1
|
Ma J, Zhou Y, Chen J, Guo S, Zhang W, Yi X, Du P, Wang Y, Chen J, Li S, Gao T, Li C, Jian Z. Exosomes enriched with miR-31-3p from keratinocytes under oxidative stress promote vitiligo progression by destructing melanocytes and activating CD8 + T cells. Int J Biol Macromol 2025; 310:143070. [PMID: 40220810 DOI: 10.1016/j.ijbiomac.2025.143070] [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: 12/03/2024] [Revised: 02/12/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Vitiligo is a skin disease characterized by the destruction of epidermal melanocytes due to oxidative stress. Keratinocytes are the main responder to oxidative stress and facilitate melanocyte loss by inducing melanocyte death and recruiting antigen-specific CD8+ T cell to skin to destroy melanocytes. It has been proved that keratinocytes secrete functional exosomes, but the role of exosomes secreted from keratinocytes under oxidative stress in vitiligo pathogenesis is unknown. The present study investigated the role of exosomes from H2O2-treated human keratinocytes in the vitiligo progression in vitro. and in vivo. The results demonstrated that oxidative stress enhanced the secretion of exosomes from keratinocytes. These exosomes (OS-Exos) suppressed the survival of melanocytes while promoting the proliferation and activation of CD8+ T cells in vitro. Then, we confirmed that OS-Exos administration aggravated melanocyte loss and CD8+ T cell infiltration in the epidermis in the vitiligo mouse model, thereby driving vitiligo progression. Further, we performed Small RNAs-seq to screen miRNAs enriched in OS-Exos. The subsequent results revealed that miR-31-3p, which was enriched in OS-Exos, facilitated melanocyte death and decreased the expression of melanogenesis-related genes through MITF signaling. Meanwhile, it was found that miR-31-3p promoted the activation of CD8+ T cells, which could depend on impaired immunosuppression and activated T-cell growth. Taken together, these data suggest that OS-Exos enriched with miR-31-3p facilitated vitiligo progression through the destruction of melanocytes and activation of CD8+ T cells. Keratinocytes-derived exosomes under oxidative stress could serve as an important mediator for oxidative stress-induced killing of melanocytes in vitiligo.
Collapse
Affiliation(s)
- Jingjing Ma
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Yuqi Zhou
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Jianru Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Sen Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Weigang Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Xiuli Yi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Pengran Du
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Yinghan Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Jiaxi Chen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Shuli Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China.
| | - Zhe Jian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 Changlexi Road, Xi'an 710032, Shaanxi, China.
| |
Collapse
|
2
|
Malik A, Sharma D, Aguirre-Gamboa R, McGrath S, Zabala S, Weber C, Jabri B. Epithelial IFNγ signalling and compartmentalized antigen presentation orchestrate gut immunity. Nature 2023; 623:1044-1052. [PMID: 37993709 PMCID: PMC11361632 DOI: 10.1038/s41586-023-06721-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/06/2023] [Indexed: 11/24/2023]
Abstract
All nucleated cells express major histocompatibility complex I and interferon-γ (IFNγ) receptor1, but an epithelial cell-specific function of IFNγ signalling or antigen presentation by means of major histocompatibility complex I has not been explored. We show here that on sensing IFNγ, colonic epithelial cells productively present pathogen and self-derived antigens to cognate intra-epithelial T cells, which are critically located at the epithelial barrier. Antigen presentation by the epithelial cells confers extracellular ATPase expression in cognate intra-epithelial T cells, which limits the accumulation of extracellular adenosine triphosphate and consequent activation of the NLRP3 inflammasome in tissue macrophages. By contrast, antigen presentation by the tissue macrophages alongside inflammasome-associated interleukin-1α and interleukin-1β production promotes a pathogenic transformation of CD4+ T cells into granulocyte-macrophage colony-stimulating-factor (GM-CSF)-producing T cells in vivo, which promotes colitis and colorectal cancer. Taken together, our study unravels critical checkpoints requiring IFNγ sensing and antigen presentation by epithelial cells that control the development of pathogenic CD4+ T cell responses in vivo.
Collapse
Affiliation(s)
- Ankit Malik
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA.
| | - Deepika Sharma
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Raúl Aguirre-Gamboa
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Shaina McGrath
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Sarah Zabala
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Christopher Weber
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA
- Department of Pathology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Bana Jabri
- Department of Medicine, Committee on Immunology, Department of Pediatrics, Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
Abstract
Inflammation is a biological process that dynamically alters the surrounding microenvironment, including participating immune cells. As a well-protected organ surrounded by specialized barriers and with immune privilege properties, the central nervous system (CNS) tightly regulates immune responses. Yet in neuroinflammatory conditions, pathogenic immunity can disrupt CNS structure and function. T cells in particular play a key role in promoting and restricting neuroinflammatory responses, while the inflamed CNS microenvironment can influence and reshape T cell function and identity. Still, the contraction of aberrant T cell responses within the CNS is not well understood. Using autoimmunity as a model, here we address the contribution of CD4 T helper (Th) cell subsets in promoting neuropathology and disease. To address the mechanisms antagonizing neuroinflammation, we focus on the control of the immune response by regulatory T cells (Tregs) and describe the counteracting processes that preserve their identity under inflammatory challenges. Finally, given the influence of the local microenvironment on immune regulation, we address how CNS-intrinsic signals reshape T cell function to mitigate abnormal immune T cell responses.
Collapse
Affiliation(s)
- Nail Benallegue
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, F-44000, Nantes, France
| | - Hania Kebir
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jorge I. Alvarez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
4
|
Liu Y, Sun Y, Bai X, Li L, Zhu G. Albiflorin Alleviates Ox-LDL-Induced Human Umbilical Vein Endothelial Cell Injury through IRAK1/TAK1 Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6584645. [PMID: 35601145 PMCID: PMC9122697 DOI: 10.1155/2022/6584645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 11/21/2022]
Abstract
Introduction Atherosclerosis (AS) is a chronic inflammatory disease characterized by lipid metabolism disorder and vascular endothelial damage. Albiflorin (AF) has been certified to be effective in the therapy of certain inflammatory diseases, while the therapeutic effect and mechanism of AF on AS have not been fully elucidated. Material and Methods. Model cells for AS were created by inducing oxidized low-density lipoprotein (Ox-LDL) in human umbilical vein endothelial cells (HUVECs). After processing with AF and interleukin-1 receptor-associated kinase 1- (IRAK1-) overexpressed plasmid, cell viability was assessed by CCK-8; cholesterol efflux was tested using liquid scintillation counter; IL-6 and TNF-α levels were determined with ELISA kits; ROS and apoptosis were confirmed using Flow cytometry. Besides, IRAK1-TAK1 pathway and apoptosis- and mitochondrial fusion-related proteins were monitored with western blotting analysis. Results Our results verified that AF could not only dramatically accelerate viability and cholesterol efflux but also attenuate inflammation, ROS production, and apoptosis in Ox-LDL-induced HUVECs. Meanwhile, AF could prominently prevent the activation of IRAK1-TAK1 pathway, downregulate apoptosis-related proteins, and upregulate mitochondrial fusion-related proteins in Ox-LDL-induced HUVECs. Moreover, we testified that IRAK1 overexpression memorably could reverse suppression of AF on inflammation, apoptosis, and IRAK1-TAK1 pathway and enhancement of AF on viability, cholesterol efflux, and mitochondrial fusion in Ox-LDL-induced HUVECs. Conclusions By blocking the IRAK1/TAK1 pathway, AF can significantly slow the course of AS, suggesting that it could be a viable therapeutic option for AS.
Collapse
Affiliation(s)
- Yeling Liu
- Department of Pharmacy, Tai'an City Central Hospital, Tai'an, Shandong 271000, China
| | - Yilai Sun
- Department of Pancreatic & Hernial Surgery Tai'an City Central Hospital, Tai'an, Shandong 271000, China
| | - Xue Bai
- Department of Cardiovascular Medicine, Tai'an City Central Hospital, Tai'an, Shandong 271000, China
| | - Lingxing Li
- Department of Cardiovascular Medicine, Tai'an City Central Hospital, Tai'an, Shandong 271000, China
| | - Guihua Zhu
- Department of Pharmacy, Tai'an City Central Hospital, Tai'an, Shandong 271000, China
| |
Collapse
|
5
|
Gao J, Xu Y, Ma S, Liang Y, Liu C, Shen J, Sun Z, Niu M, Xu K, Pan B. Inhibition of interleukin-1 receptor-associated kinase 1 decreases murine acute GVHD while preserving graft-versus-lymphoma effect. Transplant Cell Ther 2021; 28:134.e1-134.e10. [PMID: 34896653 DOI: 10.1016/j.jtct.2021.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/11/2021] [Accepted: 12/02/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Activation of antigen presenting cells (APC) is crucial in initiating inflammation and alloreaction during acute graft-versus-host disease (aGVHD), a common life-threatening complication of allogeneic hematopoietic cell transplantation. Interleukin-1 receptor-associated kinase 1 (IRAK1) regulates activation of APC in inflammatory settings. Inhibition of IRAK1 might decrease APC activation and aGVHD. OBJECTIVE To explore the impact of IRAK1 inhibition on APC activation and aGVHD in mice. STUDY DESIGN We administrated a selective IRAK1 inhibitor Jh-X-119-01 to recipient mice receiving allotransplants or co-challenged by A20 lymphoma cells. We assessed aGVHD and graft-versus-lymphoma (GVL) effect. Activations of T-cell and APC were also analyzed. RESULTS Jh-X-119-01 increased survival and decreased aGVHD of recipients. Jh-X-119-01 decreased proportions of Th1 cells and Tc1 cells in the aGVHD model and in the in vitro mixed lymphocyte reaction (MLR). The IRAK1 inhibitor reduced productions of TNFα and IFNγ in macrophages of recipient mice. In the in vitro cultured bone marrow dendric cells (BMDCs), Jh-X-119-01 decreased productions of inflammatory cytokines, reduced expressions of CD80 and CD86, and decreased protein levels of anti-apoptotic Bcl2 and phosphorylated NF-κB p65. RNA-seq analysis showed Jh-X-119-01 had an impact on several pathophysiological processes of BMDCs such as reduction of GVHD-relation genes and regulation of helper T cell differentiation. Importantly, IRAK1 inhibition did not impair cytotoxic function of T-cell or the allotransplant-related GVL effect against A20 lymphoma cells. In addition, the IRAK1 inhibitor did not retard recovery of hematopoietic cells in blood or bone marrow. CONCLUSION We show selective IRAK1 inhibition ameliorates murine aGVHD but preserves GVL effect. Our findings may have implication for using an IRAK1 inhibitor in allotransplant.
Collapse
Affiliation(s)
- Jun Gao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Yan Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Sha Ma
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Yiwen Liang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Cong Liu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Jingyi Shen
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Zengtian Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Mingshan Niu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.
| | - Bin Pan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
6
|
Cytokines and Transgenic Matrix in Autoimmune Diseases: Similarities and Differences. Biomedicines 2020; 8:biomedicines8120559. [PMID: 33271810 PMCID: PMC7761121 DOI: 10.3390/biomedicines8120559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases are increasingly recognized as disease entities in which dysregulated cytokines contribute to tissue-specific inflammation. In organ-specific and multiorgan autoimmune diseases, the cytokine profiles show some similarities. Despite these similarities, the cytokines have different roles in the pathogenesis of different diseases. Altered levels or action of cytokines can result from changes in cell signaling. This article describes alterations in the JAK-STAT, TGF-β and NF-κB signaling pathways, which are involved in the pathogenesis of multiple sclerosis and systemic lupus erythematosus. There is a special focus on T cells in preclinical models and in patients afflicted with these chronic inflammatory diseases.
Collapse
|
7
|
Downregulation of Inflammatory Cytokine Release from IL-1β and LPS-Stimulated PBMC Orchestrated by ST2825, a MyD88 Dimerisation Inhibitor. Molecules 2020; 25:molecules25184322. [PMID: 32967164 PMCID: PMC7570868 DOI: 10.3390/molecules25184322] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/31/2022] Open
Abstract
The inflammatory process implicates homeostasis disruption and increased production of inflammatory mediators. Myeloid differentiation primary response 88 (MyD88) is an essential protein recruited after lipopolysaccharide (LPS) and interleukin (IL)-1β stimulation, a process that converges in nuclear factor kappa B (NF-κB) activation, as well as a transcription of several genes of both pro- and anti-inflammatory cytokines. The inhibition of MyD88 has shown efficacy by decrease inflammatory response, and has demonstrated potential application as a therapeutic target in chronic diseases. In this study, we investigate the effect of MyD88 dimerisation inhibitor ST2825 on cytokine production from rhIL-1β and LPS-stimulated peripheral blood mononuclear cells (PBMC) from healthy blood donors (HBD). ST2825 significantly downregulates the production of IFN-γ, IL-6, IL-12, IL-2, IL-15, IL-7, VEGF, IL-1Ra, IL-4, IL-5, IL-13 and IL-9 (p < 0.05) in LPS-stimulated PBMC. Moreover, ST2825 had a relatively low impact on IL-1β signalling pathway inhibition, showing that only a few specific cytokines, such as IFN-γ and IL-1Ra, are inhibited in rhIL-1β-stimulated PBMC (p < 0.01). In conclusion, MyD88 dimerisation inhibitor ST2825 showed high efficacy by inhibiting pro- and anti-inflammatory cytokine production in LPS-stimulated PBMC. Moreover, although rhIL-1β induced a sustained cytokine production (p < 0.05), ST2825 did not show a significant effect in the secretion of neither pro- nor anti-inflammatory cytokines in rhIL-1β-stimulated PBMC.
Collapse
|
8
|
Peng H, Ning H, Wang Q, Lai J, Wei L, Stumpo DJ, Blackshear PJ, Fu M, Hou R, Hoft DF, Liu J. Tristetraprolin Regulates T H17 Cell Function and Ameliorates DSS-Induced Colitis in Mice. Front Immunol 2020; 11:1952. [PMID: 32922402 PMCID: PMC7457025 DOI: 10.3389/fimmu.2020.01952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/20/2020] [Indexed: 02/05/2023] Open
Abstract
TH17 cells have been extensively investigated in inflammation, autoimmune diseases, and cancer. The precise molecular mechanisms for TH17 cell regulation, however, remain elusive, especially regulation at the post-transcriptional level. Tristetraprolin (TTP) is an RNA-binding protein important for degradation of the mRNAs encoding several proinflammatory cytokines. With newly generated T cell-specific TTP conditional knockout mice (CD4CreTTPf/f), we found that aging CD4CreTTPf/f mice displayed an increase of IL-17A in serum and spontaneously developed chronic skin inflammation along with increased effector TH17 cells in the affected skin. TTP inhibited TH17 cell development and function by promoting IL-17A mRNA degradation. In a DSS-induced colitis model, CD4CreTTPf/f mice displayed severe colitis and had more TH17 cells and serum IL-17A compared with wild-type mice. Furthermore, neutralization of IL-17A reduced the severity of colitis. Our results reveal a new mechanism for regulating TH17 function and TH17-mediated inflammation post-transcriptionally by TTP, suggests that TTP might be a novel therapeutic target for the treatment of TH17-mediated diseases.
Collapse
Affiliation(s)
- Hui Peng
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Huan Ning
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Qinghong Wang
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Jinping Lai
- Department of Pathology, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Lin Wei
- Department of Immunology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Deborah J. Stumpo
- National Institute of Environmental Health Sciences, Research Triangle, NC, United States
| | - Perry J. Blackshear
- National Institute of Environmental Health Sciences, Research Triangle, NC, United States
| | - Mingui Fu
- Shock/Trauma Research Center and Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Rong Hou
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Daniel F. Hoft
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Jianguo Liu
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
9
|
Hamilton JA. GM-CSF in inflammation. J Exp Med 2020; 217:jem.20190945. [PMID: 31611249 PMCID: PMC7037240 DOI: 10.1084/jem.20190945] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/09/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
GM-CSF is a potential therapeutic target in inflammation and autoimmunity. This study reviews the literature on the biology of GM-CSF, in particular that describing the research leading to clinical trials targeting GM-CSF and its receptor in numerous inflammatory/autoimmune conditions, such as rheumatoid arthritis. Granulocyte–macrophage colony-stimulating factor (GM-CSF) has many more functions than its original in vitro identification as an inducer of granulocyte and macrophage development from progenitor cells. Key features of GM-CSF biology need to be defined better, such as the responding and producing cell types, its links with other mediators, its prosurvival versus activation/differentiation functions, and when it is relevant in pathology. Significant preclinical data have emerged from GM-CSF deletion/depletion approaches indicating that GM-CSF is a potential target in many inflammatory/autoimmune conditions. Clinical trials targeting GM-CSF or its receptor have shown encouraging efficacy and safety profiles, particularly in rheumatoid arthritis. This review provides an update on the above topics and current issues/questions surrounding GM-CSF biology.
Collapse
Affiliation(s)
- John A Hamilton
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Australian Institute for Musculoskeletal Science, The University of Melbourne and Western Health, St Albans, Victoria, Australia
| |
Collapse
|
10
|
Bao P, Zhao W, Mou M, Liu X. MicroRNA-21 mediates bone marrow mesenchymal stem cells protection of radiation-induced lung injury during the acute phase by regulating polarization of alveolar macrophages. Transl Cancer Res 2020; 9:231-239. [PMID: 35117177 PMCID: PMC8798259 DOI: 10.21037/tcr.2019.12.77] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/26/2019] [Indexed: 12/01/2022]
Abstract
Background Radiation-induced lung injury (RILI) often occurs in patients with non-small cell lung cancer (NSCLC) after radiotherapy, and the prognosis of patients with RILI is usually poor. This work plan to investigate the expression patterns of microRNA-21(miR-21) in NSCLC patients with RILI and the protective effects of miR-21 over-expressed bone marrow mesenchymal stem cells (BMSCs) against RILI in rat model. Methods MiR-21 expressions were determined in both serum samples and bronchoalveolar lavage fluid (BALF) samples from NSCLC patients after radiation therapy. The correlation between miR-21 expression and the follow-up clinical characterizations were determined. Further, miR-21 over-expressed BMSCs were transplanted into RILI rats and the protective effects were evaluated. BMSCs and alveolar macrophages (AMs) were co-cultured in vitro and the macrophage M1 polarization markers were determined by ELISA and qRT-PCR assays. Results Expression of miR-21 was significantly increased in NSCLC patients with RILI compared with control group, especially before or at 4 weeks after radiation therapy commenced. The miR-21 levels were highly correlated with IL-12, TNF-α, and IL-6 expressions and the severity of RILI. Animal based experiments demonstrated that BMSCs treatment had a remarkable effect on alleviating alveolitis in RILI rats, and miR-21 over-expression could enhance this effect significantly. Cell based experiments demonstrated that BMSCs notably inhibited M1 polarization of AMs and this inhibition is in a miR-21 dependent manner. Conclusions These results indicated that BMSCs could blocked the proinflammatory pathway of macrophage through miR-21 over-expression, thus could be a potential therapeutic strategy for RILI.
Collapse
Affiliation(s)
- Pengtao Bao
- Department of Respiration, the Eighth Medical Center of PLA General Hospital, Beijing 100091, China
| | - Weiguo Zhao
- Department of Respiration, the Eighth Medical Center of PLA General Hospital, Beijing 100091, China
| | - Mi Mou
- Department of Respiration, the Eighth Medical Center of PLA General Hospital, Beijing 100091, China
| | - Xiaofei Liu
- Department of Respiration, the Eighth Medical Center of PLA General Hospital, Beijing 100091, China
| |
Collapse
|