mutations in the A20/TNFAIP3 gene have been associated with a plethora of inflammatory and autoimmune pathologies in humans and in mice. Although the anti-inflammatory role of A20 is well accepted, fundamental mechanistic questions regarding its mode of action remain unclear. Here, we review new findings that further clarify the molecular and cellular mechanisms by which A20 controls inflammatory signaling and cell death, and discuss new evidence for its involvement in inflammatory and autoimmune disease development.
Collapse
Affiliation(s)
- Arne Martens
- VIB Center for Inflammation Research, 9052 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, 9052 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| |
Collapse
|
39
|
Functional analysis of deubiquitylating enzymes in tumorigenesis and development. Biochim Biophys Acta Rev Cancer 2019; 1872:188312. [DOI: 10.1016/j.bbcan.2019.188312] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 02/06/2023]
|
40
|
Lorscheid S, Müller A, Löffler J, Resch C, Bucher P, Kurschus FC, Waisman A, Schäkel K, Hailfinger S, Schulze-Osthoff K, Kramer D. Keratinocyte-derived IκBζ drives psoriasis and associated systemic inflammation. JCI Insight 2019; 4:130835. [PMID: 31622280 PMCID: PMC6948851 DOI: 10.1172/jci.insight.130835] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
The transcriptional activator IκBζ is a key regulator of psoriasis, but which cells mediate its pathogenic effect remains unknown. Here we found that IκBζ expression in keratinocytes triggers not only skin lesions but also systemic inflammation in mouse psoriasis models. Specific depletion of IκBζ in keratinocytes was sufficient to suppress the induction of imiquimod- or IL-36–mediated psoriasis. Moreover, IκBζ ablation in keratinocytes prevented the onset of psoriatic lesions and systemic inflammation in keratinocyte-specific IL-17A–transgenic mice. Mechanistically, this psoriasis protection was mediated by IκBζ deficiency in keratinocytes abrogating the induction of specific proinflammatory target genes, including Cxcl5, Cxcl2, Csf2, and Csf3, in response to IL-17A or IL-36. These IκBζ-dependent genes trigger the generation and recruitment of neutrophils and monocytes that are needed for skin inflammation. Consequently, our data uncover a surprisingly pivotal role of keratinocytes and keratinocyte-derived IκBζ as key mediators of psoriasis and psoriasis-related systemic inflammation. Deletion of IκBζ in keratinocytes is sufficient to abrogate psoriasis induction in mouse models due to changes in transcription of keratinocyte-derived chemo- and cytokines.
Collapse
Affiliation(s)
- Sebastian Lorscheid
- Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Anne Müller
- Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Jessica Löffler
- Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Claudia Resch
- Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Philip Bucher
- Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Florian C Kurschus
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany.,Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Knut Schäkel
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephan Hailfinger
- Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
| | - Klaus Schulze-Osthoff
- Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany.,German Cancer Consortium and German Cancer Research Center, Heidelberg, Germany
| | - Daniela Kramer
- Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| |
Collapse
|
41
|
Gularte MS, Anghinoni JM, Abenante L, Voss GT, de Oliveira RL, Vaucher RA, Luchese C, Wilhelm EA, Lenardão EJ, Fajardo AR. Synthesis of chitosan derivatives with organoselenium and organosulfur compounds: Characterization, antimicrobial properties and application as biomaterials. Carbohydr Polym 2019; 219:240-250. [DOI: 10.1016/j.carbpol.2019.05.040] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/11/2019] [Accepted: 05/10/2019] [Indexed: 01/09/2023]
|
42
|
Sacco KA, Milner JD. Gene-environment interactions in primary atopic disorders. Curr Opin Immunol 2019; 60:148-155. [PMID: 31302571 DOI: 10.1016/j.coi.2019.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/14/2019] [Indexed: 12/18/2022]
Abstract
Environmental factors modify disease presentation and severity in allergic disorders. Primary atopic disorders (PADs) are a heterogenous group of single gene disorders that lead to significant atopic and allergic disease manifestations. However, a number of these monogenic diseases have variable penetrance suggesting that gene-gene and/or gene-environment interactions could modulate the clinical phenotype. Environmental factors such as diet, the microbiome at the epithelial-environment interface, the presence and/or extent of infection, and psychologic stress can alter disease phenotypic expression of allergic diseases, and PADs provide discrete contexts in which to understand these influences. We outline how gene-environment interactions likely contribute to a variable penetrance and expressivity in PADs. Dietary modifications of both macronutrients and/or micronutrients alter T-cell metabolism and may influence effector T-cell function. The mucosal microbiome may affect local inflammation and may remotely influence regulatory elements, while psychologic stress can affect mast cell and other allergic effector cell function. Understanding gene-environment interactions in PADs can hopefully provide a foundation for interrogating gene-environment interactions to common allergic disorders, and also present opportunities for personalized interventions based on the altered pathways and environmental influences in affected individuals.
Collapse
Affiliation(s)
- Keith A Sacco
- Laboratory of Allergic Diseases, NIAID, NIH, 9000 Rockville Pike, NIH Building 10 Room 11N240A, United States
| | - Joshua D Milner
- Laboratory of Allergic Diseases, NIAID, NIH, 9000 Rockville Pike, NIH Building 10 Room 11N240A, United States.
| |
Collapse
|
43
|
Sahlol NY, Mostafa MS, Madkour LAEF, Salama DM. Low TNFAIP3 expression in psoriatic skin promotes disease susceptibility and severity. PLoS One 2019; 14:e0217352. [PMID: 31120955 PMCID: PMC6532901 DOI: 10.1371/journal.pone.0217352] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/29/2019] [Indexed: 12/13/2022] Open
Abstract
Psoriasis vulgaris is a systemic disorder with an underlying immune dysregulation that predisposes to inflammatory skin lesions. Meanwhile, tumor necrosis factor alpha-induced protein 3 (TNFAIP3) has been described as a protective molecule against the deleterious effects of uncontrolled inflammation. In this study, we compared the expression levels of TNFAIP3 in blood and psoriatic skin biopsies from psoriatic patients versus those in normal individuals. Additionally, the levels of TNFAIP3 protein in psoriatic skin biopsies were compared to those in normal individuals. Thirty psoriatic patients and 30 healthy participants (control group) were enrolled. The expression levels of TNFAIP3 in blood and skin were measured by quantitative reverse transcription PCR, while the skin levels of TNFAIP3 protein were measured by western blot. Psoriatic patients showed significantly lower expression levels of TNFAIP3 in psoriatic skin and blood (P< 0.001) as well as of TNFAIP3 protein in psoriatic skin (P< 0.001) compared to controls. A significant lower expression of TNFAIP3 and TNFAIP3 protein in psoriatic skin was detected in moderate/severe cases compared to mild cases (P = 0.004 and 0.003 respectively). Moreover, a significant negative correlation was found between TNFAIP3 mRNA in psoriatic tissue and psoriasis area severity index values (rs = -0.382, P-value = 0.037). In conclusion, TNFAIP3 may serve as a predictive and prognostic biomarker in psoriatic patients. Enhancing the expression and/or function of TNFAIP3 in the affected cell type may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Nahla Yassin Sahlol
- Department of Microbiology and Immunology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Marwa Salah Mostafa
- Department of Microbiology and Immunology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | |
Collapse
|
44
|
Malynn BA, Ma A. A20: A multifunctional tool for regulating immunity and preventing disease. Cell Immunol 2019; 340:103914. [PMID: 31030956 DOI: 10.1016/j.cellimm.2019.04.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/03/2019] [Indexed: 02/07/2023]
Abstract
A20, also known as TNFAIP3, is a potent regulator of ubiquitin (Ub) dependent signals. A20 prevents multiple human diseases, indicating that the critical functions of this protein are clinically as well as biologically impactful. As revealed by mouse models, cell specific functions of A20 are linked to its ability to regulate diverse signaling pathways. Aberrant expression or functions of A20 in specific cell types underlie divergent disease outcomes. Discernment of A20's biochemical functions and their phenotypic outcomes will contribute to our understanding of how ubiquitination is regulated, how Ub mediated functions can prevent disease, and will pave the way for future therapeutic interventions.
Collapse
Affiliation(s)
- Barbara A Malynn
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, United States.
| |
Collapse
|
45
|
Herter EK, Li D, Toma MA, Vij M, Li X, Visscher D, Wang A, Chu T, Sommar P, Blomqvist L, Berglund D, Ståhle M, Wikstrom JD, Xu Landén N. WAKMAR2, a Long Noncoding RNA Downregulated in Human Chronic Wounds, Modulates Keratinocyte Motility and Production of Inflammatory Chemokines. J Invest Dermatol 2018; 139:1373-1384. [PMID: 30594489 DOI: 10.1016/j.jid.2018.11.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/29/2018] [Accepted: 11/10/2018] [Indexed: 01/08/2023]
Abstract
Chronic wounds represent a major and growing health and economic burden worldwide. A better understanding of molecular mechanisms of normal as well as impaired wound healing is needed to develop effective treatment. Herein we studied the potential role of long noncoding RNA LOC100130476 in skin wound repair. LOC100130476 is an RNA polymerase II-encoded polyadenylated transcript present in both cytoplasm and nucleus. We found that its expression was lower in wound-edge keratinocytes of human chronic wounds compared to normal wounds of healthy donors and intact skin. In cultured keratinocytes, LOC100130476 expression was induced by TGF-β signaling. By reducing LOC100130476 expression with antisense oligos or activating its transcription with CRISPR/Cas9 Synergistic Activation Mediator system, we showed that LOC100130476 restricted the production of inflammatory chemokines by keratinocytes, while enhancing cell migration. In line with this, knockdown of LOC100130476 impaired re-epithelization of human ex vivo wounds. Based on these results, we named LOC100130476 wound and keratinocyte migration-associated long noncoding RNA 2 (WAKMAR2). Moreover, we identified a molecular network that may mediate the biological function of WAKMAR2 in keratinocytes using microarray. In summary, our data suggest that WAKMAR2 is an important regulator of skin wound healing and its deficiency may contribute to the pathogenesis of chronic wounds.
Collapse
Affiliation(s)
- Eva K Herter
- Dermatology and Venereology Section, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Dongqing Li
- Dermatology and Venereology Section, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Maria A Toma
- Dermatology and Venereology Section, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Manika Vij
- Dermatology and Venereology Section, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Xi Li
- Dermatology and Venereology Section, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Dani Visscher
- Dermatology and Venereology Section, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden
| | - Aoxue Wang
- Department of Dermatology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Tongbin Chu
- Department of Wound Regeneration, The Second Hospital of Dalian Medical University, Dalian, China
| | - Pehr Sommar
- Department of Molecular Medicine and Surgery, Section of Plastic Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Lennart Blomqvist
- Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - David Berglund
- Department of Immunology, Genetics and Pathology, Section of Clinical Immunology, Uppsala University, Sweden
| | - Mona Ståhle
- Dermatology and Venereology Section, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden; Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Jakob D Wikstrom
- Dermatology and Venereology Section, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden; Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Ning Xu Landén
- Dermatology and Venereology Section, Department of Medicine (Solna), Karolinska Institute, Stockholm, Sweden; Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|