1
|
Wang B, Zhang B, Wu M, Xu T. Unlocking therapeutic potential: Targeting lymphocyte activation Gene-3 (LAG-3) with fibrinogen-like protein 1 (FGL1) in systemic lupus erythematosus. J Transl Autoimmun 2024; 9:100249. [PMID: 39228513 PMCID: PMC11369448 DOI: 10.1016/j.jtauto.2024.100249] [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: 02/22/2024] [Revised: 07/21/2024] [Accepted: 07/26/2024] [Indexed: 09/05/2024] Open
Abstract
Systemic lupus erythematosus (SLE) represents an autoimmune disorder that affects multiple systems. In the treatment of this condition, the focus primarily revolves around inflammation suppression and immunosuppression. Consequently, targeted therapy has emerged as a prevailing approach. Currently, the quest for highly sensitive and specifically effective targets has gained significant momentum in the context of SLE treatment. Lymphocyte activation gene-3 (LAG-3) stands out as a crucial inhibitory receptor that binds to pMHC-II, thereby effectively dampening autoimmune responses. Fibrinogen-like protein 1 (FGL1) serves as the principal immunosuppressive ligand for LAG-3, and their combined action demonstrates a potent immunosuppressive effect. This intricate mechanism paves the way for potential SLE treatment by targeting LAG-3 with FGL1. This work provides a comprehensive summary of LAG-3's role in the pathogenesis of SLE and elucidates the feasibility of leveraging FGL1 as a therapeutic approach for SLE management. It introduces a novel therapeutic target and opens up new avenues of therapeutic consideration in the clinical context of SLE treatment.
Collapse
Affiliation(s)
- Bing Wang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Biqing Zhang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Min Wu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Ting Xu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| |
Collapse
|
2
|
Zhou Y, Liu D, Li H. FGL1 Promotes Tumor Immune Escape in Stomach Adenocarcinoma via the Notch Signaling Pathway. Mol Biotechnol 2024; 66:3203-3212. [PMID: 37902887 DOI: 10.1007/s12033-023-00928-3] [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: 01/17/2023] [Accepted: 10/05/2023] [Indexed: 11/01/2023]
Abstract
Immune escape is the major reason for immunotherapy failure in stomach adenocarcinoma (STAD). We tried to reveal the underlying mechanism of FGL1 influencing STAD in this study. Bioinformatics analyses were conducted to analyze the expression of FGL1, the signaling pathways affected by FGL1, and the relation between FGL1 and immune cell infiltration. Quantitative real-time PCR (qRT-PCR), cell counting kit-8 assay, colony formation assay, flow cytometry and Transwell assay were adopted to analyze FGL1 expression, cell viability, cell proliferation, cell apoptosis, and cell invasion, respectively. Enzyme-linked immunosorbent assay, lactate dehydrogenase method, qRT-PCR and Western blot were adopted to reveal proinflammatory cytokine expression, cytotoxicity and mRNA and protein expression of the Notch signaling-related genes, respectively, after co-culture of STAD cells and CD8+T cells. Nude mice experiment was conducted to validate the results obtained above. FGL1 expressed highly in STAD and could activate the Notch signaling pathway, and it was negatively correlated with CD8+T cell infiltration. Cell experiments confirmed that high expression of FGL1 facilitated proliferation and hindered apoptosis of STAD cells. Knockdown of FGL1 could facilitate expression of pro-inflammatory factors and the cytotoxicity of CD8+T cells in co-culture system of STAD and CD8+ T cells. Knockdown of FGL1 could suppress the expression of the Notch signaling pathway-related genes, and the addition of Notch inhibitor proved that FGL1 promoted immune escape via the Notch signaling pathway. This study investigated the influence of FGL1 on STAD immune escape and demonstrated that FGL1 inhibited CD8+ T cell activation by activating the Notch signaling pathway and thus promoted tumor immune escape in STAD, providing a new potential diagnostic marker and therapeutic target for the immunotherapy of STAD patients.
Collapse
Affiliation(s)
- Yani Zhou
- School of Health Management, Shangluo University, Shangluo, 726000, China
| | - Dan Liu
- Department of Rheumatology, First Affiliated Hospital of Xi'an Medical College, Xi'an, 710077, China
| | - Huirong Li
- Department of Mathematics and Computer Application, Shangluo University, No. 10, Beixin Street, Shangzhou District, Shangluo, 726000, Shaanxi Province, China.
| |
Collapse
|
3
|
Nie J, Qin X, Tao X, Huang J. Exploring the molecular landscape of lymphocyte activation gene-3: A literature review. Medicine (Baltimore) 2024; 103:e39622. [PMID: 39331884 PMCID: PMC11441911 DOI: 10.1097/md.0000000000039622] [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: 06/14/2024] [Accepted: 08/16/2024] [Indexed: 09/29/2024] Open
Abstract
Molecular structure and cellular distribution of lymphocyte activation gene-3 (LAG-3) have been studied extensively since 1990. However, several unresolved questions remain. It is well-established that LAG-3 plays a significant role in maintaining immune homeostasis. The presence of deficiencies in LAG-3 has been observed to be linked with autoimmune disorders, whereas the excessive expression of LAG-3 within the tumor microenvironment hinders immune responses, particularly those mediated by lymphocytes, thereby facilitating immune evasion. Consequently, investigations into these 2 aspects have become a prominent focus in both fundamental and clinical research. The objective of this review is to examine the functions and molecular characteristics of LAG-3, as well as its current clinical applications in the context of tumor immune escape and autoimmune disease. The ultimate aim is to explore and propose novel immune therapy approach.
Collapse
Affiliation(s)
- Jiaqi Nie
- Clinical Laboratory Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xue Qin
- Clinical Laboratory Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiang Tao
- Clinical Laboratory Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jin Huang
- Clinical Laboratory Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
4
|
Xi F, Sun H, Peng H, Lian Z, Wei H, Tian Z, Sun R, Chen Y. Hepatocyte-derived FGL1 accelerates liver metastasis and tumor growth by inhibiting CD8+ T and NK cells. JCI Insight 2024; 9:e173215. [PMID: 38973608 PMCID: PMC11383586 DOI: 10.1172/jci.insight.173215] [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: 06/21/2023] [Accepted: 05/17/2024] [Indexed: 07/09/2024] Open
Abstract
Fibrinogen-like protein 1 (FGL1) contributes to the proliferation and metabolism of hepatocytes; however, as a major ligand of the immune checkpoint, its role in the liver regional immune microenvironment is poorly understood. Hepatocytes specifically and highly expressed FGL1 under normal physiological conditions. Increases in hepatic CD8+ T and NK cell numbers and functions were found in Fgl1-deficient (Fgl1-/-) mice, but not in the spleen or lymph node, similar to findings in anti-FGL1 mAb-treated wild-type mice. Furthermore, Fgl1 deficiency or anti-FGL1 mAb blockade restrained liver metastasis and slowed the growth of orthotopic tumors, with significantly prolonged survival of tumor-bearing mice. Tumor-infiltrating hepatic CD8+ T and NK cells upregulated the expression of lymphocyte activation gene-3 (LAG-3) and exhibited stronger antitumor activities after anti-FGL1 treatment. The antitumor efficacy of FGL1 blockade depended on cytotoxic T lymphocytes and NK cells, demonstrated by using a cell-deficient mouse model and cell transfer in vivo. In vitro, FGL1 directly inhibited hepatic T and NK cells related to the receptor LAG-3. In conclusion, hepatocyte-derived FGL1 played critical immunoregulatory roles in the liver and contributed to liver metastasis and tumor growth by inhibiting CD8+ T and NK cell functions via the receptor LAG-3, providing a new strategy for liver cancer immunotherapy.
Collapse
Affiliation(s)
- Fengjia Xi
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Hefei, China
| | - Haoyu Sun
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Hefei, China
| | - Hui Peng
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Hefei, China
| | - Zhexiong Lian
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Hefei, China
| | - Haiming Wei
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Hefei, China
| | - Zhigang Tian
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Hefei, China
| | - Rui Sun
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Hefei, China
| | - Yongyan Chen
- Key Laboratory of Immune Response and Immunotherapy, the Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Hefei, China
| |
Collapse
|
5
|
Tampa M, Mitran MI, Mitran CI, Matei C, Georgescu SR. Psoriasis: What Is New in Markers of Disease Severity? MEDICINA (KAUNAS, LITHUANIA) 2024; 60:337. [PMID: 38399624 PMCID: PMC10889935 DOI: 10.3390/medicina60020337] [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: 01/31/2024] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024]
Abstract
Introduction. Psoriasis is a chronic inflammatory skin disease and is the result of the interaction between numerous external and internal factors. Psoriasis presents a wide range of skin manifestations encompassing individual lesions varying from pinpoint to large plaques that can evolve into generalised forms. The lesions mirror the pathophysiological mechanisms involved in psoriasis pathogenesis, such as inflammation, dysregulation of immune response, uncontrolled proliferation of keratinocytes and angiogenesis. In this article, we present the latest advances achieved regarding markers that correlate with psoriasis severity. Material and method. We have performed a narrative review on markers of psoriasis severity, including articles published between March 2018-March 2023. Results. We have identified four categories of markers: inflammation markers, oxidative stress markers, hormonal markers and cancer-related markers. The main focus was on inflammation biomarkers, including immunomodulatory molecules, haematological parameters, inflammatory cells and costimulatory molecules. Conclusions. The analysed data indicate that markers associated with inflammation, oxidative stress and hormones, and cancer-related markers could be useful in assessing the severity of psoriasis. Nevertheless, additional research is required to ascertain the practical importance of these biomarkers in clinical settings.
Collapse
Affiliation(s)
- Mircea Tampa
- Department of Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.T.); (C.M.); (S.R.G.)
- Department of Dermatology, ‘Victor Babes’ Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania
| | - Madalina Irina Mitran
- Department of Microbiology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Cristina Iulia Mitran
- Department of Microbiology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Clara Matei
- Department of Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.T.); (C.M.); (S.R.G.)
| | - Simona Roxana Georgescu
- Department of Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.T.); (C.M.); (S.R.G.)
- Department of Dermatology, ‘Victor Babes’ Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania
| |
Collapse
|
6
|
Meng M, Wang L, Yao Y, Lin D, Wang C, Yao J, Sun H, Liu M. Ganoderma lucidum polysaccharide peptide (GLPP) attenuates rheumatic arthritis in rats through inactivating NF-κB and MAPK signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:155010. [PMID: 37586160 DOI: 10.1016/j.phymed.2023.155010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/24/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Not many drugs with fewer side effects are available for the treatment of rheumatoid arthritis (RA). Ganoderma lucidum polysaccharide peptide (GLPP) has good immunomodulatory effects, but whether it is effective in managing RA is not clear. PURPOSE This study was conducted to examine the anti-RA activity and possible mechanisms of GLPP in collagen-induced arthritis (CIA) rats. METHODS Male Wistar rats were intradermally injected with bovine type II collagen in the tail base to establish the CIA model and were orally administered 100 or 200 mg/kg GLPP for 35 days. Paw thickness, clinical arthritis scores, gait analysis, organ index determination, blood cell counts, micro-CT imaging and pathological staining were performed on the rats. Liver and kidney function were measured by commercial kits, and antibody levels were measured by ELISA kits. RA-related protein levels were detected by Western blotting. RESULTS GLPP effectively alleviated CIA symptoms and reduced immune organ indexes, antibody levels and systemic organ injury. GLPP decreased the protein expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, matrix metalloproteinase (MMP)2, MMP9, MMP13, BCL-2, OPN, β-Catenin, and hypoxia inducible factor (HIF)-1α and increased the protein expression of BAX in the joint tissues of CIA rats. Moreover, GLPP decreased the phosphorylation levels of p65, IκB-α and ERK1/2. CONCLUSION GLPP effectively alleviated RA symptoms in CIA rats by inhibiting the NF-κB and MAPK pathways. This study suggests a promising therapeutic effect of mushroom-derived polysaccharide peptides on RA.
Collapse
Affiliation(s)
- Meng Meng
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Lianfu Wang
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yang Yao
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Department of Basic Medicine, Chongqing Three Gorges Medical College, Wanzhou, Chongqing, China.
| | - DongMei Lin
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Jialin Yao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Mozhen Liu
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
7
|
Zhang S, Duan Z, Liu F, Wu Q, Sun X, Ma H. The impact of exosomes derived from distinct sources on rheumatoid arthritis. Front Immunol 2023; 14:1240747. [PMID: 37575235 PMCID: PMC10414108 DOI: 10.3389/fimmu.2023.1240747] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that can induce joint deformities and functional impairment, significantly impacting the overall well-being of individuals. Exosomes, which are cellularly secreted vesicles, possess favorable biological traits such as biocompatibility, stability, and minimal toxicity. Additionally, they contain nucleic acids, lipids, proteins, amino acids, and metabolites, serving as mediators in cellular communication and information exchange. Recent studies have demonstrated the association between exosomes and the pathogenesis of RA. Exosomes derived from mesenchymal stem cells, dendritic cells, and neutrophils exert influence on the biological functions of immune cells and joint cells, however, the precise mechanism remains largely unclarified. This comprehensive review systematically analyzes and summarizes the biological characteristics and functionalities of exosomes derived from diverse cellular sources, thus establishing a scientific foundation for the utilization of exosomes as diagnostic targets and therapeutic modalities in the context of RA.
Collapse
Affiliation(s)
- Sicheng Zhang
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Zhen Duan
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Fang Liu
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Qingjie Wu
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Xiwei Sun
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Hailong Ma
- Department of Pediatric Orthopedics, Anhui Provincial Children’s Hospital, Hefei, China
| |
Collapse
|
8
|
Zhou X, Gu Y, Wang H, Zhou W, Zou L, Li S, Hua C, Gao S. From bench to bedside: targeting lymphocyte activation gene 3 as a therapeutic strategy for autoimmune diseases. Inflamm Res 2023:10.1007/s00011-023-01742-y. [PMID: 37314518 DOI: 10.1007/s00011-023-01742-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/12/2023] [Accepted: 05/12/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Immune checkpoints negatively regulate immune response, thereby playing an important role in maintaining immune homeostasis. Substantial studies have confirmed that blockade or deficiency of immune checkpoint pathways contributes to the deterioration of autoimmune diseases. In this context, focusing on immune checkpoints might provide alternative strategies for the treatment of autoimmunity. Lymphocyte activation gene 3 (LAG3), as a member of immune checkpoint, is critical in regulating immune responses as manifested in multiple preclinical studies and clinical trials. Recent success of dual-blockade of LAG3 and programmed death-1 in melanoma also supports the notion that LAG3 is a crucial regulator in immune tolerance. METHODS We wrote this review article by searching the PubMed, Web of Science and Google Scholar databases. CONCLUSION In this review, we summarize the molecular structure and the action mechanisms of LAG3. Additionally, we highlight its roles in diverse autoimmune diseases and discuss how the manipulation of the LAG3 pathway can serve as a promising therapeutic strategy as well as its specific mechanism with the aim of filling the gaps from bench to bedside.
Collapse
Affiliation(s)
- Xueyin Zhou
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yiming Gu
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huihong Wang
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wei Zhou
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lei Zou
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shuting Li
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
9
|
Gao Y, Zhang Z, Du J, Yang X, Wang X, Wen K, Sun X. Xue-Jie-San restricts ferroptosis in Crohn's disease via inhibiting FGL1/NF-κB/STAT3 positive feedback loop. Front Pharmacol 2023; 14:1148770. [PMID: 37153794 PMCID: PMC10154545 DOI: 10.3389/fphar.2023.1148770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/11/2023] [Indexed: 05/10/2023] Open
Abstract
Crohn's disease (CD) is an incurable inflammatory bowel disease due to unclear etiology and pathogenesis. Accumulating evidences have shown the harmful role of ferroptosis in CD onset and development. Additionally, fibrinogen-like protein 1 (FGL1) has been verified to be a potential therapeutic target of CD. Xue-Jie-San (XJS) is an effective prescription for treating CD. However, its therapeutic mechanism has not been fully elucidated. This study aimed to determine whether XJS alleviating CD via regulating ferroptosis and FGL1 expression. A colitis rat model was induced by 2,4,6-trinitrobenzene sulfonic acid and treated with XJS. The disease activity indices of the colitis rats were scored. Histopathological damage was assessed using HE staining. ELISA was performed to examine inflammatory cytokines. Transmission electron microscopy was utilized to observe ultrastructure changes in intestinal epithelial cells (IECs). Iron load was evaluated by examining iron concentrations, the expressions of FPN, FTH and FTL. Lipid peroxidation was investigated through detecting the levels of ROS, 4-HNE, MDA and PTGS2. Furthermore, the SLC7A11/GSH/GPX4 antioxidant system and FGL1/NF-κB/STAT3 signaling pathway were examined. The results showed that colitis was dramatically ameliorated in the XJS-treated rats as evidenced by relief of clinical symptoms and histopathological damages, downregulation of pro-inflammatory cytokines IL-6, IL-17 and TNF-α, and upregulation of anti-inflammatory cytokine IL-10. Furthermore, XJS administration led to ferroptosis inhibition in IECs by reducing iron overload and lipid peroxidation. Mechanistically, XJS enhanced the SLC7A11/GSH/GPX4 antioxidant system negatively regulated by the FGL1/NF-κB/STAT3 positive feedback loop. In conclusion, XJS might restrain ferroptosis in IECs to ameliorate experimental colitis by inhibition of FGL1/NF-κB/STAT3 positive feedback loop.
Collapse
|
10
|
Xu W, Liu X, Qu W, Wang X, Su H, Li W, Cheng Y. Exosomes derived from fibrinogen-like protein 1-overexpressing bone marrow-derived mesenchymal stem cells ameliorates rheumatoid arthritis. Bioengineered 2022; 13:14545-14561. [PMID: 36694465 PMCID: PMC9995129 DOI: 10.1080/21655979.2022.2090379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Rheumatoid arthritis (RA) is a most common chronic joint disease belonging to inflammatory autoimmune disease. The aim of this study was to determine the role and mechanism of bone marrow mesenchymal stem cells (BMSCs)-derived exosomes and fibrinogen-like protein 1 (FGL1) overexpression exosomes shuttled by BMSCs (FGL1-Exos) on RA. All of the exosomes were visualized by transmission electron microscope (TEM) and the characteristic proteins were detected by western blot. To investigate the therapeutic effect of FGL1-Exos, RA-FLSs were activated by TNF-α and RA rat model was established by collagen incomplete Freund's adjuvant. Cell viability, apoptosis, inflammation factors, and protein levels were detected by CCK-8, flow cytometry, enzyme-linked immunosorbent assay and western blot, respectively. Hematoxylin and eosin and safranin O staining were used to detect the histopathology changes. Cell apoptosis and FGL1 expression in knee joint were detected by immunofluorescence. The results showed that FGL1-Exos could inhibit the cell viability meanwhile increase the cell apoptosis in RA-FLSs. Meanwhile, FGL1-Exos could effectively suppress the inflammation score, joint destruction, and inflammatory response in RA rat model. FGL1-Exos directly inhibited cell apoptosis of RA-FLSs and RA rat model by suppressing the inflammatory cytokines, specific rheumatoid markers, immunological markers meanwhile meditating the NF-κB pathway. Our results indicate that FGL1 was a therapeutic potential target in RA therapy.
Collapse
Affiliation(s)
- Wenqiang Xu
- Department of Orthopaedics, the Affiliated Laishan Branch of Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Xiaofeng Liu
- Department of Traumatic Orthopaedics, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Wenqing Qu
- Department of Orthopaedics, YanTaiShan Hospital, Yantai, Shandong, China
| | - Xin Wang
- Department of Traumatic Orthopaedics, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Hao Su
- Department of Traumatic Orthopaedics, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Wenliang Li
- Department of Orthopaedics, YanTaiShan Hospital, Yantai, Shandong, China
| | - Yiheng Cheng
- Department of Orthopaedics, YanTaiShan Hospital, Yantai, Shandong, China
| |
Collapse
|