1
|
Liu D, Wang X, Han Y, Wang J, Sun Y, Hou Y, Wu Q, Zeng C, Ding X, Chang Y, Hu J, Huang X, Lu L. A donor PD-1 +CD8 + T SCM-like regulatory subset mobilized by G-CSF alleviates recipient acute graft-versus-host-disease. Signal Transduct Target Ther 2025; 10:120. [PMID: 40175340 PMCID: PMC11965471 DOI: 10.1038/s41392-025-02183-1] [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] [Received: 10/09/2024] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 04/04/2025] Open
Abstract
Donor selection determines the occurrence of acute graft-versus-host-disease (aGVHD) following allogeneic hematopoietic stem cell transplantation (allo-HSCT). To optimize the current clinical donor selection criteria and identify putative donor lymphocyte subsets associated with better recipient outcomes, we analyzed the peripheral CD4+ and CD8+ subsets in 80 granulocyte colony-stimulating factor (G-CSF) mobilized donors and examined the aGVHD incidence of the corresponding 80 haploidentical and identical allo-HSCT recipients. The G-CSF-induced expansion of subsets varied among donors. We discovered a novel PD-1+CD8+CD45RA+CCR7+ T lymphocyte subset in suitable donors that was significantly correlated with lower incidence of aGVHD and post-transplant anti-infection. The anti-aGVHD activity of this subset was confirmed in a validation cohort (n = 30). Single-cell RNA sequencing revealed that this T cell subset exhibited transcriptomic features of stem cell-like memory T cell (TSCM) with both Treg and Teff activities which indicated its dual functions in aGVHD inhibition and graft-versus-leukemia (GVL) effect. Intriguingly, upon G-CSF mobilization, the donor PD-1+CD8+ TSCM-like regulatory cells increased the PD-1 expression in a BCL6-dependent manner. Next, we showed that the mouse counterpart of this subset (PD-1+CD8+CD44-CD62L+) ameliorated aGVHD, and confirmed the existence of this subset in clinical recipients. In summary, we, for the first time, identified a novel donor peripheral T cell subset suppressing aGVHD while promoting the immune reconstitution of recipients. It may serve as an indicator for optimal haploidentical and identical donor selection. Importantly, the dual Treg and Teff function of these T cells makes it a promising treatment for not only aGVHD but also auto-immune diseases.
Collapse
Affiliation(s)
- Dan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Xue Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuheng Han
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wang
- Blood and Marrow Transplantation Center, Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yidan Sun
- Blood and Marrow Transplantation Center, Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yafei Hou
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Wu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cong Zeng
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuping Ding
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingjun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Jiong Hu
- Blood and Marrow Transplantation Center, Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing, China.
- Blood and Marrow Transplantation Center, Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Liming Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Reddy NR, Maachi H, Xiao Y, Simic MS, Yu W, Tonai Y, Cabanillas DA, Serrano-Wu E, Pauerstein PT, Tamaki W, Allen GM, Parent AV, Hebrok M, Lim WA. Engineering synthetic suppressor T cells that execute locally targeted immunoprotective programs. Science 2024; 386:eadl4793. [PMID: 39636990 PMCID: PMC11831968 DOI: 10.1126/science.adl4793] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 09/22/2024] [Indexed: 12/07/2024]
Abstract
Immune homeostasis requires a balance of inflammatory and suppressive activities. To design cells potentially useful for local immune suppression, we engineered conventional CD4+ T cells with synthetic Notch (synNotch) receptors driving antigen-triggered production of anti-inflammatory payloads. Screening a diverse library of suppression programs, we observed the strongest suppression of cytotoxic T cell attack by the production of both anti-inflammatory factors (interleukin-10, transforming growth factor-β1, programmed death ligand 1) and sinks for proinflammatory cytokines (interleukin-2 receptor subunit CD25). Engineered cells with bespoke regulatory programs protected tissues from immune attack without systemic suppression. Synthetic suppressor T cells protected transplanted beta cell organoids from cytotoxic T cells. They also protected specific tissues from unwanted chimeric antigen receptor (CAR) T cell cross-reaction. Synthetic suppressor T cells are a customizable platform to potentially treat autoimmune diseases, organ rejection, and CAR T cell toxicities with spatial precision.
Collapse
Affiliation(s)
- Nishith R. Reddy
- UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Hasna Maachi
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Yini Xiao
- UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Milos S. Simic
- UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Wei Yu
- UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Yurie Tonai
- UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Daniela A. Cabanillas
- UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Ella Serrano-Wu
- UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - Philip T. Pauerstein
- UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Whitney Tamaki
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA, USA
| | - Greg M. Allen
- UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Audrey V. Parent
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Matthias Hebrok
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Wendell A. Lim
- UCSF Cell Design Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
3
|
Bellotti P, Ladd Z, Leroy V, Su G, Sharma S, Hartman JB, Krebs J, Viscardi C, Maile R, Moldawer LL, Efron PA, Sharma AK, Upchurch GR. Resolvin D2/GPR18 signaling enhances monocytic myeloid-derived suppressor cell function to mitigate abdominal aortic aneurysm formation. FASEB J 2024; 38:e70067. [PMID: 39320982 PMCID: PMC11433576 DOI: 10.1096/fj.202400414rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
Abdominal aortic aneurysm (AAA) formation is a chronic vascular pathology characterized by inflammation, leukocyte infiltration, and vascular remodeling. The aim of this study was to delineate the protective role of Resolvin D2 (RvD2), a bioactive isoform of specialized pro-resolving lipid mediators, via G-protein-coupled receptor 18 (GPR18) receptor signaling in attenuating AAAs. Importantly, RvD2 and GPR18 levels were significantly decreased in aortic tissue of AAA patients compared with controls. Furthermore, using an established murine model of AAA in C57BL/6 (WT) mice, we observed that treatment with RvD2 significantly attenuated aortic diameter, pro-inflammatory cytokine production, immune cell infiltration (neutrophils and macrophages), elastic fiber disruption, and increased smooth muscle cell α-actin expression as well as increased TGF-β2 and IL-10 expressions compared to untreated mice. Moreover, the RvD2-mediated protection from vascular remodeling and AAA formation was blocked when mice were previously treated with siRNA for GPR18 signifying the importance of RvD2/GPR18 signaling in vascular inflammation. Mechanistically, RvD2-mediated protection significantly enhanced infiltration and activation of monocytic myeloid-derived suppressor cells (M-MDSCs) by increasing TGF-β2 and IL-10 secretions in a GPR18-dependent manner to attenuate aortic inflammation and vascular remodeling. Collectively, this study demonstrates that RvD2 treatment induces an expansion of myeloid-lineage committed progenitors, such as M-MDSCs, activates GPR18-dependent signaling to enhance TGF-β2 and IL-10 secretion, and mitigates SMC activation that contributes to resolution of aortic inflammation and remodeling during AAA formation.
Collapse
Affiliation(s)
- Paolo Bellotti
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Zachary Ladd
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Victoria Leroy
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Gang Su
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Shiven Sharma
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Joseph B. Hartman
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Jonathan Krebs
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Chelsea Viscardi
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Robert Maile
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Lyle L. Moldawer
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Phillip A. Efron
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | - Ashish K. Sharma
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, FL, 32610, USA
| | - Gilbert R. Upchurch
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
4
|
Leroy V, Manual Kollareth DJ, Tu Z, Valisno JAC, Woolet-Stockton M, Saha B, Emtiazjoo AM, Rackauskas M, Moldawer LL, Efron PA, Cai G, Atkinson C, Upchurch GR, Sharma AK. MerTK-dependent efferocytosis by monocytic-MDSCs mediates resolution of ischemia/reperfusion injury after lung transplant. JCI Insight 2024; 9:e179876. [PMID: 39172530 PMCID: PMC11466183 DOI: 10.1172/jci.insight.179876] [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: 01/29/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024] Open
Abstract
Lung transplantation (LTx) outcomes are impeded by ischemia/reperfusion injury (IRI) and subsequent chronic lung allograft dysfunction (CLAD). We examined the undefined role of receptor Mer tyrosine kinase (MerTK) on monocytic myeloid-derived suppressor cells (M-MDSCs) in efferocytosis to facilitate resolution of lung IRI. Single-cell RNA sequencing of lung tissue and bronchoalveolar lavage (BAL) from patients after LTx were analyzed. Murine lung hilar ligation and allogeneic orthotopic LTx models of IRI were used with BALB/c (WT), Cebpb-/- (MDSC-deficient), Mertk-/-, or MerTK-cleavage-resistant mice. A significant downregulation in MerTK-related efferocytosis genes in M-MDSC populations of patients with CLAD was observed compared with healthy individuals. In the murine IRI model, a significant increase in M-MDSCs, MerTK expression, and efferocytosis and attenuation of lung dysfunction was observed in WT mice during injury resolution that was absent in Cebpb-/- and Mertk-/- mice. Adoptive transfer of M-MDSCs in Cebpb-/- mice significantly attenuated lung dysfunction and inflammation. Additionally, in a murine orthotopic LTx model, increases in M-MDSCs were associated with resolution of lung IRI in the transplant recipients. In vitro studies demonstrated the ability of M-MDSCs to efferocytose apoptotic neutrophils in a MerTK-dependent manner. Our results suggest that MerTK-dependent efferocytosis by M-MDSCs can substantially contribute to the resolution of post-LTx IRI.
Collapse
Affiliation(s)
- Victoria Leroy
- Department of Surgery
- Department of Pharmacology and Therapeutics
| | | | - Zhenxiao Tu
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | | | - Biplab Saha
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Amir M. Emtiazjoo
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | | | | | - Guoshuai Cai
- Department of Surgery
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Carl Atkinson
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | - Ashish K. Sharma
- Department of Surgery
- Department of Pharmacology and Therapeutics
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
5
|
Yang F, Men R, Lv L, Zhou L, Deng Q, Wang X, Liu J, Yang L. Engaging natural regulatory myeloid cells to restrict T-cell hyperactivation-induced liver inflammation via extracellular vesicle-mediated purine metabolism regulation. Theranostics 2024; 14:4874-4893. [PMID: 39239508 PMCID: PMC11373623 DOI: 10.7150/thno.97427] [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: 04/17/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024] Open
Abstract
Rationale: Dysregulated T-cell immune response-mediated inflammation plays critical roles in the pathology of diverse liver diseases, but the underlying mechanism of liver immune homeostasis control and the specific therapies for limiting T-cell overactivation remain unclear. Methods: The metabolic changes in concanavalin A (ConA) mice and autoimmune hepatitis (AIH) patients and their associations with liver injury were analyzed. The expression of purine catabolism nucleases (e.g., CD39 and CD73) on liver cells and immune cells was assessed. The effects of MCregs and their extracellular vesicles (EVs) on CD4+ T-cell overactivation and the underlying mechanism were also explored. Results: Our findings revealed significant alterations in purine metabolism in ConA mice and AIH patients, which correlated with liver injury severity and therapeutic response. CD39 and CD73 were markedly upregulated on CD11b+Gr-1+ MCs under liver injury conditions. The naturally expanded CD39+CD73+Gr-1highCD11b+ MCreg subset during early liver injury effectively suppressed CD4+ T-cell hyperactivation and liver injury both in vitro and in vivo. Mechanistically, MCregs released CD73high EVs, which converted extracellular AMP to immunosuppressive metabolites (e.g., adenosine and inosine), activating the cAMP pathway and inhibiting glycolysis and cytokine secretion in activated CD4+ T cells. Conclusions: This study provides insights into the mechanism controlling immune homeostasis during the early liver injury phase and highlights that MCreg or MCreg-EV therapy may be a specific strategy for preventing diverse liver diseases induced by T-cell overactivation.
Collapse
Affiliation(s)
- Fan Yang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ruoting Men
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Linling Lv
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Leyu Zhou
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Qiaoyu Deng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xianglin Wang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Li Yang
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
6
|
Wang H, Guo Y, Zhang X, Zhou X. Bexarotene Induce Differentiation of Myeloid-Derived Suppressor Cells through Arg-1 Signalling Pathway. Transplant Proc 2024; 56:1469-1477. [PMID: 38981763 DOI: 10.1016/j.transproceed.2024.03.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 02/21/2024] [Accepted: 03/18/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Cellular therapy has emerged as a promising strategy to minimize the use of conventional immunosuppressive drugs and ultimately induce long-term graft survival. Myeloid-derived suppressor cells (MDSCs) can be used for immunosuppressive treatment of solid organ transplants. METHODS Granular macrophage colony-stimulating factor (GM-CSF) and bexarotene, an X receptor-selective retinoid, were used for in vitro MDSC induction. Cell phenotypes were detected using flow cytometry, while mRNA was detected via real-time PCR. A mouse skin transplantation model was used to verify the inhibitory effects of this treatment. RESULTS The combination of GM-CSF and bexarotene-induced MDSC differentiation. MDSCs induce immune tolerance by inhibiting T-cell proliferation, influencing cytokine secretion, and inducing T-cell transformation into Treg cells. Combination treatment significantly up-regulated Arg-1 expression in MDSCs. The Arg-1 inhibitor nor-NOHA neutralized the immunosuppressive activity of MDSCs, suggesting the involvement of Arg-1 in MDSC-mediated immunosuppression. GM-CSF and bexarotene-induced MDSCs prolong graft survival in mouse skin transplants, exhibiting in vivo immunosuppressive effects. CONCLUSIONS A new method for inducing MDSCs is presented. The combination of GM-CSF and bexarotene induces MDSCs with remarkable regulatory functions. Adoptive transfer of the induced MDSCs extended allograft survival. These results suggest that MDSCs can potentially be used in future clinical transplants to inhibit rejection, reduce adverse events, and induce operative tolerance.
Collapse
Affiliation(s)
- Haozhou Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yinan Guo
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiaodong Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaoguang Zhou
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
van Geffen C, Lange T, Kolahian S. Myeloid-derived suppressor cells in influenza virus-induced asthma exacerbation. Front Immunol 2024; 15:1342497. [PMID: 38694499 PMCID: PMC11061804 DOI: 10.3389/fimmu.2024.1342497] [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: 11/21/2023] [Accepted: 04/01/2024] [Indexed: 05/04/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a phenotypically heterogenous group of cells that potently suppress the immune response. A growing body of evidence supports the important role of MDSCs in a variety of lung diseases, such as asthma. However, the role of MDSCs in asthma exacerbation has so far not been investigated. Here, we studied the role of MDSCs in a murine model of influenza virus-induced asthma exacerbation. BALB/c mice were exposed to house dust mite (HDM) three times a week for a total of five weeks to induce a chronic asthmatic phenotype, which was exacerbated by additional exposure to the A/Hamburg/5/2009 hemagglutinin 1 neuraminidase 1 (H1N1) influenza virus. Induction of lung inflammatory features, production of T helper (Th) 1- and Th2- associated inflammatory cytokines in the lavage fluid and an increased airway hyper-responsiveness were observed, establishing the asthma exacerbation model. The number and activity of pulmonary M-MDSCs increased in exacerbated asthmatic mice compared to non-exacerbated asthmatic mice. Furthermore, depletion of MDSCs aggravated airway hyper-responsiveness in exacerbated asthmatic mice. These findings further denote the role of MDSCs in asthma and provide some of the first evidence supporting a potential important role of MDSCs in asthma exacerbation.
Collapse
Affiliation(s)
- Chiel van Geffen
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), The Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, University of Tübingen, Tübingen, Germany
| | - Tim Lange
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), The Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
| | - Saeed Kolahian
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL), The Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, Marburg, Germany
- Small Animal Imaging Core Facility, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| |
Collapse
|
8
|
Bellotti P, Ladd Z, Leroy V, Su G, Sharma S, Hartman JB, Krebs J, Viscardi C, Maile R, Moldawer LL, Efron P, Sharma AK, Upchurch GR. Resolvin D2/GPR18 signaling enhances monocytic myeloid-derived suppressor cell function to mitigate abdominal aortic aneurysm formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581672. [PMID: 38464077 PMCID: PMC10925138 DOI: 10.1101/2024.02.23.581672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Abdominal aortic aneurysm (AAA) formation is a chronic vascular pathology characterized by inflammation, leukocyte infiltration and vascular remodeling. The aim of this study was to delineate the protective role of Resolvin D2 (RvD2), a bioactive isoform of specialized proresolving lipid mediators, via G-protein coupled receptor 18 (GPR18) receptor signaling in attenuating AAAs. Importantly, RvD2 and GPR18 levels were significantly decreased in aortic tissue of AAA patients compared with controls. Furthermore, using an established murine model of AAA in C57BL/6 (WT) mice, we observed that treatment with RvD2 significantly attenuated aortic diameter, pro-inflammatory cytokine production, immune cell infiltration (neutrophils and macrophages), elastic fiber disruption and increased smooth muscle cell α-actin expression as well as increased TGF-β2 and IL-10 expressions compared to untreated mice. Moreover, the RvD2-mediated protection from vascular remodeling and AAA formation was blocked when mice were previously treated with siRNA for GPR18 signifying the importance of RvD2/GPR18 signaling in vascular inflammation. Mechanistically, RvD2-mediated protection significantly enhanced infiltration and activation of monocytic myeloid-derived suppressor cells (M-MDSCs) by increasing TGF-β2 and IL-10 secretions that mitigated smooth muscle cell activation in a GPR18-dependent manner to attenuate aortic inflammation and vascular remodeling via this intercellular crosstalk. Collectively, this study demonstrates RvD2 treatment induces an expansion of myeloid-lineage committed progenitors, such as M-MDSCs, and activates GPR18-dependent signaling to enhance TGF-β2 and IL-10 secretion that contributes to resolution of aortic inflammation and remodeling during AAA formation.
Collapse
|
9
|
Leroy V, Manual Kollareth DJ, Tu Z, Valisno JAC, Woolet-Stockton M, Saha B, Emtiazjoo AM, Rackauskas M, Moldawer LL, Efron PA, Cai G, Atkinson C, Upchurch GR, Sharma AK. MerTK-dependent efferocytosis by monocytic-MDSCs mediates resolution of post-lung transplant injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576261. [PMID: 38328174 PMCID: PMC10849528 DOI: 10.1101/2024.01.18.576261] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Rationale Patients with end stage lung diseases require lung transplantation (LTx) that can be impeded by ischemia-reperfusion injury (IRI) leading to subsequent chronic lung allograft dysfunction (CLAD) and inadequate outcomes. Objectives We examined the undefined role of MerTK (receptor Mer tyrosine kinase) on monocytic myeloid-derived suppressor cells (M-MDSCs) in efferocytosis (phagocytosis of apoptotic cells) to facilitate resolution of lung IRI. Methods Single-cell RNA sequencing of lung tissue and BAL from post-LTx patients was analyzed. Murine lung hilar ligation and allogeneic orthotopic LTx models of IRI were used with Balb/c (WT), cebpb -/- (MDSC-deficient), Mertk -/- or MerTK-CR (cleavage resistant) mice. Lung function, IRI (inflammatory cytokine and myeloperoxidase expression, immunohistology for neutrophil infiltration), and flow cytometry of lung tissue for efferocytosis of apoptotic neutrophils were assessed in mice. Measurements and Main Results A significant downregulation in MerTK-related efferocytosis genes in M-MDSC populations of CLAD patients compared to healthy subjects was observed. In the murine IRI model, significant increase in M-MDSCs, MerTK expression and efferocytosis was observed in WT mice during resolution phase that was absent in cebpb -/- Land Mertk -/- mice. Adoptive transfer of M-MDSCs in cebpb -/- mice significantly attenuated lung dysfunction, and inflammation leading to resolution of IRI. Additionally, in a preclinical murine orthotopic LTx model, increases in M-MDSCs were associated with resolution of lung IRI in the transplant recipients. In vitro studies demonstrated the ability of M-MDSCs to efferocytose apoptotic neutrophils in a MerTK-dependent manner. Conclusions Our results suggest that MerTK-dependent efferocytosis by M-MDSCs can significantly contribute to the resolution of post-LTx IRI.
Collapse
|
10
|
Hua S, Wang S, Cai J, Wu L, Cao Y. Myeloid-derived suppressor cells: Are they involved in gestational diabetes mellitus? Am J Reprod Immunol 2023:e13711. [PMID: 37157925 DOI: 10.1111/aji.13711] [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: 03/21/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/10/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is currently the most common metabolic complication during pregnancy, with an increasing prevalence worldwide. Maternal immune dysregulation might be partly responsible for the pathophysiology of GDM. Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of cells, emerging as a new immune regulator with potent immunosuppressive capacity. Although the fate and function of these cells were primarily described in pathological conditions such as cancer and infection, accumulating evidences have spotlighted their beneficial roles in homeostasis and physiological conditions. Recently, several studies have explored the roles of MDSCs in the diabetic microenvironment. However, the fate and function of these cells in GDM are still unknown. The current review summarized the existing knowledges about MDSCs and their potential roles in diabetes during pregnancy in an attempt to highlight our current understanding of GDM-related immune dysregulation and identify areas where further study is required.
Collapse
Affiliation(s)
- Siyu Hua
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Shanshan Wang
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinyang Cai
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lamei Wu
- Department of Perinatal Healthcare, Huai'an District Maternity and Child Health Hospital, Huai'an, Jiangsu, China
| | - Yan Cao
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
11
|
Ostrand-Rosenberg S, Lamb TJ, Pawelec G. Here, There, and Everywhere: Myeloid-Derived Suppressor Cells in Immunology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1183-1197. [PMID: 37068300 PMCID: PMC10111205 DOI: 10.4049/jimmunol.2200914] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/06/2023] [Indexed: 04/19/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) were initially identified in humans and mice with cancer where they profoundly suppress T cell- and NK cell-mediated antitumor immunity. Inflammation is a central feature of many pathologies and normal physiological conditions and is the dominant driving force for the accumulation and function of MDSCs. Therefore, MDSCs are present in conditions where inflammation is present. Although MDSCs are detrimental in cancer and conditions where cellular immunity is desirable, they are beneficial in settings where cellular immunity is hyperactive. Because MDSCs can be generated ex vivo, they are being exploited as therapeutic agents to reduce damaging cellular immunity. In this review, we discuss the detrimental and beneficial roles of MDSCs in disease settings such as bacterial, viral, and parasitic infections, sepsis, obesity, trauma, stress, autoimmunity, transplantation and graft-versus-host disease, and normal physiological settings, including pregnancy and neonates as well as aging. The impact of MDSCs on vaccination is also discussed.
Collapse
Affiliation(s)
- Suzanne Ostrand-Rosenberg
- Division of Microbiology and Immunology, Department of Pathology, University of Utah 84112, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Tracey J. Lamb
- Division of Microbiology and Immunology, Department of Pathology, University of Utah 84112, Salt Lake City, UT
| | - Graham Pawelec
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany, and Health Sciences North Research Institute, Sudbury, ON, Canada
| |
Collapse
|
12
|
Shi ZY, Yang C, Lu LY, Lin CX, Liang S, Li G, Zhou HM, Zheng JM. Inhibition of hexokinase 2 with 3-BrPA promotes MDSCs differentiation and immunosuppressive function. Cell Immunol 2023; 385:104688. [PMID: 36774675 DOI: 10.1016/j.cellimm.2023.104688] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/08/2022] [Accepted: 02/03/2023] [Indexed: 02/08/2023]
Abstract
The adoptive transfer of ex vivo generated myeloid-derived suppressor cells (MDSCs) may be a promising therapeutic strategy for preventing allograft rejection after solid organ transplantation. Currently, the precise role of immune-metabolic pathways in the differentiation and function of MDSCs is not fully understood. Hexokinase 2 (HK2) is an isoform of hexokinase and is a key enzyme involved in the increased aerobic glycolysis of different immune cells during their activation and function. Here, we demonstrate that the addition of HK2 inhibitor 3-Bromopyruvic acid (3-BrPA) into traditional MDSCs induction system in vitro significantly promoted MDSCs production and enhanced their immunosuppressive function. Treatment with 3-BrPA increased the expression of MDSC-related immunosuppressive molecules, such as iNOS, Arg1, and CXCR2. Moreover, the adoptive transfer of 3-BrPA-treated MDSCs significantly prolonged the survival time of mouse heart allografts. This study provides a novel strategy to solve the problems of harvesting enough autologous cells for MDSC production from sick patients, and producing functionally enhanced MDSCs for preventing graft rejection and inducing tolerance.
Collapse
Affiliation(s)
- Zhan-Yue Shi
- Department of Cardiothoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chao Yang
- Department of Organ Transplantation, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510000, China
| | - Liu-Yi Lu
- Department of Cardiothoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Research Center of Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Can-Xiang Lin
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shi Liang
- Department of Cardiothoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Gen Li
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Min Zhou
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun-Meng Zheng
- Department of Cardiothoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
13
|
Local and Systemic Injections of Human Cord Blood Myeloid-Derived Suppressor Cells to Prevent Graft Rejection in Corneal Transplantation. Biomedicines 2022; 10:biomedicines10123223. [PMID: 36551981 PMCID: PMC9776015 DOI: 10.3390/biomedicines10123223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are therapeutic agents to prevent graft rejection in organ transplants by modulating inflammation. Herein, the immunosuppressive effect of human cord blood MDSCs on corneal allograft models was confirmed. CB-MDSCs were locally (subconjuctival, 5 × 105) or systemically (intravenous, 1 × 106) injected twice on days 0 and 7. A corneal transplantation model was established using C57BL/6 and BALB/c mice, and corneal graft opacity was measured to evaluate graft rejection up to 6 weeks. Results showed that graft survival in the MDSCs groups increased compared to vehicle groups after 42 days. Systemic and local MDSC administration inhibited the maturation (MHC-IIhi CD11c+) of dendritic cells (DCs) and the differentiation of interferon γ+ CD4+ Th1 in draining lymph nodes (LNs). However, vehicle groups increased the infiltration of CD3+ T cells and F4/80+ macrophages and produced prominent neovascular and lymphatic vessels into the graft site with increased mRNA expression of VEGF-A/C and VEGFR-1/R-3. Local MDSCs administration showed prominent anti-angiogenic/anti-lymphangiogenic effects even at lower MDSCs doses. Thus, CB-MDSCs could relatively suppress the infiltration of pathological T cells/macrophages into the corneas and the migration of mature DCs into draining LNs Therefore, ocular and systemic MDSCs administration showed therapeutic potential for preventing corneal allograft rejection.
Collapse
|
14
|
Yu K, Yu C, Jiao L, Miao K, Ni L, Rao X, Zhou L, Zhao C. The Function and Therapeutic Implications of TNF Signaling in MDSCs. Biomolecules 2022; 12:1627. [PMID: 36358977 PMCID: PMC9687347 DOI: 10.3390/biom12111627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 09/27/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of immature and heterogeneous myeloid cells with immunosuppressive functions. MDSCs play important roles in the pathogenesis of cancer, chronic inflammatory diseases, and many autoimmune disorders. The accumulation and activation of MDSCs can be regulated by tumor necrosis factor α (TNF-α). In this review, we summarize the roles played by TNF-α in the recruitment, immunosuppressive functions, and chemotaxis of MDSCs, and discuss the potential therapeutic effects of TNF-α upon these cells in tumor growth and some inflammatory disorders.
Collapse
Affiliation(s)
- Kun Yu
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chengxin Yu
- GI Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Liping Jiao
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Kun Miao
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Ni
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoquan Rao
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ling Zhou
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chunxia Zhao
- Division of Cardiology, Departments of Internal Medicine and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
15
|
Radojević D, Bekić M, Gruden-Movsesijan A, Ilić N, Dinić M, Bisenić A, Golić N, Vučević D, Đokić J, Tomić S. Myeloid-derived suppressor cells prevent disruption of the gut barrier, preserve microbiota composition, and potentiate immunoregulatory pathways in a rat model of experimental autoimmune encephalomyelitis. Gut Microbes 2022; 14:2127455. [PMID: 36184742 PMCID: PMC9543149 DOI: 10.1080/19490976.2022.2127455] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Over-activated myeloid cells and disturbance in gut microbiota composition are critical factors contributing to the pathogenesis of Multiple Sclerosis (MS). Myeloid-derived suppressor cells (MDSCs) emerged as promising regulators of chronic inflammatory diseases, including autoimmune diseases. However, it remained unclear whether MDSCs display any therapeutic potential in MS, and how this therapy modulates gut microbiota composition. Here, we assessed the potential of in vitro generated bone marrow-derived MDSCs to ameliorate experimental autoimmune encephalomyelitis (EAE) in Dark Agouti rats and investigated how their application associates with the changes in gut microbiota composition. MDSCs differentiated with prostaglandin (PG)E2 (MDSC-PGE2) and control MDSCs (differentiated without PGE2) displayed strong immunosuppressive properties in vitro, but only MDSC-PGE2 significantly ameliorated EAE symptoms. This effect correlated with a reduced infiltration of Th17 and IFN-γ-producing NK cells, and an increased proportion of regulatory T cells in the CNS and spleen. Importantly, both MDSCs and MDSC-PGE2 prevented EAE-induced reduction of gut microbiota diversity, but only MDSC-PGE2 prevented the extensive alterations in gut microbiota composition following their early migration into Payer's patches and mesenteric lymph nodes. This phenomenon was related to the significant enrichment of gut microbial taxa with potential immunoregulatory properties, as well as higher levels of butyrate, propionate, and putrescine in feces. This study provides new insights into the host-microbiota interactions in EAE, suggesting that activated MDSCs could be potentially used as an efficient therapy for acute phases of MS. Considering a significant association between the efficacy of MDSC-PGE2 and gut microbiota composition, our findings also provide a rationale for further exploring the specific microbial metabolites in MS therapy.
Collapse
Affiliation(s)
- Dušan Radojević
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marina Bekić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Alisa Gruden-Movsesijan
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Nataša Ilić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia
| | - Miroslav Dinić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Aleksandar Bisenić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Nataša Golić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Dragana Vučević
- Medical Faculty of the Military Medical Academy, University of Defense in Belgrade, Belgrade, Serbia
| | - Jelena Đokić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia,CONTACT Jelena Đokić Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, 111042 Belgrade, Vojvode Stepe 444a, Belgrade, Serbia
| | - Sergej Tomić
- Department for Immunology and Immunoparasitology, Institute for the Application of Nuclear Energy, University of Belgrade, Belgrade, Serbia,Sergej Tomić Institute for the Application of Nuclear Energy, 11080 Belgrade, Banatska 31b, Belgrade, Serbia
| |
Collapse
|
16
|
Zuo H, Tao J, Wang M, Xie X, Sun M. A novel immunochemotherapy based on immunogenicity-activated and immunosuppression-reversed biomimetic nanoparticles. RSC Adv 2022; 12:28104-28112. [PMID: 36320259 PMCID: PMC9527569 DOI: 10.1039/d2ra04326b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/25/2022] [Indexed: 01/24/2023] Open
Abstract
Studies show that infiltrated myeloid-derived suppressor cells (MDSCs) are vital in the immunosuppressive tumor microenvironment and account for lymphoma refractoriness and recurrence. Here, we developed a biomimetic nanoplatform (PM-PLGA-DOX/GEM) in which platelet membranes (PM) wrap PLGA nanoparticles co-loaded with doxorubicin (DOX) and gemcitabine (GEM). PM-PLGA-DOX/GEM would accumulate in tumor tissues because of the enhanced permeation and retention (EPR) effect and the tumor cell-induced platelet aggregation (TCIPA) effect. GEM could eliminate the MDSCs in tumor tissues, thereby reversing the immunosuppressive tumor microenvironment. Furthermore, DOX could invoke the immunogenic cell death (ICD) of lymphoma cells. Consequently, numerous T cells were recruited and activated to improve the therapeutic effects. This study will offer a potential platform for clinical treatment of lymphoma and other solid tumors.
Collapse
Affiliation(s)
- Huaqin Zuo
- Department of Hematology, Northern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouJiangsu225001P. R. China
| | - Junxian Tao
- Department of Endocrinology, Northern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouJiangsu225001P. R. China
| | - Manli Wang
- Graduate School of Dalian Medical UniversityDalianLiaoning116044P. R. China
| | - Xiaoyan Xie
- Department of Hematology, Northern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouJiangsu225001P. R. China
| | - Mei Sun
- Department of Hematology, Northern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouJiangsu225001P. R. China
| |
Collapse
|
17
|
Schroeter A, Roesel MJ, Matsunaga T, Xiao Y, Zhou H, Tullius SG. Aging Affects the Role of Myeloid-Derived Suppressor Cells in Alloimmunity. Front Immunol 2022; 13:917972. [PMID: 35874716 PMCID: PMC9296838 DOI: 10.3389/fimmu.2022.917972] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are defined as a group of myeloid cells with potent immunoregulatory functions that have been shown to be involved in a variety of immune-related diseases including infections, autoimmune disorders, and cancer. In organ transplantation, MDSC promote tolerance by modifying adaptive immune responses. With aging, however, substantial changes occur that affect immune functions and impact alloimmunity. Since the vast majority of transplant patients are elderly, age-specific modifications of MDSC are of relevance. Furthermore, understanding age-associated changes in MDSC may lead to improved therapeutic strategies. Here, we provide a comprehensive update on the effects of aging on MDSC and discuss potential consequences on alloimmunity.
Collapse
Affiliation(s)
- Andreas Schroeter
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Regenerative Medicine and Experimental Surgery, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Maximilian J. Roesel
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Institute of Medical Immunology, Charite Universitaetsmedizin Berlin, Berlin, Germany
| | - Tomohisa Matsunaga
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Urology, Osaka Medical and Pharmaceutical University, Takatsuki City, Japan
| | - Yao Xiao
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Hao Zhou
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Stefan G. Tullius
- Transplant Surgery Research Laboratory and Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
18
|
Park Y, Kwok SK. Recent Advances in Cell Therapeutics for Systemic Autoimmune Diseases. Immune Netw 2022; 22:e10. [PMID: 35291648 PMCID: PMC8901702 DOI: 10.4110/in.2022.22.e10] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/03/2022] Open
Abstract
Systemic autoimmune diseases arise from loss of self-tolerance and immune homeostasis between effector and regulator functions. There are many therapeutic modalities for autoimmune diseases ranging from conventional disease-modifying anti-rheumatic drugs and immunosuppressants exerting nonspecific immune suppression to targeted agents including biologic agents and small molecule inhibitors aiming at specific cytokines and intracellular signal pathways. However, such current therapeutic strategies can rarely induce recovery of immune tolerance in autoimmune disease patients. To overcome limitations of conventional treatment modalities, novel approaches using specific cell populations with immune-regulatory properties have been attempted to attenuate autoimmunity. Recently progressed biotechnologies enable sufficient in vitro expansion and proper manipulation of such ‘tolerogenic’ cell populations to be considered for clinical application. We introduce 3 representative cell types with immunosuppressive features, including mesenchymal stromal cells, Tregs, and myeloid-derived suppressor cells. Their cellular definitions, characteristics, mechanisms of immune regulation, and recent data about preclinical and clinical studies in systemic autoimmune diseases are reviewed here. Challenges and limitations of each cell therapy are also addressed.
Collapse
Affiliation(s)
- Youngjae Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
19
|
Lin ECY, Chen SW, Chen LK, Lin TA, Wu YX, Juan CC, Chang YI. Glucosamine Interferes With Myelopoiesis and Enhances the Immunosuppressive Activity of Myeloid-Derived Suppressor Cells. Front Nutr 2021; 8:762363. [PMID: 34901113 PMCID: PMC8660085 DOI: 10.3389/fnut.2021.762363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/19/2021] [Indexed: 12/16/2022] Open
Abstract
Glucosamine (GlcN) is the most widely consumed dietary supplement and exhibits anti-inflammatory effects. However, the influence of GlcN on immune cell generation and function is largely unclear. In this study, GlcN was delivered into mice to examine its biological function in hematopoiesis. We found that GlcN promoted the production of immature myeloid cells, known as myeloid-derived suppressor cells (MDSCs), both in vivo and in vitro. Additionally, GlcN upregulated the expression of glucose transporter 1 in hematopoietic stem and progenitor cells (HSPCs), influenced HSPC functions, and downregulated key genes involved in myelopoiesis. Furthermore, GlcN increased the expression of arginase 1 and inducible nitric oxide synthase to produce high levels of reactive oxygen species, which was regulated by the STAT3 and ERK1/2 pathways, to increase the immunosuppressive ability of MDSCs. We revealed a novel role for GlcN in myelopoiesis and MDSC activity involving a potential link between GlcN and immune system, as well as the new therapeutic benefit.
Collapse
Affiliation(s)
- Eric Chang-Yi Lin
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Shuoh-Wen Chen
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Luen-Kui Chen
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Ting-An Lin
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan.,Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Yu-Xuan Wu
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Chi-Chang Juan
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Yuan-I Chang
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| |
Collapse
|
20
|
Ling Z, Yang C, Tan J, Dou C, Chen Y. Beyond immunosuppressive effects: dual roles of myeloid-derived suppressor cells in bone-related diseases. Cell Mol Life Sci 2021; 78:7161-7183. [PMID: 34635950 PMCID: PMC11072300 DOI: 10.1007/s00018-021-03966-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/11/2021] [Accepted: 09/29/2021] [Indexed: 02/08/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells (IMCs) with immunosuppressive functions, whereas IMCs originally differentiate into granulocytes, macrophages, and dendritic cells (DCs) to participate in innate immunity under steady-state conditions. At present, difficulties remain in identifying MDSCs due to lacking of specific biomarkers. To make identification of MDSCs accurately, it also needs to be determined whether having immunosuppressive functions. MDSCs play crucial roles in anti-tumor, angiogenesis, and metastasis. Meanwhile, MDSCs could make close interaction with osteoclasts, osteoblasts, chondrocytes, and other stromal cells within microenvironment of bone and joint, and thereby contributing to poor prognosis of bone-related diseases such as cancer-related bone metastasis, osteosarcoma (OS), rheumatoid arthritis (RA), osteoarthritis (OA), and orthopedic trauma. In addition, MDSCs have been shown to participate in the procedure of bone repair. In this review, we have summarized the function of MDSCs in cancer-related bone metastasis, the interaction with stromal cells within the bone microenvironment as well as joint microenvironment, and the critical role of MDSCs in bone repair. Besides, the promising value of MDSCs in the treatment for bone-related diseases is also well discussed.
Collapse
Affiliation(s)
- Zhiguo Ling
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chuan Yang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiulin Tan
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ce Dou
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yueqi Chen
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
21
|
Myeloid-derived suppressor cells: Multi-talented immune suppressive cells that can be either helpful or harmful. Cell Immunol 2021; 365:104374. [PMID: 34038759 DOI: 10.1016/j.cellimm.2021.104374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|