1
|
Zaccagnini G, Baci D, Tastsoglou S, Cozza I, Madè A, Voellenkle C, Nicoletti M, Ruatti C, Longo M, Perani L, Gaetano C, Esposito A, Martelli F. miR-210 overexpression increases pressure overload-induced cardiac fibrosis. Noncoding RNA Res 2025; 12:20-33. [PMID: 40034123 PMCID: PMC11874870 DOI: 10.1016/j.ncrna.2025.01.009] [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: 03/21/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 03/05/2025] Open
Abstract
Aortic stenosis, a common valvular heart disease, can lead to left ventricular pressure overload, triggering pro-fibrotic responses in the heart. miR-210 is a microRNA that responds to hypoxia and ischemia and plays a role in immune regulation and in cardiac remodeling upon myocardial infarction. This study investigated the effects of miR-210 on cardiac fibrosis caused by pressure overload. Using a mouse model with inducible miR-210 over-expression, we subjected mice to transverse aortic constriction (TAC) to induce pressure overload. Mice with miR-210 over-expression developed eccentric hypertrophy, heightened expression of hypertrophic markers (Nppa and Nppb) and increased cross sectional area of cardiomyocytes, impacting the free wall of the left ventricle. These findings suggest that miR-210 worsens cardiac dysfunction. Furthermore, miR-210 over-expression led to a more robust and sustained inflammatory response in the heart, increased interstitial and perivascular fibrosis, and activation of myofibroblasts. miR-210 also promoted angiogenesis. In vitro, cardiac fibroblasts over-expressing miR-210 showed increased adhesion, wound healing and migration capacity. Our results demonstrate that miR-210 contributes to adverse cardiac remodeling in response to pressure overload, including eccentric hypertrophy, inflammation, and fibrosis.
Collapse
Affiliation(s)
- G. Zaccagnini
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - D. Baci
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - S. Tastsoglou
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - I. Cozza
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - A. Madè
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - C. Voellenkle
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - M. Nicoletti
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - C. Ruatti
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - M. Longo
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
| | - L. Perani
- Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
| | - C. Gaetano
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| | - A. Esposito
- Preclinical Imaging Facility, Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
- Vita-Salute San Raffaele University, Milan, 20132, Italy
| | - F. Martelli
- Laboratory of Molecular Cardiology, IRCCS Policlinico San Donato, San Donato Milanese, Milan, 20097, Italy
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| |
Collapse
|
2
|
Dashti M, Mohammadi M, Dehnavi S, Sadeghi M. Immunomodulatory interactions between mesenchymal stromal/stem cells and immune cells in psoriasis: therapeutic potential and challenges. Stem Cell Res Ther 2025; 16:241. [PMID: 40369681 PMCID: PMC12080136 DOI: 10.1186/s13287-025-04375-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
Psoriasis is defined as a persistent autoimmune disease characterized by the appearance of psoriatic lesions on the surface of the skin. Currently, various approaches including chemicals, corticosteroids, phototherapy, and biological agents are being proposed and implemented to improve psoriatic lesions by modulating immune system activity or metabolic processes, often with unintended consequences and side effects. Currently, mesenchymal stromal/stem cells (MSCs) have attracted considerable interest among researchers due to their ability to modulate immune responses and their ease of application, representing a promising strategy for alleviating clinical symptoms in the treatment of allergic reactions, autoimmune diseases, cancer, and more. This study will investigate how MSCs interact with immune system cells involved in psoriasis development, such as neutrophils, keratinocytes, dendritic cells (DC), and T cell subtypes, for potential therapeutic use in psoriasis management. In this case, several immunomodulatory mechanisms are involved, including expression of chemokines, pro-inflammatory cytokines, matrix metalloproteinase and other factors involved in cell proliferation and neutrophil extracellular trap (NET) formation are among the effects of MSCs on keratinocytes and neutrophils. keratinocytes and neutrophils as pro-inflammatory cells involved in psoriasis pathogenesis and pathogenesis and progression of psoriasis. On the other hand, MSCs interact with DCs and various subsets of T cells, including Th1, Th2, Th17 and Tregs, to generate tolerogenic DCs and increase the differentiation of Tregs and modulate the Th17/Treg towards a regulatory state through overexpression of anti-inflammatory and immunomodulatory and immunomodulatory cytokines, including IL-10 and transforming growth Factor beta (TGF-β). Finally, we will focus on the challenges and obstacles in psoriasis treatment using MSCs, including limitations in the case of using MSCs from different sources and side effects that may be encountered by whole cell therapy strategies, which are attracting attention towards the implication of cell-free regimens such as using MSC-derived secretome or extracellular vesicles and exosomes to provide similar therapeutic outcomes without presumed side effects.
Collapse
Affiliation(s)
- Mohammadreza Dashti
- Kashmar School of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Mohammadi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Dehnavi
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mahvash Sadeghi
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Lin L, Li W, Gao X, Li Q, Zhou X, Liu W, Zhong X, Yang Y, Zhang X, Luo Q. The effect of plasma exosomal microRNA- 148a- 3p on the CD4 + T cell function and its mechanism in the pathogenesis of psoriasis. Arch Dermatol Res 2025; 317:732. [PMID: 40261424 DOI: 10.1007/s00403-025-04197-9] [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: 01/02/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/24/2025]
Abstract
Psoriasis represents a chronic inflammatory skin disease occurring globally. We investigated the role of plasma exosomal microRNA (miR)- 148a- 3p and its target gene Bim in psoriasis. Plasma exosomes (Exos) and CD4+ T cells were extracted from psoriatic patients. miR- 148a- 3p expression in Exos and CD4+ T cells of psoriatic patients, the proportions of CD4+ T cell subsets, and the contents of anti-inflammatory/pro-inflammatory factors were determined by RT-qPCR, flow cytometry and ELISA. The correlation between plasma exosomal miR- 148a- 3p and CD4+ T cell subsets in psoriatic patients was analyzed by Pearson's analysis. The psoriatic mouse was treated with Exos/antagomir miR- 148a- 3p. Histopathological changes in the skin were observed. The CD4+ T cell subset levels, serum cytokine contents and miR- 148a- 3p expression in the blood were measured. The miR- 148a- 3p-Bim targeted binding relationship was predicted and verified by Starbase database and dual-luciferase assay. The Bim expression in psoriatic mice was determined. Psoriatic patients had highly-expressed miR- 148a- 3p in both Exos and CD4+ T cells, and abnormal CD4+ T cell subsets and cytokine levels. Plasma exosomal miR- 148a- 3p was correlated with the CD4+ T cell subsets in psoriatic patients. Exos caused down-regulated miR- 148a- 3p level in skin tissues of mouse, regulated CD4+ T cell function and aggravated the symptoms in psoriatic mice. miR- 148a- 3p repression partially reversed the role of Exos in CD4+ T cell function and psoriasis-like symptoms. Exos-carried miR- 148a- 3p targeted Bim in CD4+ T cells of psoriatic mice. Plasma exosomal miR- 148a- 3p targeted Bim to affect the dysfunction of CD4+ T cells in psoriatic mice, thereby aggravating the psoriasis-like symptoms.
Collapse
Grants
- No. 2024A03J0483 Guangzhou Basic Research Plan Jointly the City, School (Hospital), and/or Enterprise
- No. 2024A03J0483 Guangzhou Basic Research Plan Jointly the City, School (Hospital), and/or Enterprise
- No. 2024A03J0483 Guangzhou Basic Research Plan Jointly the City, School (Hospital), and/or Enterprise
- No. 2024A03J0483 Guangzhou Basic Research Plan Jointly the City, School (Hospital), and/or Enterprise
- No. 2024A03J0483 Guangzhou Basic Research Plan Jointly the City, School (Hospital), and/or Enterprise
- No. 2024A03J0483 Guangzhou Basic Research Plan Jointly the City, School (Hospital), and/or Enterprise
- No. 2024A03J0483 Guangzhou Basic Research Plan Jointly the City, School (Hospital), and/or Enterprise
- No. 2024A03J0483 Guangzhou Basic Research Plan Jointly the City, School (Hospital), and/or Enterprise
- No. 2024A03J0483 Guangzhou Basic Research Plan Jointly the City, School (Hospital), and/or Enterprise
- No. 2024A03J0483 Guangzhou Basic Research Plan Jointly the City, School (Hospital), and/or Enterprise
Collapse
Affiliation(s)
- Ling Lin
- Department of Dermatology, Guangzhou Dermatology Hospital, No. 56 Hengfu Road, Guangzhou, 510095, China
| | - Wei Li
- Department of Dermatology, Guangzhou Dermatology Hospital, No. 56 Hengfu Road, Guangzhou, 510095, China
| | - Xinjing Gao
- Department of Dermatology, Guangzhou Women and Children'S Medical Center, Guangzhou Medical University, Guangzhou, 510620, People's Republic of China
| | - Qian Li
- Department of Dermatology, Guangzhou Dermatology Hospital, No. 56 Hengfu Road, Guangzhou, 510095, China
| | - Xin Zhou
- Department of Dermatology, Guangzhou Dermatology Hospital, No. 56 Hengfu Road, Guangzhou, 510095, China
| | - Weiyu Liu
- Department of Dermatology, Guangzhou Dermatology Hospital, No. 56 Hengfu Road, Guangzhou, 510095, China
| | - Xuelian Zhong
- Department of Dermatology, Guangzhou Dermatology Hospital, No. 56 Hengfu Road, Guangzhou, 510095, China
| | - Yunqing Yang
- Department of Dermatology, Guangzhou Dermatology Hospital, No. 56 Hengfu Road, Guangzhou, 510095, China
| | - Xibao Zhang
- Department of Dermatology, Guangzhou Dermatology Hospital, No. 56 Hengfu Road, Guangzhou, 510095, China
| | - Quan Luo
- Department of Dermatology, Guangzhou Dermatology Hospital, No. 56 Hengfu Road, Guangzhou, 510095, China.
| |
Collapse
|
4
|
Shu F, Wang Y, Zhang F, Li L, Mao D. Exploring the Causal Role of miR-941 in Chronic Rhinosinusitis: Insights from Transcriptomics and Genomics. Int J Gen Med 2025; 18:1983-1998. [PMID: 40231245 PMCID: PMC11994465 DOI: 10.2147/ijgm.s511534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Purpose Despite significant advancements in the epigenetics of chronic rhinosinusitis (CRS), particularly in the domain of microRNA (miRNA), little is known about miRNAs that play a causal role in CRS. This study aims to identify miRNAs with a causal relationship to CRS and explore their potential clinical value and mechanisms in CRS. Methods We conducted small RNA sequencing on blood and nasal samples to find miRNAs with consistent expression differences in CRS. These miRNAs were confirmed via qRT-PCR and assessed for clinical relevance through Spearman correlation and statistical analysis used to evaluate diagnostic accuracy. Bidirectional Mendelian randomization (MR, a genetic causal inference method) analyzed their causal links to CRS. Target genes of causally significant miRNAs were identified using miRWalk, and their mechanisms were explored through pathway enrichment and validation studies. Results We identified differentially expressed miRNAs in blood and nasal tissues using a |log2(Fold Change)| > 0.58 and P-value < 0.05 threshold. Following False Discovery Rate correction, hsa-miR-941 was identified as an upregulated miRNA in both CRS patient samples. The experimental validation of miR-941 expression closely matched sequencing results. Spearman and statistical analysis associated miR-941 expression changes with CRS severity and diagnostic accuracy. Bidirectional MR demonstrated a significant association of miR-941 with CRS risk, without evidence of reverse causality. Target gene and Western blot assays suggested miR-941's potential influence on CRS through the PI3K/AKT pathway. Conclusion There is a positive causal relationship between hsa-miR-941 and CRS, making hsa-miR-941 a valuable target for the diagnosis and treatment of CRS. These findings position miR-941 as a valuable biomarker and therapeutic target, providing new opportunities for precision medicine in CRS treatment. miR-941 may exert its effects by modulating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Fu Shu
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400000, People’s Republic of China
| | - Yaping Wang
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing, 400000, People’s Republic of China
| | - Feng Zhang
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing, 400000, People’s Republic of China
| | - Linglong Li
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing, 400000, People’s Republic of China
| | - Dehong Mao
- Department of Otorhinolaryngology, Yongchuan Chinese Medicine Hospital Affiliated to Chongqing Medical University, Chongqing, 400000, People’s Republic of China
| |
Collapse
|
5
|
Wu A, Wang Y, Mao R, Tan Z, Xu S, Long J, Wang Q, Zhao Z, Xie H, Deng Z, Li J, Chen M. Naturally-occurring carnosic acid as a promising therapeutic agent for skin inflammation via targeting STAT1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156442. [PMID: 39919329 DOI: 10.1016/j.phymed.2025.156442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/30/2024] [Accepted: 01/28/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Psoriasis and rosacea are prevalent chronic inflammatory skin disorders driven by aberrant interactions between skin-resident keratinocytes and immune cells. Natural products represent a largely untapped source of novel therapeutic agents for various diseases. This study aimed to identify an effective natural product for treating psoriasis and rosacea and to elucidate its underlying mechanism of action. METHODS Bioinformatics and network pharmacology approaches were employed to identify potential drug candidates for these conditions. Psoriasis-like and rosacea-like inflammation models were established in mice to assess the in vivo therapeutic effects of carnosic acid. In vitro experiments were performed to investigate the molecular mechanisms underlying carnosic acid's anti-inflammatory activity. RESULTS Through bioinformatics and network pharmacology, carnosic acid, a plant-derived phenolic diterpene, was identified as a promising candidate for these skin disorders. Functional assays demonstrated that carnosic acid effectively inhibited skin inflammation in both imiquimod-induced psoriasis and LL37-induced rosacea mouse models. Mechanistically, carnosic acid bound directly to STAT1, inhibiting its phosphorylation and subsequent transcriptional activation, which led to a reduction in the production of STAT1-mediated inflammatory factors in keratinocytes. Topical application of carnosic acid significantly alleviated clinical symptoms in both psoriasis and rosacea models. CONCLUSION These findings suggest that carnosic acid holds potential as a therapeutic agent for STAT1-mediated skin inflammation.
Collapse
Affiliation(s)
- Aike Wu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yunying Wang
- Department of Dermatology, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Rui Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zixin Tan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - San Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Juan Long
- Department of Dermatology, Hunan Children's Hospital, Changsha, China
| | - Qian Wang
- Hunan Binsis Biotechnology Co., Ltd, Changsha, China
| | - Zhixiang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
6
|
Qiu Y, Jiang W, Feng D, Yu Y, Hou H, Deng M, Chen X, Liu L, Wu R, Lu Q, Zhao M. Resolving 3-Dimensional Genomic Landscape of CD4+ T Cells in the Peripheral Blood of Patients with Psoriasis. J Invest Dermatol 2025; 145:831-841.e10. [PMID: 39182560 DOI: 10.1016/j.jid.2024.08.006] [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: 05/22/2024] [Revised: 07/20/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024]
Abstract
A precise regulation of gene expression depends on the accuracy of the 3-dimensional (3D) structure of chromatin; however, the effects of the 3D genome on gene expression in psoriasis remain unknown. In this study, we conducted Hi-C and RNA sequencing on CD4+ T cells collected from 5 patients with psoriasis and 3 healthy controls and constructed a comprehensive 3D chromatin interaction map to delineate the genomic hierarchies, including A/B compartments, topologically associated domains, and chromatin loops. Then, the specific superenhancers related to psoriasis were identified by Hi-C and H3K27ac chromatin immunoprecipitation sequencing data. Subsequently, comprehensive analyses were carried out on the differentially expressed genes that are associated with altered topologically associated domains, loops, and superenhancers in psoriasis. Finally, we screened the candidate target genes and examined the potential functional SNP in psoriasis affected by disruptions of the spatial organization. This study provides a comprehensive reference for examining the 3D genome interactions in psoriasis and elucidating the interplay between spatial organization disruption and gene regulation. We hope that our findings can help clarify the mechanisms underlying the pathogenesis of psoriasis and shed light on the role of 3D genomic structure, therefore informing potential therapeutic approaches.
Collapse
Affiliation(s)
- Yueqi Qiu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wenjuan Jiang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China
| | - Delong Feng
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yaqin Yu
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Huihui Hou
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; School of Public Health, Nanjing Medical University, Nanjing, China
| | - Min Deng
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyun Chen
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Liu
- Epigenetic Group, Frasergen Bioinformatics, Wuhan, China
| | - Ruifang Wu
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Ming Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, China; Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China; School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
7
|
Zhang Y, Zang C, Mao M, Zhang M, Tang Z, Chen W, Zhu W. Advances in RNA therapy for the treatment of autoimmune diseases. Autoimmun Rev 2025; 24:103753. [PMID: 39842534 DOI: 10.1016/j.autrev.2025.103753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Autoimmune diseases (ADs) are a group of complex, chronic conditions characterized by disturbance of immune tolerance, with examples including systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, and psoriasis. These diseases have unclear pathogenesis, and traditional therapeutic approaches remain limited. However, advances in high-throughput histology technology and scientific discoveries have led to the identification of various pathogenic factors contributing to ADs. Coupled with improvements in RNA nucleic acid-based drug synthesis, design, and delivery, RNA-based therapies have been extensively investigated for their potential in treating ADs. This paper reviews the progress in the use of miRNAs, lncRNAs, circRNAs, siRNAs, antisense oligonucleotides (ASOs), aptamers, mRNAs, and other RNA-based therapies in ADs, focusing on their therapeutic potential and application prospects, providing insights for future research and clinical treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Ying Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Chenyang Zang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Manyun Mao
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Mi Zhang
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Zhenwei Tang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wangqing Chen
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
| | - Wu Zhu
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Changsha, Hunan, China; Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
| |
Collapse
|
8
|
Tian M, Hao F, Jin X, Wang X, Chang T, He S, Wang H, Jiang Y, Wang Y, Liu J, Feng Y, Li D, Yin Z, Ba X, Wei M. KLHL25-ACLY module functions as a switch in the fate determination of the differentiation of iTreg/Th17. Commun Biol 2025; 8:471. [PMID: 40119138 PMCID: PMC11928475 DOI: 10.1038/s42003-025-07917-z] [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: 08/02/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
The differentiation of Th17 and iTreg is tightly associated with fatty acid metabolism. TGFβ1-induced iTreg differentiation from Th0 relies on fatty acid oxidation (FAO), whereas IL-6 with TGFβ1 shifts metabolism to Th17-preferred fatty acid synthesis (FAS). However, how IL-6 reprograms fatty acid metabolism remains unclear. Here, we unveiled that TGFβ1-activated JNK is recruited to the Klhl25 promoter by NF-YA. JNK then phosphorylates histone H3 at Ser10 to activate Klhl25 transcription, leading to the ubiquitination-dependent degradation of ATP-citrate lyase (ACLY) and the switch from FAS to FAO, which supports iTreg generation. Whereas, upon IL-6 signaling, NF-YA is phosphorylated by ERK, losing its DNA binding ability, which shuts off TGFβ1-JNK-mediated Klhl25 transcription and ACLY ubiquitination, thereby increasing FAS and supporting Th17 differentiation. This study demonstrated that KLHL25-ACLY module functions as a switch in response to TGFβ1 and IL-6 signals, playing a decisive role in the fate determination of iTreg/Th17 differentiation.
Collapse
Affiliation(s)
- Miaomiao Tian
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Fengqi Hao
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
- School of Physical Education, Northeast Normal University, Changchun, Jilin, China
| | - Xin Jin
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Xinyu Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Tianyi Chang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Shuang He
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Huiyue Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Ying Jiang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Yang Wang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Jia Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Yunpeng Feng
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China
| | - Dan Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Zhuhai, China
| | - Xueqing Ba
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China.
| | - Min Wei
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), Northeast Normal University, Changchun, Jilin, China.
| |
Collapse
|
9
|
Shan Z, Zhou L, Ma Y, Huang Y. Circ_0090100 induces AHNAK expression to inhibit trophoblast cell proliferation and invasion and accelerate cell apoptosis by segregating miR-139-5p in preeclampsia. Hum Cell 2025; 38:67. [PMID: 40063163 DOI: 10.1007/s13577-025-01185-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 02/06/2025] [Indexed: 05/09/2025]
Abstract
The etiology of preeclampsia (PE) is complex and is known to involve the expression of circular RNAs (circRNAs). Among these, the function of circ_0090100 in PE is yet to be fully understood. This study was conducted to examine the expression profile of circ_0090100 in placental tissues from PE patients and to assess its influence on trophoblast cell functions. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to measure the RNA expression levels of circ_0090100, microRNA-139-5p (miR-139-5p), and AHNAK nucleoprotein (AHNAK). Cell viability, proliferation, apoptosis, and invasion were assessed using the CCK-8 assay, EdU incorporation, flow cytometry, and transwell migration assays, respectively. Western blot analysis was performed to detect the protein expression of N-cadherin, Vimentin, and AHNAK. Dualluciferase reporter assay and RNA immunoprecipitation (RIP) assay were conducted to elucidate the relationships between circ_0090100, miR-139-5p, and AHNAK. We found that the expression of circ_0090100 and AHNAK was upregulated, while miR-139-5p. expression was downregulated in PE placental tissues compared to controls. Transfection of a plasmid overexpressing circ_0090100 into JEG-3 and HTR-8/SVneo trophoblast cell lines resulted in reduced cell proliferation and invasion, but increased apoptosis. Mechanistically, circ_0090100 functioned as a miR-139-5p sponge, and miR-139-5p targeted AHNAK. Furthermore, upregulating miR-139-5p reversed the effects of circ_0090100 in JEG-3 and HTR-8/SVneo cells. AHNAK was found to be involved in the regulation of miR-139-5p effects on trophoblast cells. Additionally, circ_0090100 induced the production of AHNAK through miR-139-5p. Therefore, circ_0090100 activated AHNAK to suppress trophoblast cell proliferation and invasion, and promote cell apoptosis, via miR-139-5p.
Collapse
Affiliation(s)
- Zhenli Shan
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Jing'an District, No. 580 Changle Road, Shanghai, 200092, China
| | - Linfang Zhou
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Jing'an District, No. 580 Changle Road, Shanghai, 200092, China
| | - Yan Ma
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Jing'an District, No. 580 Changle Road, Shanghai, 200092, China
| | - Yiying Huang
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Jing'an District, No. 580 Changle Road, Shanghai, 200092, China.
| |
Collapse
|
10
|
Mohammed OA, Alghamdi M, Bahashwan E, Al Jarallah AlQahtani A, Alfaifi A, Hassan RH, Alfaifi J, Alamri MMS, Alhalafi AH, Adam MIE, BinAfif WF, Abdel-Reheim MA, Mageed SSA, S Doghish A. Emerging insights into the role of natural products and miRNAs in psoriasis: from pathophysiology to precision medicine. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2487-2509. [PMID: 39466441 DOI: 10.1007/s00210-024-03528-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024]
Abstract
Psoriasis is a sustainable skin disease characterized by inflammation resulting from the interaction between immune cells and keratinocytes. Significant advancements have been achieved in studying the molecular process behind noncoding and coding genes, leading to valuable insights for clinical therapy. Nevertheless, our comprehension of this intricate ailment remains ambiguous. Natural products such as curcumin, vitamin D, omega-3, vitamin E, psoralen, gallic acid (GA), and resveratrol offer a promising alternative or adjunct therapy for psoriasis by modulating multiple pathways and exhibiting fewer side effects compared to conventional treatments. MicroRNAs (miRNAs) are short RNAs that are involved in regulating gene expression after transcription, namely by suppressing gene activity. Recent research on miRNAs has uncovered their significant significance in the development of psoriasis. In this review, we examined the latest developments in the investigation of miRNAs in psoriasis. Previous studies have revealed that imbalanced miRNAs in psoriasis have a significant impact on the processes of keratinocyte differentiation, proliferation, and the progression of inflammation. Furthermore, miRNAs exert an impact on the activity of immune cells involved in psoriasis, such as Langerhans cells, dendritic cells, and CD4+ T cells. Furthermore, we explore potential miRNA-focused treatment options for psoriasis, including the localized administration of external miRNA mimics, and miRNA inhibitors. The effectiveness of natural products and miRNAs in treating psoriasis, as well as the signaling pathways that may be involved, are summarized in this article.
Collapse
Affiliation(s)
- Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Mushabab Alghamdi
- Department of Internal Medicine, Division of Rheumatology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Emad Bahashwan
- Department of Internal Medicine, Division of Dermatology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - AbdulElah Al Jarallah AlQahtani
- Department of Internal Medicine, Division of Dermatology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Adel Alfaifi
- Department of Dermatology, Armed Forces Hospital - Southern Region, 62413, Khamis Mushait, Saudi Arabia
| | - Rania H Hassan
- Dermatology Clinic, Abbasseya Psychiatric Hospital, Abbasseya, Cairo, 11517, Egypt
| | - Jaber Alfaifi
- Department of Child Health, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Mohannad Mohammad S Alamri
- Department of Family and Community Medicine, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Abdullah Hassan Alhalafi
- Department of Family and Community Medicine, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Masoud I E Adam
- Department of Medical Education and Internal Medicine, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Waad Fuad BinAfif
- Department of Internal Medicine, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | | | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Badr City, 11829, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Badr City, , 11829, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Nasr City, 11231, Egypt.
| |
Collapse
|
11
|
Bolha L, Hočevar A, Jurčić V. Current state of epigenetics in giant cell arteritis: Focus on microRNA dysregulation. Autoimmun Rev 2025; 24:103739. [PMID: 39732382 DOI: 10.1016/j.autrev.2024.103739] [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: 09/18/2024] [Revised: 12/23/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
Giant cell arteritis (GCA) is a primary systemic vasculitis affecting the elderly, characterized by a granulomatous vessel wall inflammation of large- and medium-sized arteries. The immunopathology of GCA is complex, involving both the innate and adaptive arms of the immune system, where a maladaptive inflammatory-driven vascular repair process ultimately results in vessel wall thickening, intramural vascular smooth muscle cell proliferation, neovascularization and vessel lumen occlusion, which can lead to serious ischemic complications such as visual loss and ischemic stroke. Over the past decade, microRNA (miRNA) dysregulation has been highlighted as an important contributing factor underlying the pathogenesis of GCA. Since current understanding of miRNA involvement in GCA remains largely based on extrapolation of previously determined miRNA functions in vitro or in loss- or gain-of-function studies, an overall insight into the role of miRNA alteration in GCA pathophysiology remains limited. In this narrative review, we summarize the current knowledge on aberrantly expressed miRNAs in GCA and thoroughly discuss the impact of their altered regulatory role in the context of GCA setting. Furthermore, we address challenges and future perspectives in utilization of miRNA-based diagnostic and prognostic biomarkers of GCA in clinical settings.
Collapse
Affiliation(s)
- Luka Bolha
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| | - Alojzija Hočevar
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vesna Jurčić
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
12
|
Teo LTK, Juantuah-Kusi N, Subramanian G, Sampath P. Psoriasis Treatments: Emerging Roles and Future Prospects of MicroRNAs. Noncoding RNA 2025; 11:16. [PMID: 39997616 PMCID: PMC11858470 DOI: 10.3390/ncrna11010016] [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: 12/19/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Psoriasis, a widespread and chronic inflammatory skin disorder, is marked by its persistence and the lack of a definitive cure. The pathogenesis of psoriasis is increasingly understood, with ongoing research highlighting the intricate interplay of genetic, immunological, and environmental factors. Recent advancements have illuminated the pivotal role of microRNAs in orchestrating complex processes in psoriasis and other hyperproliferative skin diseases. This narrative review highlights the emerging significance of miRNAs as key regulators in psoriasis pathogenesis and examines their potential as therapeutic targets. We discuss current treatment approaches and the promising future of miRNAs as next-generation therapeutic agents for this condition.
Collapse
Affiliation(s)
- Li Tian Keane Teo
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, South Kensington, London SW7 2AZ, UK
| | - Nerissa Juantuah-Kusi
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore
| | - Gowtham Subramanian
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore
- Skin Research Institute of Singapore (SRIS), 11 Mandalay Road #17-01 Clinical Sciences Building, Singapore 308232, Singapore
| | - Prabha Sampath
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Singapore
- Skin Research Institute of Singapore (SRIS), 11 Mandalay Road #17-01 Clinical Sciences Building, Singapore 308232, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
13
|
Xin L, Xu HS, Fan LJ, Liu C, Zou YH, Zhou Q, Yue ZQ, Gan JH, Liu J. Methionine Restriction Exerts Anti-Tumor Immunity via Joint Intervention of T-Bet Palmitoylation in Gastric Cancer. Biotechnol J 2025; 20:e202400574. [PMID: 39989253 DOI: 10.1002/biot.202400574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Methionine restriction (MR) exerts an anti-tumor immunomodulatory role. Th1 cells facilitate CD8+ cytotoxic T cell activation and targeted tumor cell killing. Our previous work shows that MR enhances the immunotherapy effect of PD-L1/PD-1 blockade on gastric cancer, MR can simultaneously inhibit Th1 cell differentiation, which may affect their synergistic therapeutic outcome. We aim to elucidate the molecular mechanism of MR regulating Th1 cell activation in gastric cancer. METHODS Murine Foregastric Carcinoma (MFC) cells were injected into 615 mice to establish transplanted tumor models, which were then treated with an MR diet or combined with 2-bromopalmitate (2-BP). CD4+T cells were cultured with deficient methionine. The acyl-biotinyl exchange (ABE) method was to detect T-bet palmitoylation and cycloheximide experiments to detect protein stability. GPS-Palm tool was employed to screen palmitoyltransferases. The impact of T-bet palmitoylation on the pro-tumor-killing effect of Th1 cells was examined. RESULTS MR enhanced anti-PD-1's inhibition of tumor growth, while concurrently suppressing the increased Th1 cells. Combined with 2-BP further inhibited tumor and increased Th1 cells. Suppressing Th1 activity attenuated 2-BP's synergistic therapeutic effect and reduced CD8+ GZMB+ T cells. MR inhibited Th1 differentiation by reducing T-bet expression, 2-BP treatment restored, while T-bet interference reversed 2-BP's effect. MR increased palmitoylation and T-bet underwent palmitoylation modification. ZDHHC23 mediated T-bet palmitoylation and promoted T-bet degradation. MR promoted T-bet degradation, thereby decreasing T-bet content, inhibiting Th1 cell polarization and CD8+ T cell killing effect. CONCLUSIONS MR combined with T-bet palmitoylation intervention promotes Th1 polarization and CD8+ T cell toxicity, thereby enhancing anti-tumor immunity in gastric cancer.
Collapse
Affiliation(s)
- Lin Xin
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - He-Song Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Luo-Jun Fan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Chuan Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yong-Hui Zou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Qi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhen-Qi Yue
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jin-Heng Gan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jiang Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
14
|
Zhu Y, Zhao L, Yan W, Ma H, Zhao W, Qu J, Zheng W, Zhang C, Du H, Yu M, Wan N, Ye H, Xie Y, Ke B, Xu Q, Sun H, Sun Y, Ouyang Z. Celastrol directly targets LRP1 to inhibit fibroblast-macrophage crosstalk and ameliorates psoriasis progression. Acta Pharm Sin B 2025; 15:876-891. [PMID: 40177548 PMCID: PMC11959968 DOI: 10.1016/j.apsb.2024.12.041] [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: 07/29/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 04/05/2025] Open
Abstract
Psoriasis is an incurable chronic inflammatory disease that requires new interventions. Here, we found that fibroblasts exacerbate psoriasis progression by promoting macrophage recruitment via CCL2 secretion by single-cell multi-omics analysis. The natural small molecule celastrol was screened to interfere with the secretion of CCL2 by fibroblasts and improve the psoriasis-like symptoms in both murine and cynomolgus monkey models. Mechanistically, celastrol directly bound to the low-density lipoprotein receptor-related protein 1 (LRP1) β-chain and abolished its binding to the transcription factor c-Jun in the nucleus, which in turn inhibited CCL2 production by skin fibroblasts, blocked fibroblast-macrophage crosstalk, and ameliorated psoriasis progression. Notably, fibroblast-specific LRP1 knockout mice exhibited a significant reduction in psoriasis like inflammation. Taken together, from clinical samples and combined with various mouse models, we revealed the pathogenesis of psoriasis from the perspective of fibroblast-macrophage crosstalk, and provided a foundation for LRP1 as a novel potential target for psoriasis treatment.
Collapse
Affiliation(s)
- Yuyu Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Lixin Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Wei Yan
- Department of Dermatology and Venereology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyue Ma
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wanjun Zhao
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Wei Zheng
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Chenyang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Haojie Du
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Meng Yu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ning Wan
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Ye
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yicheng Xie
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Bowen Ke
- Department of Anesthesiology, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
| | - Haiyan Sun
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing 210008, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Zijun Ouyang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen 518055, China
| |
Collapse
|
15
|
Pan J, Chen S, Chen X, Song Y, Cheng H. Histone Modifications and DNA Methylation in Psoriasis: A Cellular Perspective. Clin Rev Allergy Immunol 2025; 68:6. [PMID: 39871086 DOI: 10.1007/s12016-024-09014-1] [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] [Accepted: 12/08/2024] [Indexed: 01/29/2025]
Abstract
In recent years, epigenetic modifications have attracted significant attention due to their unique regulatory mechanisms and profound biological implications. Acting as a bridge between environmental stimuli and changes in gene activity, they reshape gene expression patterns, providing organisms with regulatory mechanisms to respond to environmental changes. A growing body of evidence indicates that epigenetic regulation plays a crucial role in the pathogenesis and progression of psoriasis. A deeper understanding of these epigenetic mechanisms not only helps unveil the molecular mechanisms underlying the initiation and progression of psoriasis but may also provide new insights into diagnostic and therapeutic strategies. Given the unique roles and significant contributions of various cell types involved in the process of psoriasis, a thorough analysis of specific epigenetic patterns in different cell types becomes a key entry point for elucidating the mechanisms of disease development. Although epigenetic modifications encompass multiple complex layers, this review will focus on histone modifications and DNA methylation, describing how they function in different cell types and subsequently impact the pathophysiological processes of psoriasis. Finally, we will summarize the current problems in research concerning histone modifications and DNA methylation in psoriasis and discuss the clinical application prospects and challenges of targeting epigenetic modifications as therapeutic strategies for psoriasis.
Collapse
Affiliation(s)
- Jing Pan
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Siji Chen
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xianzhen Chen
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yinjing Song
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Hao Cheng
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
16
|
Li D, Li X, Duan M, Xue X, Tang X, Nan N, Zhao R, Zhang W, Zhang W. Knockdown of PELI1 promotes Th2 and Treg cell differentiation in juvenile idiopathic arthritis. Exp Cell Res 2025; 444:114360. [PMID: 39617092 DOI: 10.1016/j.yexcr.2024.114360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
Pellino1 (PELI1) is a key regulator of inflammatory and autoimmune diseases. The role of PELI1 in juvenile idiopathic arthritis (JIA) is unclear. The correlation between serum PELI1 mRNA levels and clinical indicators of JIA patients was evaluated by Pearson correlation analysis. The percentage of Th1, Th2, Th17 and Treg cells was analyzed by flow cytometry. ELISA kits were used to detect cytokine levels in serum and cell supernatants. Co-immunoprecipitation experiments were performed to validate PELI1 and TCF-1 interactions. The protein and ubiquitination levels of TCF-1 were detected by western blot. The results showed that JIA patients have high serum PELI1 levels. PELI1 levels were positively correlated with erythrocyte sedimentation rate, C-reactive protein levels and JADAS27 scores in JIA patients. Interfering with PELI1 promoted naïve CD4+ T cell differentiation to Th2 and Treg cells and increased IL-4 and IL-10 levels, while inhibiting their differentiation to Th1 and Th17 cells and decreasing IFN-γ and IL-17 levels. PELI1 increased TCF-1 ubiquitination levels and accelerated its degradation. Inhibition of TCF-1 reduced the effects of interfering with PELI1 on cell differentiation and cytokine levels. In conclusion, Silencing of PELI1 facilitated the naïve CD4+ T cell differentiation into Th2 and Treg cells by TCF-1.
Collapse
Affiliation(s)
- Dan Li
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China; Department of Rheumatology and Immunology, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Xiaoqing Li
- Department of Rheumatology and Immunology, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Mingyue Duan
- Department of Clinical Laboratory, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Xiuhong Xue
- Department of Rheumatology and Immunology, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Xianyan Tang
- Department of Rheumatology and Immunology, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Nan Nan
- Department of Rheumatology and Immunology, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Rui Zhao
- Department of Rheumatology and Immunology, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Wenhua Zhang
- Department of Rheumatology and Immunology, Xi'an Children's Hospital, Xi'an, Shaanxi, 710003, China
| | - Wanggang Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China.
| |
Collapse
|
17
|
Liang Y, Wang Y, Peng A, Li J, Zhang K. Molecular mechanisms and drug therapy of metabolism disorders in psoriasis. J DERMATOL TREAT 2024; 35:2375580. [PMID: 39013549 DOI: 10.1080/09546634.2024.2375580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/26/2024] [Indexed: 07/18/2024]
Abstract
Psoriasis is a prevalent skin disease affecting approximately 1%-3% of the population and imposes significant medical, social and economic burdens. Psoriasis involves multiple organs and is often complicated with obesity, diabetes, dyslipidemia, and hypertension. Because of the benefits of lipid-lowering agents and antidiabetic medications for psoriasis, metabolic abnormalities possibly play a pathogenic role in psoriasis. This review focuses on the impacts of a variety of metabolic disorders on psoriasis and the underlying mechanisms. In psoriasis, enhanced glycolysis, glutamine metabolism and altered fatty acid composition in the psoriatic lesion and plasma result in the excessive proliferation of keratinocytes and secretion of inflammatory cytokines. Altered metabolism is associated with the activation of MTORC signaling pathway and transcription factors such as HIF and S6K1. Therefore, MTORC1 can be a target for the treatment of psoriasis. Additionally, there are diabetes drugs and lipid-lowering drugs including TZDs, GLP-1 RAs, Metformin, statins and fibrates, which improve both metabolic levels and psoriasis symptoms.
Collapse
Affiliation(s)
- Yanyang Liang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Ying Wang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Aihong Peng
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Junqin Li
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
18
|
Sun J, Yuan H, Yu Y, Li A, Zhao Z, Tang Y, Zheng F. Immunomodulatory potential of primary cilia in the skin. Front Immunol 2024; 15:1456875. [PMID: 39676858 PMCID: PMC11638010 DOI: 10.3389/fimmu.2024.1456875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Primary cilia (PC) are essential signaling hubs for proper epithelial formation and the maintenance of skin homeostasis. Found on most cells in the human body, including skin cells, PC facilitate signal transduction that allows ciliated cells to interact with the immune system via multiple pathways, helping to maintain immune system homeostasis. PC can be altered by various microenvironmental stimuli to develop corresponding regulatory functions. Both PC and ciliary signaling pathways have been shown to be involved in the immune processes of various skin lesions. However, the mechanisms by which PC regulate cellular functions and maintain immune homeostasis in tissues are highly complex, and our understanding of them in the skin remains limited. In this paper, we discuss key ciliary signaling pathways and ciliated cells in the skin, with a focus on their immunomodulatory functions. We have compiled evidence from various cells, tissues and disease models to help explore the potential immunomodulatory effects of PC in the skin and their molecular mechanisms.
Collapse
Affiliation(s)
- Jingwei Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huimin Yuan
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yanru Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Aorou Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zihe Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fengjie Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
19
|
Jin L, Jiang Q, Huang H, Zhou X. Topical histone deacetylase inhibitor remetinostat improves IMQ-induced psoriatic dermatitis via suppressing dendritic cell maturation and keratinocyte differentiation and inflammation. Eur J Pharmacol 2024; 983:177011. [PMID: 39304110 DOI: 10.1016/j.ejphar.2024.177011] [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: 06/05/2024] [Revised: 09/07/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by excessive proliferation of keratinocytes and infiltration of immune cells. Although psoriasis has entered the era of biological treatment, there is still a need to explore more effective therapeutic targets and drugs due to the presence of resistance and adverse reactions to biologics. Remetinostat, an HDAC inhibitor, can maintain its potency within the skin with minimal systemic effects, making it a promising topical medication for treating psoriasis. But its effectiveness in treating psoriasis has not been evaluated. In this study, the topical application of remetinostat significantly improved psoriasiform inflammation in an imiquimod-induced mice model by inhibiting CD86 expression of CD11C+I-A/I-E+ dendritic cells (DCs) in the skin. Moreover, remetinostat could dampen the maturation and activation of bone marrow-derived DCs in vitro, as well as the expression of psoriasis-related inflammatory mediators by keratinocytes. In addition, remetinostat could promote keratinocyte differentiation without affecting its proliferation. Our findings demonstrate that remetinostat improves psoriasis by inhibiting the maturation and activation of DCs and the differentiation and inflammation of keratinocytes, which may facilitate the potential application of remetinostat in anti-psoriasis therapy.
Collapse
Affiliation(s)
- Liping Jin
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qian Jiang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Huining Huang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xingchen Zhou
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
20
|
Mu S, Wang W, Liu Q, Ke N, Li H, Sun F, Zhang J, Zhu Z. Autoimmune disease: a view of epigenetics and therapeutic targeting. Front Immunol 2024; 15:1482728. [PMID: 39606248 PMCID: PMC11599216 DOI: 10.3389/fimmu.2024.1482728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Autoimmune diseases comprise a large group of conditions characterized by a complex pathogenesis and significant heterogeneity in their clinical manifestations. Advances in sequencing technology have revealed that in addition to genetic susceptibility, various epigenetic mechanisms including DNA methylation and histone modification play critical roles in disease development. The emerging field of epigenetics has provided new perspectives on the pathogenesis and development of autoimmune diseases. Aberrant epigenetic modifications can be used as biomarkers for disease diagnosis and prognosis. Exploration of human epigenetic profiles revealed that patients with autoimmune diseases exhibit markedly altered DNA methylation profiles compared with healthy individuals. Targeted cutting-edge epigenetic therapies are emerging. For example, DNA methylation inhibitors can rectify methylation dysregulation and relieve patients. Histone deacetylase inhibitors such as vorinostat can affect chromatin accessibility and further regulate gene expression, and have been used in treating hematological malignancies. Epigenetic therapies have opened new avenues for the precise treatment of autoimmune diseases and offer new opportunities for improved therapeutic outcomes. Our review can aid in comprehensively elucidation of the mechanisms of autoimmune diseases and development of new targeted therapies that ultimately benefit patients with these conditions.
Collapse
Affiliation(s)
- Siqi Mu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Wanrong Wang
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Qiuyu Liu
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Naiyu Ke
- Department of Ophthalmology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hao Li
- Department of Urology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Feiyang Sun
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Jiali Zhang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
| | - Zhengwei Zhu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
| |
Collapse
|
21
|
Chen L, Liu C, Xiang X, Qiu W, Guo K. miR‑155 promotes an inflammatory response in HaCaT cells via the IRF2BP2/KLF2/NF‑κB pathway in psoriasis. Int J Mol Med 2024; 54:91. [PMID: 39219281 PMCID: PMC11374146 DOI: 10.3892/ijmm.2024.5415] [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: 02/01/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Psoriasis is a chronic inflammatory skin condition with numerous causes, including genetic, immunological and infectious factors. The course of psoriasis is long and recurrence is common; pathogenesis is not completely understood. However, there is an association between advancement of psoriasis and aberrant microRNA (miR or miRNA)‑155 expression. Through bioinformatics, the present study aimed to analyze the differentially expressed genes and miRNAs in psoriasis and its biological mechanism and function psoriatic inflammation. First of all, differentially expressed genes (DEGs) and miRNAs (DEMs) in patients with psoriasis were identified using GEO2R interactive web application. A psoriasis inflammatory model was established using lipopolysaccharide (LPS)‑treated HaCaT keratinocytes, which were transfected with miR‑155 mimic or inhibitor. Cell Counting Kit‑8 was used for the assessment of cell viability and proliferation, and changes in the cell cycle were examined using flow cytometry. ELISA and reverse transcription‑quantitative PCR (RT‑qPCR) were used to detect the expression levels of the inflammatory factors IL‑1β and IL‑6. The dual‑luciferase reporter assay was used to verify the targeting association between miR‑155‑5p and IFN regulatory factor 2 binding protein 2 (IRF2BP2). To verify the targeting association of miR‑155 and the IRF2BP2/kruppel‑like factor 2 (KLF2)/NF‑κB signaling pathway, expression levels of IRF2BP2, KLF2 and p65 were identified by RT‑qPCR and western blotting. IRF2BP2 levels were also confirmed by immunofluorescence, in conjunction with bioinformatics database analysis. Overexpression of miR‑155 inhibited proliferation of HaCaT cells and increased the number of cells in S phase and decreasing number of cells in G1 and G2 phase. In the LPS‑induced inflammatory state, miR‑155 overexpression heightened the inflammatory response of HaCaT cells while inhibition of miR‑155 lessened it. Suppression of inflammatory cytokine expression by miR‑155‑5p inhibitor was reversed by knockdown of IRF2BP2. miR‑155 was shown to interact with IRF2BP2 to negatively regulate its expression, leading to decreased KLF2 expression and increased p65 expression and secretion of inflammatory factors, intensifying the inflammatory response of HaCaT cells. Therefore, miR‑155 may contribute to development of psoriasis by inducing tissue and cell damage by increasing the inflammatory response of HaCaT cells via the IRF2BP2/KLF2/NF‑κB pathway. In conclusion, the results of the present study offer novel perspectives on the role of miR‑155 in the onset and progression of psoriasis.
Collapse
Affiliation(s)
- Lu Chen
- Department of Immunology, School of Medicine, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Chang Liu
- Department of Immunology, School of Medicine, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Xuesong Xiang
- Department of Immunology, School of Medicine, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Wenhong Qiu
- Department of Immunology, School of Medicine, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Kaiwen Guo
- Department of Pathogenic Biology, Medical College, Wuhan University of Science and Technology, Wuhan, Hubei 430065, P.R. China
| |
Collapse
|
22
|
Wang R, Huang Y, Shao K, Yan J, Sun Q. High Expression of miR-6785-5p in the Serum Exosomes of Psoriasis Patients Alleviates Psoriasis-Like Skin Damage by Interfering with the MNK2/p-eIF4E Axis in Keratinocytes. Inflammation 2024; 47:1585-1599. [PMID: 38472599 DOI: 10.1007/s10753-024-01995-7] [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: 12/02/2023] [Revised: 01/29/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024]
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by abnormal keratinocyte proliferation and inflammation. MiRNAs and serum exosomes participate in the pathogenesis of many diseases. The objective of this study is to explore the function of miR-6785-5p in psoriatic keratinocytes and its upstream and downstream mechanisms. For our study, we employed qRT-PCR and fluorescence in situ hybridization to evaluate miR-6785-5p in psoriatic keratinocytes and conducted a microRNA microarray for identifying differentially expressed miRNAs in patient serum exosomes. We then cocultured keratinocytes with these exosomes, using immunofluorescence staining and qRT-PCR to assess uptake and miR-6785-5p overexpression. We explored miR-6785-5p's role through transfection with specific mimics and inhibitors and confirmed MNK2 as its target using a luciferase assay. MNK2's function was further examined using siRNA technology. Lastly, we applied an imiquimod-induced psoriasis mouse model, also employing siRNA, to investigate MNK2's role in psoriasis. MiR-6785-5p demonstrates a notable overexpression in the keratinocytes of psoriasis patients as well as in their serum exosomes. These keratinocytes actively uptake the miR-6785-5p-enriched serum exosomes. Functionally, miR-6785-5p appears to alleviate psoriasis-like skin damage, observable both in vitro and in vivo, by downregulating MNK2 expression. Psoriasis keratinocytes uptake serum exosomes highly expressing miR-6785-5p. MiR-6785-5p inhibits the abnormal proliferation and inflammatory state of keratinocytes by reducing MNK2 expression and interfering with the MNK2/p-eIF4E axis.
Collapse
Affiliation(s)
- Ruijie Wang
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
- Laboratory of Basic Medical Science, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Yingjian Huang
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
- Laboratory of Basic Medical Science, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Kaixin Shao
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
- Laboratory of Basic Medical Science, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Jianjun Yan
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China.
| | - Qing Sun
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
23
|
Chen X, Fang M, Hong J, Guo Y. JNK Pathway-Associated Phosphatase Deficiency Facilitates Atherosclerotic Progression by Inducing T-Helper 1 and 17 Polarization and Inflammation in an ERK- and NF-κB Pathway-Dependent Manner. J Atheroscler Thromb 2024; 31:1460-1478. [PMID: 38797677 PMCID: PMC11456371 DOI: 10.5551/jat.64754] [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: 05/29/2024] Open
Abstract
AIM JNK pathway-associated phosphatase (JKAP) regulates T cell-mediated immunity and inflammation, which are involved in atherosclerosis pathogenesis. This study investigated the effects of JKAP on T-helper (Th) cell polarization, inflammation, and atherosclerotic progression. METHODS Serum JKAP levels were measured in 30 patients with coronary heart disease (CHD) and 30 controls. CHD blood naïve CD4+ T cells were acquired, followed by JKAP overexpression and knockdown with or without treatment with PD98059 (ERK inhibitor) or BAY-11-7082 (NF-κB inhibitor) in vitro. CD4+ T-cell conditional JKAP ablation mice were established in vivo, followed by the construction of an atherosclerosis model. RESULTS JKAP was reduced and negatively correlated with the Gensini score, CRP, Th1 cells, Th17 cells, and proinflammatory cytokines in patients with CHD. In vitro, JKAP overexpression suppressed Th1 and Th17 cell differentiation and proinflammatory cytokines, whereas JKAP knockdown exerted the opposite effect; however, JKAP modification did not affect Th2 cell differentiation. Interestingly, JKAP negatively regulated the ERK and NF-κB pathways; meanwhile, the PD98059 and BAY-11-7082 treatments repressed Th1 and Th17 cell differentiation, and attenuated the effect of JKAP knockdown on these indices. In vivo, conditional CD4+ T-cell JKAP ablation increased Th1 and Th17 cell polarization in the spleen, lymph node, blood, and/or aortic root. Furthermore, CD4+ T-cell conditional JKAP ablation exaggerated atherosclerotic lesions in the aorta, elevated CD4+ cell infiltration and proinflammatory cytokines in the aortic root, and activated the ERK and NF-κB pathways in the aortic root. CONCLUSION JKAP ablation facilitates atherosclerosis progression by promoting Th1 and 17 polarization and inflammation through regulation of the ERK and NF-κB pathways.
Collapse
Affiliation(s)
- Xinjing Chen
- Department of Cardiology, Provincial Clinical Medical College of Fujian Medical University, Fujian Institute of Cardiovascular Disease, Fujian Provincial Hospital
| | - Mingcheng Fang
- Department of Cardiology, Provincial Clinical Medical College of Fujian Medical University, Fujian Institute of Cardiovascular Disease, Fujian Provincial Hospital
| | - Jingxuan Hong
- Department of Cardiology, Provincial Clinical Medical College of Fujian Medical University, Fujian Institute of Cardiovascular Disease, Fujian Provincial Hospital
| | - Yansong Guo
- Department of Cardiology, Provincial Clinical Medical College of Fujian Medical University, Fujian Institute of Cardiovascular Disease, Fujian Provincial Hospital
| |
Collapse
|
24
|
Chen W, Liang J, He S, Liang Q, Tian W, Lu A, Li D, Huang Z, Wu G. She-Chuang-Si-Wu-Tang Alleviates Inflammation and Itching Symptoms in a Psoriasis Mouse Model by Regulating the Th17/IL-17 Axis via the STAT3/MAPK Pathways. J Inflamm Res 2024; 17:5957-5975. [PMID: 39247836 PMCID: PMC11380483 DOI: 10.2147/jir.s472417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/28/2024] [Indexed: 09/10/2024] Open
Abstract
Purpose Psoriasis is an immune-related disorder characterized by silver scales, epidermis thickness, and itching. She-Chuang-Si-Wu-Tang (SSWT), a traditional Chinese medicine decoction, has been used clinically for 400 years. Although it benefits psoriasis treatment, the mechanism of action is still unclear. This study explores SSWT's molecular mechanism in treating psoriasis through network pharmacology analysis and experiments. Methods We identified relevant SSWT and psoriasis targets using network pharmacology and conducted SSWT quality control with high-performance liquid chromatography (HPLC). A mouse model of psoriasis was established using imiquimod (IMQ), with the drug administered continuously for seven days, spanning an eight-day period. During the experiment, we observed spontaneous scratching behaviors and assessed the Psoriasis Area and Severity Index (PASI) scores. At the conclusion of the experiment, we examined skin tissue pathology under an optical microscope and measured epidermal thickness. Additionally, we used enzyme-linked immunosorbent assay (ELISA) and quantitative reverse transcription polymerase chain reaction (qRT-PCR) to measure interleukin (IL)-23, IL-17A, IL-17F, and interferon (IFN)-γ levels in the mice's serum and their mRNA expression in the skin. Western blot analysis was conducted to assess protein levels related to signaling pathways. Results Results indicate that SSWT may target IL-17 signaling pathways and T helper (Th) 17 cell differentiation, as predicted by network pharmacology. SSWT significantly improved the PASI and Baker scores, reduced epidermal thickness, and decreased spontaneous scratching in IMQ-induced mice. Additionally, SSWT treatment significantly lowered the concentrations of inflammatory factors in the serum and skin lesions, as well as mRNA expression levels, compared to the IMQ group. Furthermore, SSWT significantly inhibited the phosphorylation of both the signal transducer and activator of transcription 3 (STAT3) and mitogen-activated protein kinase (MAPK) pathways. Conclusion In summary, this study unveiled the potential anti-psoriatic mechanism of SSWT, offering new evidence for its clinical application.
Collapse
Affiliation(s)
- Weixiong Chen
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People’s Republic of China
| | - Jianqiang Liang
- Department of Dermatology, the First People’s Hospital of Yulin, Yulin, Guangxi, 537000, People’s Republic of China
| | - Shuang He
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
- Key Laboratory for Complementary and Alternative Medicine Experimental Animal Models of Guangxi, Nanning, Guangxi, 530299, People’s Republic of China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Qingsong Liang
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
- Key Laboratory for Complementary and Alternative Medicine Experimental Animal Models of Guangxi, Nanning, Guangxi, 530299, People’s Republic of China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Wenting Tian
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Aobo Lu
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Demin Li
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Zhicheng Huang
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Guanyi Wu
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
- Key Laboratory for Complementary and Alternative Medicine Experimental Animal Models of Guangxi, Nanning, Guangxi, 530299, People’s Republic of China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| |
Collapse
|
25
|
Wang J, Yan P, Jia Y, Guo Z, Guo Y, Yin R, Wang L, Fan Z, Zhou Y, Yuan J, Yin R. Expression profiles of miRNAs in the lung tissue of piglets infected with Glaesserella parasuis and the roles of ssc-miR-135 and ssc-miR-155-3p in the regulation of inflammation. Comp Immunol Microbiol Infect Dis 2024; 111:102214. [PMID: 39002176 DOI: 10.1016/j.cimid.2024.102214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024]
Abstract
MicroRNAs (miRNAs) have been shown to play an important regulatory role in the process of pathogenic infection. However, the miRNAs that regulate the pathogenic process of G. parasuis and their functions are still unknown. Here, high-throughput sequencing was used to quantify the expression of miRNA in piglet lung tissue after G. parasuis XX0306 strain infection. A total of 25 differentially expressed microRNAs (DEmiRNAs) were identified. GO and KEGG pathway enrichment analysis showed that many of the functions of genes that may be regulated by DEmiRNA are related to inflammatory response and immune regulation. Further studies found that ssc-miR-135 may promote the expression of inflammatory factors through NF-κB signaling pathway. Whereas, ssc-miR-155-3p inhibited the inflammatory response induced by G. parasuis, and its regulatory mechanism remains to be further investigated. This study provides a valuable reference for revealing the regulatory effects of miRNAs on the pathogenesis of G. parasuis. DATA AVAILABILITY: The datasets generated during the current study are not publicly available due to this study is currently in the ongoing research stage, and some of the data cannot be made public sooner yet, but are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Jingyi Wang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China; College of Animal Husbandry and Veterinary Medicine, Jinzhou Medical University, Jinzhou 121000, China.
| | - Ping Yan
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Yongchao Jia
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Zhongbo Guo
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Ying Guo
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Ronglan Yin
- Research Academy of Animal Husbandry and Veterinary Medicine Sciences of Jilin Province, Changchun 130062, China.
| | - Linxi Wang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Zenglei Fan
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Yuanyuan Zhou
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Jing Yuan
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| | - Ronghuan Yin
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China.
| |
Collapse
|
26
|
Tian K, Xu W, Chen M, Deng F. miR-155 promotes Th17 differentiation by targeting FOXP3 to aggravate inflammation in MRSA pneumonia. Cytokine 2024; 180:156662. [PMID: 38824863 DOI: 10.1016/j.cyto.2024.156662] [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: 02/06/2024] [Revised: 05/04/2024] [Accepted: 05/21/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Previous researches have clarified that miR-155 is increased in methicillin-resistant Staphylococcus aureus (MRSA) pneumonia, and modulates Th9 differentiation. Like Th9 cells, Th17 cells were also a subset of CD4+ T cells and involved in MRSA pneumonia progression. This work aimed to investigate the role and mechanism of miR-155 in Th17 differentiation. METHODS Bronchoalveolar lavage fluid (BALF) was collected from children with MRSA pneumonia and bronchial foreign bodies. MRSA-infected murine model was established followed by collecting BALF and lung tissues. qRT-PCR, ELISA and flow cytometry were performed to examine the mRNA expression and concentration of IL-17 and the number of Th17 cells in above samples. HE and ELISA were used to evaluate inflammatory responses in lung. Furthermore, CD4+ T cells were isolated from BALF of children for in vitro experiments. After treatments with miR-155 mimic/inhibitor, the roles of miR-155 in Th17/IL-17 regulation were determined. The downstream of miR-155 was explored by qRT-PCR, western blotting, dual luciferase reporter analysis and RIP assay. RESULTS The levels of IL-17 and the proportion of Th17 cells were increased in children with MRSA pneumonia. A similar pattern was observed in MRSA-infected mice. On the contrary, IL-17 neutralization abolished the activation of Th17/IL-17 induced by MRSA infection. Furthermore, IL-17 blockade diminished the inflammation caused by MRSA. In vitro experiments demonstrated miR-155 positively regulated IL-17 expression and Th17 differentiation. Mechanistically, FOXP3 was a direct target of miR-155. miR-155 inhibited FOXP3 level via binding between FOXP3 and Argonaute 2 (AGO2), the key component of RNA-induced silencing complex (RISC). FOXP3 overexpression reversed elevated IL-17 levels and Th17 differentiation induced by miR-155. CONCLUSIONS miR-155 facilitates Th17 differentiation by reducing FOXP3 through interaction of AGO2 and FOXP3 to promote the pathogenesis of MRSA pneumonia. IL-17 blockade weakens the inflammation due to MRSA, which provides a nonantibiotic treatment strategy for MRSA pneumonia.
Collapse
Affiliation(s)
- Keyin Tian
- Children's Medical Center of Anhui Medical University, Department of Pediatric nephrology, Hefei 230051, Anhui, China; The Fifth Clinical College of Anhui Medical University, Hefei 230051, Anhui, China; Anhui Provincial Children's Hospital, Department of Pediatric emergency, Hefei 230051, Anhui, China
| | - Weihua Xu
- Anhui Provincial Children's Hospital, Department of Pediatric emergency, Hefei 230051, Anhui, China
| | - Mingxiao Chen
- Anhui Provincial Children's Hospital, Department of Pediatric emergency, Hefei 230051, Anhui, China
| | - Fang Deng
- Children's Medical Center of Anhui Medical University, Department of Pediatric nephrology, Hefei 230051, Anhui, China; The Fifth Clinical College of Anhui Medical University, Hefei 230051, Anhui, China; Anhui Provincial Children's Hospital, Department of Pediatric nephrology, Hefei 230051, Anhui, China.
| |
Collapse
|
27
|
Liu R, Zhang L, Zhang K. Histone modification in psoriasis: Molecular mechanisms and potential therapeutic targets. Exp Dermatol 2024; 33:e15151. [PMID: 39090854 DOI: 10.1111/exd.15151] [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: 12/23/2023] [Revised: 06/24/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024]
Abstract
Psoriasis is an immune-mediated, inflammatory disease. Genetic and environmental elements are involved in the nosogenesis of this illness. Epigenetic inheritance serves as the connection between genetic and environmental factors. Histone modification, an epigenetic regulatory mechanism, is implicated in the development of numerous diseases. The basic function of histone modification is to regulate cellular functions by modifying gene expression. Modulation of histone modification, such as regulation of enzymes pertinent to histone modification, can be an alternative approach for treating some diseases, including psoriasis. Herein, we reviewed the regulatory mechanisms and biological effects of histone modifications and their roles in the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Ruifeng Liu
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| | - Luyao Zhang
- Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kaiming Zhang
- Shanxi Key Laboratory of Stem Cells for Immunological Dermatosis, Institute of Dermatology, Taiyuan Central Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
28
|
Toghiani R, Azimian Zavareh V, Najafi H, Mirian M, Azarpira N, Abolmaali SS, Varshosaz J, Tamaddon AM. Hypoxia-preconditioned WJ-MSC spheroid-derived exosomes delivering miR-210 for renal cell restoration in hypoxia-reoxygenation injury. Stem Cell Res Ther 2024; 15:240. [PMID: 39080774 PMCID: PMC11289969 DOI: 10.1186/s13287-024-03845-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Recent advancements in mesenchymal stem cell (MSC) technology have paved the way for innovative treatment options for various diseases. These stem cells play a crucial role in tissue regeneration and repair, releasing local anti-inflammatory and healing signals. However, challenges such as homing issues and tumorigenicity have led to exploring MSC-exosomes as a promising alternative. MSC-exosomes have shown therapeutic potential in conditions like renal ischemia-reperfusion injury, but low production yields hinder their clinical use. METHODS To address this limitation, we examined hypoxic preconditioning of Wharton jelly-derived MSCs (WJ-MSCs) 3D-cultured in spheroids on isolated exosome yields and miR-21 expression. We then evaluated their capacity to load miR-210 into HEK-293 cells and mitigate ROS production, consequently enhancing their survival and migration under hypoxia-reoxygenation conditions. RESULTS MiR-210 overexpression was significantly induced by optimized culture and preconditioning conditions, which also improved the production yield of exosomes from grown MSCs. The exosomes enriched with miR-210 demonstrated a protective effect by improving survival, reducing apoptosis and ROS accumulation in damaged renal cells, and ultimately promoting cell migration. CONCLUSION The present study underscores the possibility of employing advanced techniques to maximize the therapeutic attributes of exosomes produced from WJ-MSC spheroid for improved recovery outcomes in ischemia-reperfusion injuries.
Collapse
Affiliation(s)
- Reyhaneh Toghiani
- Department of Pharmaceutical Nanotechnology, Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vajihe Azimian Zavareh
- Department of Plant and Animal Biology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | - Hanyieh Najafi
- Department of Pharmaceutical Nanotechnology, Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Mirian
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
29
|
Chen L, Wang X, Liu C, Chen X, Li P, Qiu W, Guo K. Integrative analysis of gene and microRNA expression profiles reveals candidate biomarkers and regulatory networks in psoriasis. Medicine (Baltimore) 2024; 103:e39002. [PMID: 39028999 PMCID: PMC11398825 DOI: 10.1097/md.0000000000039002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 06/28/2024] [Indexed: 07/21/2024] Open
Abstract
Psoriasis (PS) is a chronic inflammatory skin disease with a long course and tendency to recur, the pathogenesis of which is not fully understood. This article aims to identify the key differentially expressed genes (DEGs) and microRNA (miRNAs) of PS, construct the core miRNA-mRNA regulatory network, and investigate the underlying molecular mechanism through integrated bioinformatics approaches. Two gene expression profile datasets and 2 miRNA expression profile datasets were downloaded from the gene expression omnibus (GEO) database and analyzed by GEO2R. Intersection DEGs and intersection differentially expressed miRNAs (DEMs) were each screened. The Metascape database and R software were used to perform enrichment analysis of intersecting DEGs and study their functions. Target genes of DEMs were predicted from the online database miRNet. The protein-protein interaction files of the overlapping target genes were obtained from string and the miRNA-mRNA network was constructed by Cytoscape software. In addition, the online web tool CIBERSORT was used to analyze the immune infiltration of dataset GSE166388, and the relative abundance of 22 immune cells in the diseased and normal control tissues was calculated and assessed. Finally, quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to verify the relative expression of the screened miRNAs and mRNAs to assess the applicability of DEMs and DEGs as biomarkers in PS. A total of 205 mating DEGs and 6 mating DEMs were screened. 103 dysregulated crossover genes from 205 crossover DEGs and 7878 miRNA target genes were identified. The miRNA-mRNA regulatory network was constructed and the top 10 elements were obtained from CytoHubba, including hsa-miR-146a-5p, hsa-miR-17-5p, hsa-miR-106a-5p, hsa-miR-18a-5p, CDK1, CCNA2, CCNB1, MAD2L1, RRM2, and CCNB2. QRT-PCR revealed significant differences in miRNA and gene expression between inflammatory and normal states. In this study, the miRNA-mRNA core regulator pairs hsa-miR-146a-5p, hsa-miR-17-5p, hsa-miR-106a-5p, hsa-miR-18a-5p, CDK1, CCNA2, CCNB1, MAD2L1, RRM2, and CCNB2 may be involved in the course of PS. This study provides new insights to discover new potential targets and biomarkers to further investigate the molecular mechanism of PS.
Collapse
Affiliation(s)
- Lu Chen
- Department of Immunology, Jianghan University, School of Medicine, Wuhan, Hubei, PR China
| | - Xiaochen Wang
- Department of Immunology, Jianghan University, School of Medicine, Wuhan, Hubei, PR China
| | - Chang Liu
- Department of Immunology, Jianghan University, School of Medicine, Wuhan, Hubei, PR China
| | - Xiaoqing Chen
- Department of Immunology, Jianghan University, School of Medicine, Wuhan, Hubei, PR China
| | - Peng Li
- Department of Dermatology, Wuhan Central Hospital, Wuhan, Hubei, PR China
| | - Wenhong Qiu
- Department of Immunology, Jianghan University, School of Medicine, Wuhan, Hubei, PR China
| | - Kaiwen Guo
- Department of Pathogenic Biology, Wuhan University of Science and Technology, Medical College, Wuhan, Hubei, PR China
| |
Collapse
|
30
|
Li D, Shan S, Mao X, Zhao Y, Chen B, Xiong Q, Pan D, Huang S. CS12192, a novel JAK3/JAK1/TBK1 inhibitor, attenuates autoimmune dermatoses in murine models. Immunopharmacol Immunotoxicol 2024:1-9. [PMID: 38918174 DOI: 10.1080/08923973.2024.2373223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
OBJECTIVE Autoimmune dermatosis (AID) occurs when the body's immune system attacks skin or tissue, leading to various types of skin disorders or injuries. Recent studies show that Janus kinases (JAKs) play critical roles in autoimmune diseases including AID by regulating multiple cytokine signaling pathways. CS12192, a novel JAK3/JAK1/TBK1 inhibitor, has been reported to exert ameliorative effects in rheumatoid arthritis. However, the efficacy of CS12192 on AID is undetermined. This study aims to investigate the therapeutic efficacy of CS12192 on psoriasis (PSO), systemic lupus erythematosus (SLE) and atopic dermatitis (AD) in mouse models. METHODS Interleukin-23 (IL-23)-induced PSO model, spontaneous SLE model of MRL/MpJ-Faslpr/J (MRL/lpr) mice, and oxazolone (OXA) and dinitrochlorobenzene (DNCB)-induced murine AD models were used for the evaluation of curative effects of CS12192, respectively. The skin lesion, biochemical parameters, ear thickness, ear weight and histopathology were assessed accordingly. RESULTS In PSO model, mice treated with CS12192 show reduced ear thickness and ear weight as compared with vehicle. In SLE model, CS12192 ameliorates cutaneous parameters such as lymphadenectasis and skin lesion but not systematic parameters such as proteinuria concentration and score, serum dsDNA and BUN concentration. In AD models, CS12192 dose-dependently improves ear swelling and reduces histological scores, exerting equivalent efficacy with baricitinib, a marketed JAK1/JAK2 inhibitor. CONCLUSION Our findings suggest that the novel JAK3/JAK1/TBK1 inhibitor CS12192 is potentially to alleviate autoimmune dermatosis.
Collapse
Affiliation(s)
- Dan Li
- Chengdu Chipscreen Pharmaceutical Ltd, Chengdu, Sichuan, China
| | - Song Shan
- Shenzhen Chipscreen Biosciences Co., Ltd, Shenzhen, Guangdong, China
| | - Xuhua Mao
- Chengdu Chipscreen Pharmaceutical Ltd, Chengdu, Sichuan, China
| | - Yiru Zhao
- Chengdu Chipscreen Pharmaceutical Ltd, Chengdu, Sichuan, China
| | - Beizhong Chen
- Chengdu Chipscreen Pharmaceutical Ltd, Chengdu, Sichuan, China
| | - Qiuyun Xiong
- Chengdu Chipscreen Pharmaceutical Ltd, Chengdu, Sichuan, China
| | - Desi Pan
- Shenzhen Chipscreen Biosciences Co., Ltd, Shenzhen, Guangdong, China
| | - Shengjian Huang
- Chengdu Chipscreen Pharmaceutical Ltd, Chengdu, Sichuan, China
- Shenzhen Chipscreen Biosciences Co., Ltd, Shenzhen, Guangdong, China
| |
Collapse
|
31
|
Ma J, Gan L, Chen H, Chen L, Hu Y, Luan C, Chen K, Zhang J. Upregulated miR-374a-5p drives psoriasis pathogenesis through WIF1 downregulation and Wnt5a/NF-κB activation. Cell Signal 2024; 119:111171. [PMID: 38604345 DOI: 10.1016/j.cellsig.2024.111171] [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: 12/26/2023] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Psoriasis is a chronic, inflammatory skin disease. MicroRNAs (miRNAs) are an abundant class of non-coding RNA molecules. Recent studies have shown that multiple miRNAs are abnormally expressed in patients with psoriasis. The upregulation of miR-374a-5p has been associated with psoriasis severity. However, the specific role of miR-374a-5p in the pathogenesis of psoriasis remain unclear. METHODS qRT-PCR was employed to validate the expression of miR-374a-5p in psoriatic lesions and in a psoriasis-like cell model constructed using a mixture of M5 (IL-17A, IL-22, OSM, IL-1α, and TNF-α). HaCaT cells were transfected with miR-374a-5p mimic/inhibitor, and assays including EdU, CCK-8, and flow cytometry were conducted to evaluate the effect of miR-374a-5p on cell proliferation. The expression of inflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α was verified by qRT-PCR. Bioinformatics analysis and dual-luciferase reporter gene assay were performed to detect the downstream target genes and upstream transcription factors of miR-374a-5p, followed by validation of their expression through qRT-PCR and Western blotting. A psoriasis-like mouse model was established using imiquimod cream topical application. The psoriasis area and severity index scoring, hematoxylin-eosin histology staining, and Ki67 immunohistochemistry were employed to validate the effect of miR-374a-5p on the psoriatic inflammation phenotype after intradermal injection of miR-374a-5p agomir/NC. Additionally, the expression of pathway-related molecules and inflammatory factors such as IL-1β, IL-17a, and TNF-α was verified by immunohistochemistry. RESULTS Upregulation of miR-374a-5p was observed in psoriatic lesions and the psoriasis-like cell model. In vitro experiments demonstrated that miR-374a-5p not only promoted the proliferation of HaCaT cells but also upregulated the expression of inflammatory cytokines, including IL-1β, IL-6, IL-8, and TNF-α. Furthermore, miR-374a-5p promoted skin inflammation and epidermal thickening in the Imiquimod-induced psoriasis-like mouse model. Mechanistic studies revealed that miR-374a-5p led to downregulation of WIF1, thereby activating the Wnt5a/NF-κB signaling pathway. The transcription factor p65 encoded by RELA, as a subunit of NF-κB, further upregulated the expression of miR-374a-5p upon activation. This positive feedback loop promoted keratinocyte proliferation and abnormal inflammation, thereby facilitating the development of psoriasis. CONCLUSION Our findings elucidate the role of miR-374a-5p upregulation in the pathogenesis of psoriasis through inhibition of WIF1 and activation of the Wnt5a/NF-κB pathway, providing new potential therapeutic targets for psoriasis.
Collapse
Affiliation(s)
- Jing Ma
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Lu Gan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Hongying Chen
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Lihao Chen
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yu Hu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Chao Luan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China.
| | - Kun Chen
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China.
| | - Jiaan Zhang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China.
| |
Collapse
|
32
|
Liu X, Sun H, Zheng L, Zhang J, Su H, Li B, Wu Q, Liu Y, Xu Y, Song X, Yu Y. Adipose-derived miRNAs as potential biomarkers for predicting adulthood obesity and its complications: A systematic review and bioinformatic analysis. Obes Rev 2024; 25:e13748. [PMID: 38590187 DOI: 10.1111/obr.13748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/25/2024] [Accepted: 03/10/2024] [Indexed: 04/10/2024]
Abstract
Adipose tissue is the first and primary target organ of obesity and the main source of circulating miRNAs in patients with obesity. This systematic review aimed to analyze and summarize the generation and mechanisms of adipose-derived miRNAs and their role as early predictors of various obesity-related complications. Literature searches in the PubMed and Web of Science databases using terms related to miRNAs, obesity, and adipose tissue. Pre-miRNAs from the Human MicroRNA Disease Database, known to regulate obesity-related metabolic disorders, were combined for intersection processing. Validated miRNA targets were sorted through literature review, and enrichment analysis using the Kyoto Encyclopedia of Genes and Genomes via the KOBAS online tool, disease analysis, and miRNA transcription factor prediction using the TransmiR v. 2.0 database were also performed. Thirty miRNAs were identified using both obesity and adipose secretion as criteria. Seventy-nine functionally validated targets associated with 30 comorbidities of these miRNAs were identified, implicating pathways such as autophagy, p53 pathways, and inflammation. The miRNA precursors were analyzed to predict their transcription factors and explore their biosynthesis mechanisms. Our findings offer potential insights into the epigenetic changes related to adipose-driven obesity-related comorbidities.
Collapse
Affiliation(s)
- Xiyan Liu
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning, China
- Health Sciences Institute, China Medical University, Shenyang, Liaoning, China
| | - Huayi Sun
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning, China
- Department of Colorectal Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lixia Zheng
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning, China
- Health Sciences Institute, China Medical University, Shenyang, Liaoning, China
| | - Jian Zhang
- Health Sciences Institute, China Medical University, Shenyang, Liaoning, China
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| | - Han Su
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning, China
- Health Sciences Institute, China Medical University, Shenyang, Liaoning, China
| | - Bingjie Li
- Health Sciences Institute, China Medical University, Shenyang, Liaoning, China
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| | - Qianhui Wu
- Health Sciences Institute, China Medical University, Shenyang, Liaoning, China
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| | - Yunchan Liu
- Health Sciences Institute, China Medical University, Shenyang, Liaoning, China
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| | - Yingxi Xu
- Health Sciences Institute, China Medical University, Shenyang, Liaoning, China
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaoyu Song
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning, China
- Health Sciences Institute, China Medical University, Shenyang, Liaoning, China
| | - Yang Yu
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang, Liaoning, China
- Health Sciences Institute, China Medical University, Shenyang, Liaoning, China
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
33
|
Chen X, Deng G, Chen K, Chen Y, Ye W, Sun P. Targeting the NLRP3 inflammasome in psoriasis. Int J Dermatol 2024; 63:844-851. [PMID: 38345734 DOI: 10.1111/ijd.17073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 07/05/2024]
Abstract
The NLRP3 inflammasome, a complex consisting of the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3, has emerged as a critical mediator of pathological inflammation and a significant therapeutic target for various inflammatory diseases. Psoriasis, a chronic inflammatory skin condition without a definitive cure, has shown promising results in animal models through the inhibition of the NLRP3 inflammasome. This review aims to explore the development of the NLRP3 inflammasome in psoriasis and the molecular mechanisms responsible for its inhibition by natural products and small molecules currently being developed for psoriasis treatment. Furthermore, we are examining clinical trials using agents that block the NLRP3 pathway for the treatment of psoriasis. This study is timely to provide a new perspective on managing psoriasis.
Collapse
Affiliation(s)
- Xiuhui Chen
- Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan, China
- Department of Pharmacy, Dongguan Eighth People's Hospital, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Guoliang Deng
- Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan, China
- Department of Pharmacy, Dongguan Eighth People's Hospital, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Kaifeng Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yanhong Chen
- Department of Pharmacy, Baoan Central Hospital of Shenzhen, Shenzhen, China
| | - Weijun Ye
- Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan, China
- Department of Pharmacy, Dongguan Eighth People's Hospital, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, China
| | - Ping Sun
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
34
|
Letonja J, Petrovič D. A Review of MicroRNAs and lncRNAs in Atherosclerosis as Well as Some Major Inflammatory Conditions Affecting Atherosclerosis. Biomedicines 2024; 12:1322. [PMID: 38927529 PMCID: PMC11201627 DOI: 10.3390/biomedicines12061322] [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: 04/25/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
It is generally accepted that atherosclerosis is a chronic inflammatory disease. The link between atherosclerosis and other inflammatory diseases such as psoriasis, type 2 diabetes mellitus (T2DM), and rheumatoid arthritis (RA) via metabolic, inflammatory, and immunoregulatory pathways is well established. The aim of our review was to summarize the associations between selected microRNAs (miRs) and long non-coding RNAs (lncRNAs) and atherosclerosis, psoriasis, T2DM, and RA. We reviewed the role of miR-146a, miR-210, miR-143, miR-223, miR-126, miR-21, miR-155, miR-145, miR-200, miR-133, miR-135, miR-221, miR-424, let-7, lncRNA-H19, lncRNA-MEG3, lncRNA-UCA1, and lncRNA-XIST in atherosclerosis and psoriasis, T2DM, and RA. Extracellular vesicles (EVs) are a method of intracellular signal transduction. Their function depends on surface expression, cargo, and the cell from which they originate. The majority of the studies that investigated lncRNAs and some miRs had relatively small sample sizes, which limits the generalizability of their findings and indicates the need for more research. Based on the studies reviewed, miR-146a, miR-155, miR-145, miR-200, miR-133, and lncRNA-H19 are the most promising potential biomarkers and, possibly, therapeutic targets for atherosclerosis as well as T2DM, RA, and psoriasis.
Collapse
Affiliation(s)
- Jernej Letonja
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia;
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| | - Danijel Petrovič
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia;
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| |
Collapse
|
35
|
Chen X, Wu Y, Jia S, Zhao M. Fibroblast: A Novel Target for Autoimmune and Inflammatory Skin Diseases Therapeutics. Clin Rev Allergy Immunol 2024; 66:274-293. [PMID: 38940997 DOI: 10.1007/s12016-024-08997-1] [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] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
Fibroblasts are crucial components of the skin structure. They were traditionally believed to maintain the skin's structure by producing extracellular matrix and other elements. Recent research illuminated that fibroblasts can respond to external stimuli and exhibit diverse functions, such as the secretion of pro-inflammatory factors, adipogenesis, and antigen presentation, exhibiting remarkable heterogeneity and plasticity. This revelation positions fibroblasts as active contributors to the pathogenesis of skin diseases, challenging the traditional perspective that views fibroblasts solely as structural entities. Based on their diverse functions, fibroblasts can be categorized into six subtypes: pro-inflammatory fibroblasts, myofibroblasts, adipogenic fibroblasts, angiogenic fibroblasts, mesenchymal fibroblasts, and antigen-presenting fibroblasts. Cytokines, metabolism, and epigenetics regulate functional abnormalities in fibroblasts. The dynamic changes fibroblasts exhibit in different diseases and disease states warrant a comprehensive discussion. We focus on dermal fibroblasts' aberrant manifestations and pivotal roles in inflammatory and autoimmune skin diseases, including psoriasis, vitiligo, lupus erythematosus, scleroderma, and atopic dermatitis, and propose targeting aberrantly activated fibroblasts as a potential therapeutic strategy for inflammatory and autoimmune skin diseases.
Collapse
Affiliation(s)
- Xiaoyun Chen
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yutong Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Sujie Jia
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
36
|
Zhang B, Mei X, Zhao M, Lu Q. The new era of immune skin diseases: Exploring advances in basic research and clinical translations. J Transl Autoimmun 2024; 8:100232. [PMID: 39022635 PMCID: PMC11252396 DOI: 10.1016/j.jtauto.2024.100232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024] Open
Affiliation(s)
- Bo Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Xiaole Mei
- Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ming Zhao
- Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| |
Collapse
|
37
|
Chen X, Fu C, Zheng Y, Li X, Liao Y, Zheng Y, Liang W, Zhao Y, Huang J, Huang T, Bu J, Shen E. Intermittent fasting alleviates IMQ-induced psoriasis-like dermatitis via reduced γδT17 and monocytes in mice. Arch Dermatol Res 2024; 316:176. [PMID: 38758283 DOI: 10.1007/s00403-024-02886-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/19/2024] [Accepted: 04/14/2024] [Indexed: 05/18/2024]
Abstract
Psoriasis is a chronic immune mediated inflammatory skin disease with systemic manifestations. It has been reported that caloric restriction could improve severity of psoriasis patients. However, the mechanism of intermittent fasting effects on psoriasis has not been investigated. Caloric restriction is known to reduce the number of circulating inflammatory monocytes in a CCL2-dependent manner. However, it is still unknown whether caloric restriction can improve psoriasis by regulating monocytes through CCL2. In this study, we used imiquimod (IMQ)-induced psoriasis-like mouse model to explore the effects and the mechanisms of intermittent fasting on psoriasis-like dermatitis. We found that intermittent fasting could significantly improve IMQ-induced psoriasis-like dermatitis, and reduce the number of γδT17 cells and IL-17 production in draining lymph nodes and psoriatic lesion via inhibiting proliferation and increasing death of γδT17 cells. Furthermore, intermittent fasting could significantly decrease monocytes in blood, and this was associated with decreased monocytes, macrophages and DC in psoriasis-like skin inflammation. Reduced monocytes in circulation and increased monocytes in BM of fasting IMQ-induced psoriasis-like mice is through reducing the production of CCL2 from BM to inhibit monocyte egress to the periphery. Our above data shads light on the mechanisms of intermittent fasting on psoriasis.
Collapse
Affiliation(s)
- Xiangming Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Cheng Fu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
- Shenzhen Children's Hospital, Shenzhen, China
| | - Yanling Zheng
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaomin Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yue Liao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yu Zheng
- National Center for STD Control, Chinese Center for Disease Control and Prevention, Nanjing, 200042, Jiangsu, China
| | - Wenting Liang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yuyang Zhao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Jijun Huang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Ting Huang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Jin Bu
- National Center for STD Control, Chinese Center for Disease Control and Prevention, Nanjing, 200042, Jiangsu, China.
- Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.
| | - Erxia Shen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Xiao W, Sha K, Wang M, Tan Z, Wang Y, Xu S, Zhao Z, Wang Q, Xie H, Chen M, Deng Z, Li J. SERPINB3/B4 Is Increased in Psoriasis and Rosacea Lesions and Has Proinflammatory Effects in Mouse Models of these Diseases. J Invest Dermatol 2024:S0022-202X(24)00367-1. [PMID: 38735363 DOI: 10.1016/j.jid.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024]
Abstract
Psoriasis and rosacea are both chronic inflammatory skin disorders resulted from aberrant keratinocyte-immune cell crosstalk, but the common molecular foundations for these 2 conditions are poorly understood. In this study, we reveal that both patients with psoriasis and those with rosacea as well as their mouse models have significantly elevated expressions of SERPINB3/B4 (members of serine protease inhibitor) in the lesional skin. Skin inflammation in mice that resembles both psoriasis and rosacea is prevented by SERPINB3/B4 deficiency. Mechanistically, we demonstrate that SERPINB3/B4 positively induces NF-κB signaling activation, thereby stimulating disease-characteristic inflammatory chemokines and cytokines production in keratinocytes and promoting the chemotaxis of CD4+ T cells. Our results suggest that in keratinocytes, SERPINB3/B4 may be involved in the pathogenesis of both psoriasis and rosacea by stimulating NF-κB signaling, and they indicate a possible treatment overlap between these 2 diseases.
Collapse
Affiliation(s)
- Wenqin Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ke Sha
- Department of Dermatology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Mei Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zixin Tan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yunying Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - San Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Wang
- Hunan Binsis Biotechnology, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
39
|
Shellard EM, Rane SS, Eyre S, Warren RB. Functional Genomics and Insights into the Pathogenesis and Treatment of Psoriasis. Biomolecules 2024; 14:548. [PMID: 38785955 PMCID: PMC11117854 DOI: 10.3390/biom14050548] [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: 03/11/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Psoriasis is a lifelong, systemic, immune mediated inflammatory skin condition, affecting 1-3% of the world's population, with an impact on quality of life similar to diseases like cancer or diabetes. Genetics are the single largest risk factor in psoriasis, with Genome-Wide Association (GWAS) studies showing that many psoriasis risk genes lie along the IL-23/Th17 axis. Potential psoriasis risk genes determined through GWAS can be annotated and characterised using functional genomics, allowing the identification of novel drug targets and the repurposing of existing drugs. This review is focused on the IL-23/Th17 axis, providing an insight into key cell types, cytokines, and intracellular signaling pathways involved. This includes examination of currently available biological treatments, time to relapse post drug withdrawal, and rates of primary/secondary drug failure, showing the need for greater understanding of the underlying genetic mechanisms of psoriasis and how they can impact treatment. This could allow for patient stratification towards the treatment most likely to reduce the burden of disease for the longest period possible.
Collapse
Affiliation(s)
- Elan May Shellard
- Faculty of Biology, Medicine and Health, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK
| | - Shraddha S. Rane
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, The University of Manchester, Manchester M13 9PT, UK; (S.S.R.); (S.E.)
| | - Stephen Eyre
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, The University of Manchester, Manchester M13 9PT, UK; (S.S.R.); (S.E.)
| | - Richard B. Warren
- Dermatology Centre, Northern Care Alliance NHS Foundation Trust, Manchester M6 8HD, UK;
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M23 9LT, UK
| |
Collapse
|
40
|
Zhang Y, Lu Q. Immune cells in skin inflammation, wound healing, and skin cancer. J Leukoc Biol 2024; 115:852-865. [PMID: 37718697 DOI: 10.1093/jleuko/qiad107] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/22/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023] Open
Abstract
Given the self-evident importance of cutaneous immunity in the maintenance of body-surface homeostasis, disturbance of the steady-state skin is inextricably intertwined with dysfunction in cutaneous immunity. It is often overlooked by people that skin, well-known as a solid physical barrier, is also a strong immunological barrier, considering the abundant presence of immune cells including lymphocytes, granulocytes, dendritic cells, and macrophages. What's more, humoral immune components including cytokines, immunoglobulins, and antimicrobial peptides are also rich in the skin. This review centers on skin inflammation (acute and chronic, infection and aseptic inflammation), wound healing, and skin cancer to elucidate the elaborate network of immune cells in skin diseases.
Collapse
Affiliation(s)
- Yuhan Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
| |
Collapse
|
41
|
Lin ZC, Hung CF, Aljuffali IA, Lin MH, Fang JY. RNA-Based Antipsoriatic Gene Therapy: An Updated Review Focusing on Evidence from Animal Models. Drug Des Devel Ther 2024; 18:1277-1296. [PMID: 38681207 PMCID: PMC11055533 DOI: 10.2147/dddt.s447780] [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: 11/01/2023] [Accepted: 04/07/2024] [Indexed: 05/01/2024] Open
Abstract
Psoriasis presents as a complex genetic skin disorder, characterized by the interaction between infiltrated immune cells and keratinocytes. Substantial progress has been made in understanding the molecular mechanisms of both coding and non-coding genes, which has positively impacted clinical treatment approaches. Despite extensive research into the genetic aspects of psoriasis pathogenesis, fully grasping its epigenetic component remains a challenging endeavor. In response to the pressing demand for innovative treatments to alleviate inflammatory skin disorders, various novel strategies are under consideration. These include gene therapy employing antisense nucleotides, silencing RNA complexes, stem cell therapy, and antibody-based therapy. There is a pressing requirement for a psoriasis-like animal model that replicates human psoriasis to facilitate early preclinical evaluations of these novel treatments. The authors conduct a comprehensive review of various gene therapy in different psoriasis-like animal models utilized in psoriasis research. The animals included in the list underwent skin treatments such as imiquimod application, as well as genetic and biologic injections, and the results of these interventions are detailed. Animal models play a crucial role in translating drug discoveries from the laboratory to clinical practice, and these models aid in improving the reproducibility and clinical applicability of preclinical data. Numerous animal models with characteristics similar to those of human psoriasis have proven to be useful in understanding the development of psoriasis. In this review, the article focuses on RNA-based gene therapy exploration in different types of psoriasis-like animal models to improve the treatment of psoriasis.
Collapse
Affiliation(s)
- Zih-Chan Lin
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi, Chiayi, Taiwan
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Program in Pharmaceutical Biotechnology, Fu Jen Catholic University, New Taipei City, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ibrahim A Aljuffali
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ming-Hsien Lin
- Department of Dermatology, Chi Mei Medical Center, Tainan, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
| |
Collapse
|
42
|
Han Y, Xi L, Leng F, Xu C, Zheng Y. Topical Delivery of microRNA-125b by Framework Nucleic Acids for Psoriasis Treatment. Int J Nanomedicine 2024; 19:2625-2638. [PMID: 38505169 PMCID: PMC10950082 DOI: 10.2147/ijn.s441353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/29/2024] [Indexed: 03/21/2024] Open
Abstract
Purpose Psoriasis is a chronic and recurrent inflammatory dermatitis characterized by T cell imbalance and abnormal keratinocyte proliferation. MicroRNAs (miRNAs) hold promise as therapeutic agents for this disease; however, their clinical application is hindered by poor stability and limited skin penetration. This study demonstrates the utilization of Framework Nucleic Acid (FNA) for the topical delivery of miRNAs in psoriasis treatment. Methods By utilizing miRNA-125b as the model drug, FNA-miR-125b was synthesized via self-assembly. The successful synthesis and stability of FNA-miR-125b in bovine fetal serum (FBS) were verified through gel electrophoresis. Subsequently, flow cytometry was employed to investigate the cell internalization on HaCaT cells, while qPCR determined the effects of FNA-miR-125b on cellular functions. Additionally, the skin penetration ability of FNA-miR-125b was assessed. Finally, a topical administration study involving FNA-miR-125b cream on imiquimod (IMQ)-induced psoriasis mice was conducted to evaluate its therapeutic efficacy. Results The FNA-miR-125b exhibited excellent stability, efficient cellular internalization, and potent inhibition of keratinocyte proliferation. In the psoriasis mouse model, FNA-miR-125b effectively penetrated the skin tissue, resulting in reduced epidermal thickness and PASI score, as well as decreased levels of inflammatory cytokines.
Collapse
Affiliation(s)
- Yunfeng Han
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People’s Republic of China
| | - Long Xi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People’s Republic of China
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, People’s Republic of China
| | - Fang Leng
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People’s Republic of China
| |
Collapse
|
43
|
Mihu C, Popescu CA, Cenariu D, Vesa Ş, Baican A, Melincovici CS, Drulă R, Tigu AB, Buzoianu AD. Is Mir-205 a possible biomarker for evaluating treatment response in psoriasis? J Med Life 2024; 17:353-359. [PMID: 39044928 PMCID: PMC11262607 DOI: 10.25122/jml-2024-0264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 05/20/2024] [Indexed: 07/25/2024] Open
Abstract
Psoriasis is a chronic skin disease that affects a significant number of patients and can severely impair quality of life. Although the diagnosis is normally clinical, paraclinical determination can occasionally be useful either in differential diagnosis or in evaluating the inflammatory response to treatment. MicroRNAs (miRNAs) are small non-coding parts of the RNA family that regulate gene expression and may have an important role as biomarkers in evaluating treatment response. The dysregulation of miRNAs has been well studied in other diseases, especially in oncology, but their role in chronic skin conditions such as psoriasis is still not fully understood. This study aims to evaluate the levels of three miRNAs (miR-155, miR-210, and miR-205) in patients with psoriasis, treated either systemically or topically, compared to a control group, and to assess the possible relationship between miRNA levels and systemic therapy. Our findings show a constant dysregulation of miR-205 in patients with psoriasis, with significantly higher levels compared to the control group, which can be explained as conferring a protective effect to treated patients. Further studies are needed in order to fully understand the role of miRNAs in the physiopathology of psoriasis and even, potentially, to provide more targeted genetic therapies in the future.
Collapse
Affiliation(s)
- Carina Mihu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Codruța Alina Popescu
- Department of Human Sciences, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Cenariu
- Medfuture Research Centre for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ştefan Vesa
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Adrian Baican
- Department of Dermatology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Rareş Drulă
- Medfuture Research Centre for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Adrian Bogdan Tigu
- Medfuture Research Centre for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anca Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
44
|
Carmona-Rocha E, Rusiñol L, Puig L. New and Emerging Oral/Topical Small-Molecule Treatments for Psoriasis. Pharmaceutics 2024; 16:239. [PMID: 38399292 PMCID: PMC10892104 DOI: 10.3390/pharmaceutics16020239] [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: 01/07/2024] [Revised: 01/30/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
The introduction of biologic therapies has led to dramatic improvements in the management of moderate-to-severe psoriasis. Even though the efficacy and safety of the newer biologic agents are difficult to match, oral administration is considered an important advantage by many patients. Current research is focused on the development of oral therapies with improved efficacy and safety compared with available alternatives, as exemplified by deucravacitinib, the first oral allosteric Tyk2 inhibitor approved for the treatment of moderate to severe psoriasis in adults. Recent advances in our knowledge of psoriasis pathogenesis have also led to the development of targeted topical molecules, mostly focused on intracellular signaling pathways such as AhR, PDE-4, and Jak-STAT. Tapinarof (an AhR modulator) and roflumilast (a PDE-4 inhibitor) have exhibited favorable efficacy and safety outcomes and have been approved by the FDA for the topical treatment of plaque psoriasis. This revision focuses on the most recent oral and topical therapies available for psoriasis, especially those that are currently under evaluation and development for the treatment of psoriasis.
Collapse
Affiliation(s)
- Elena Carmona-Rocha
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (E.C.-R.); (L.R.)
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain
- Sant Pau Teaching Unit, School of Medicine, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| | - Lluís Rusiñol
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (E.C.-R.); (L.R.)
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain
- Sant Pau Teaching Unit, School of Medicine, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| | - Lluís Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (E.C.-R.); (L.R.)
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain
- Sant Pau Teaching Unit, School of Medicine, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| |
Collapse
|
45
|
Li J, Yan W, Yuan H, Ren F. Theacrine enhances autophagy and inhibits inflammation via regulating SIRT3/FOXO3a/Parkin pathway. Int J Rheum Dis 2024; 27:e15085. [PMID: 38402443 DOI: 10.1111/1756-185x.15085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Psoriasis, a common chronic inflammatory skin condition, impacts around 2%-3% of the global population. Theacrine is recognized for its potential anti-inflammatory and antioxidant properties. However, the role of theacrine in psoriasis remains unclear. PURPOSES To investigate the effects of theacrine on psoriasis and explore the underlying signaling pathways. METHODS For imiquimod (IMQ)-induced Psoriasis-like mice, the psoriatic inflammation was monitored using Psoriasis Area and Severity Index (PASI). The skin damage was observed using Hematoxylin and Eosin staining. The KI67 and CD4 in skin tissues were assessed using Immunohistochemistry analysis. Western blots were performed to evaluate the expression of Keratin 1 (KRT1), KRT6, LC3, P62, Beclin1, T-bet, GATA3, RAR-related orphan receptor (ROR)-γt, Sirtuin-3 (SIRT3), Forkhead Box O3a (FOXO3a) and Parkin. Additionally, LC3B expression was analyzed using an immunofluorescent assay, while flow cytometry was performed to analyze the percentage of Th17, Th1, and Th2 positive cells in skin-draining lymph node. RESULTS Theacrine improved skin condition by reducing hyperkeratosis and acanthosis, lowering PASI scores, and decreasing KI67-positive cells. Theacrine also modulated keratin expression, elevating KRT1 while reducing KRT6 levels. Theacrine enhanced autophagy indicated by an increased LC3-II/LC3-I ratio and Beclin1, while reduced P62 levels. Additionally, Theacrine reduced CD4-positive cells and suppressed Th17 and Th1 cell activation. Theacrine activated the FOXO3a/Parkin pathway by upregulating SIRT3 expression, and down-regulation of SIRT3 counteracted theacrine's effects in psoriasis-like mice. CONCLUSION Theacrine inhibits skin damage, promotes autophagy, and mediates inflammation in IMQ-induced psoriasis mice via upregulating SIRT3 to activate FOXO3a/Parkin pathway, positioning theacrine as a candidate for psoriasis treatment.
Collapse
Affiliation(s)
- Jin Li
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenliang Yan
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hongshan Yuan
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Fang Ren
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
46
|
Zhang L, Guo H, Zhang X, Wang L, Wei F, Zhao Y, Wang B, Meng Y, Li Y. Small nucleolar RNA Snora73 promotes psoriasis progression by sponging miR-3074-5p and regulating PBX1 expression. Funct Integr Genomics 2024; 24:15. [PMID: 38240925 PMCID: PMC10799104 DOI: 10.1007/s10142-024-01300-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/11/2024] [Accepted: 01/13/2024] [Indexed: 01/22/2024]
Abstract
Chronic psoriasis is a kind of immune-mediated skin illness and the underlying molecular mechanisms of pathogenesis remain incompletely understood. Here, we used small RNA microarray assays to scan the differential expressed RNAs in psoriasis patient samples. The downstream miRNAs and its targets were predicted using bioinformatics analysis from online bases and confirmed using fluorescence in situ hybridization and dual‑luciferase report gene assay. Cell ability of proliferation and migration were detected using CCK-8 and transwell assays. The results showed that a new snoRNA Snora73 was upregulated in psoriasis patient samples. Overexpression of Snora73 significantly increased psoriasis cells viability and migration, while knockdown of Snora73 got the opposite results. Mechanistically, our results showed that Snora73 acted as a sponge for miR-3074-5p and PBX1 is a direct target of miR-3074-5p in psoriasis cells. Furthermore, miR-3074-5p suppressed psoriasis cell proliferation and migration, while PBX1 promoted cell proliferation and migration in psoriasis. Collectively, these findings reveal a crucial role of Snora73 in progression of psoriasis through miR-3074-5p/PBX1 signaling pathway and suggest a potential therapeutic strategy.
Collapse
Affiliation(s)
- Lihua Zhang
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereal Disease in Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Hui Guo
- Key Laboratory of Infection and Immunity of CAS, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaoguang Zhang
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereal Disease in Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Ling Wang
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereal Disease in Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Feng Wei
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereal Disease in Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yike Zhao
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereal Disease in Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Bo Wang
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereal Disease in Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yibo Meng
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereal Disease in Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yanling Li
- Department of Dermatology, Clinical Medical Research Center of Dermatology and Venereal Disease in Hebei Province, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
- Construction Unit of the Sub-Center of the National Center for Clinical Medical Research On Skin and Immunological Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
47
|
Rusiñol L, Puig L. Multi-Omics Approach to Improved Diagnosis and Treatment of Atopic Dermatitis and Psoriasis. Int J Mol Sci 2024; 25:1042. [PMID: 38256115 PMCID: PMC10815999 DOI: 10.3390/ijms25021042] [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: 12/16/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Psoriasis and atopic dermatitis fall within the category of cutaneous immune-mediated inflammatory diseases (IMIDs). The prevalence of IMIDs is increasing in industrialized societies, influenced by both environmental changes and a genetic predisposition. However, the exact immune factors driving these chronic, progressive diseases are not fully understood. By using multi-omics techniques in cutaneous IMIDs, it is expected to advance the understanding of skin biology, uncover the underlying mechanisms of skin conditions, and potentially devise precise and personalized approaches to diagnosis and treatment. We provide a narrative review of the current knowledge in genomics, epigenomics, and proteomics of atopic dermatitis and psoriasis. A literature search was performed for articles published until 30 November 2023. Although there is still much to uncover, recent evidence has already provided valuable insights, such as proteomic profiles that permit differentiating psoriasis from mycosis fungoides and β-defensin 2 correlation to PASI and its drop due to secukinumab first injection, among others.
Collapse
Affiliation(s)
- Lluís Rusiñol
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain
- Unitat Docent Hospital Universitari Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| | - Lluís Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain
- Unitat Docent Hospital Universitari Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| |
Collapse
|
48
|
Xu SJ, Chen JH, Chang S, Li HL. The role of miRNAs in T helper cell development, activation, fate decisions and tumor immunity. Front Immunol 2024; 14:1320305. [PMID: 38264670 PMCID: PMC10803515 DOI: 10.3389/fimmu.2023.1320305] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
T helper (Th) cells are central members of adaptive immunity and comprise the last line of defense against pathogen infection and malignant cell invasion by secreting specific cytokines. These cytokines then attract or induce the activation and differentiation of other immune cells, including antibody-producing B cells and cytotoxic CD8+ T cells. Therefore, the bidirectional communication between Th cells and tumor cells and their positioning within the tumor microenvironment (TME), especially the tumor immune microenvironment (TIME), sculpt the tumor immune landscape, which affects disease initiation and progression. The type, number, and condition of Th cells in the TME and TIME strongly affect tumor immunity, which is precisely regulated by key effectors, such as granzymes, perforins, cytokines, and chemokines. Moreover, microRNAs (miRNAs) have emerged as important regulators of Th cells. In this review, we discuss the role of miRNAs in regulating Th cell mediated adaptive immunity, focusing on the development, activation, fate decisions, and tumor immunity.
Collapse
Affiliation(s)
- Shi-Jun Xu
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- Henan Medical Device Engineering Research Center of Interventional Therapy for Non-vascular Tumors, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Jin-Hua Chen
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Suhwan Chang
- Department of Physiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hai-Liang Li
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
- Henan Medical Device Engineering Research Center of Interventional Therapy for Non-vascular Tumors, Henan Cancer Hospital, Zhengzhou, Henan, China
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
49
|
Li L, Lu J, Liu J, Wu J, Zhang X, Meng Y, Wu X, Tai Z, Zhu Q, Chen Z. Immune cells in the epithelial immune microenvironment of psoriasis: emerging therapeutic targets. Front Immunol 2024; 14:1340677. [PMID: 38239345 PMCID: PMC10794746 DOI: 10.3389/fimmu.2023.1340677] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/15/2023] [Indexed: 01/22/2024] Open
Abstract
Psoriasis is a chronic autoimmune inflammatory disease characterized by erroneous metabolism of keratinocytes. The development of psoriasis is closely related to abnormal activation and disorders of the immune system. Dysregulated skin protective mechanisms can activate inflammatory pathways within the epithelial immune microenvironment (EIME), leading to the development of autoimmune-related and inflammatory skin diseases. In this review, we initially emphasized the pathogenesis of psoriasis, paying particular attention to the interactions between the abnormal activation of immune cells and the production of cytokines in psoriasis. Subsequently, we delved into the significance of the interactions between EIME and immune cells in the emergence of psoriasis. A thorough understanding of these immune processes is crucial to the development of targeted therapies for psoriasis. Finally, we discussed the potential novel targeted therapies aimed at modulating the EIME in psoriasis. This comprehensive examination sheds light on the intricate underlying immune mechanisms and provides insights into potential therapeutic avenues of immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Lisha Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Jiaye Lu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Jun Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Junchao Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Xinyue Zhang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Yu Meng
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Xiying Wu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai University, School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of External Chinese Medicine, Shanghai, China
| |
Collapse
|
50
|
Lian M, Mortoglou M, Uysal-Onganer P. Impact of Hypoxia-Induced miR-210 on Pancreatic Cancer. Curr Issues Mol Biol 2023; 45:9778-9792. [PMID: 38132457 PMCID: PMC10742176 DOI: 10.3390/cimb45120611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Pancreatic cancer (PC) poses significant clinical challenges, with late-stage diagnosis and limited therapeutic options contributing to its dismal prognosis. A hallmark feature of PC is the presence of a profoundly hypoxic tumour microenvironment, resulting from various factors such as fibrotic stroma, rapid tumour cell proliferation, and poor vascularization. Hypoxia plays a crucial role in promoting aggressive cancer behaviour, therapeutic resistance, and immunosuppression. Previous studies have explored the molecular mechanisms behind hypoxia-induced changes in PC, focusing on the role of hypoxia-inducible factors (HIFs). Among the myriad of molecules affected by hypoxia, microRNA-210 (miR-210) emerges as a central player. It is highly responsive to hypoxia and regulated by HIF-dependent and HIF-independent pathways. miR-210 influences critical cellular processes, including angiogenesis, metastasis, and apoptosis, all of which contribute to PC progression and resistance to treatment. Understanding these pathways provides insights into potential therapeutic targets. Furthermore, investigating the role of miR-210 and its regulation in hypoxia sheds light on the potential development of early diagnostic strategies, which are urgently needed to improve outcomes for PC patients. This review delves into the complexities of PC and introduces the roles of hypoxia and miR-210 in the progression of PC.
Collapse
Affiliation(s)
| | | | - Pinar Uysal-Onganer
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK; (M.L.); (M.M.)
| |
Collapse
|