1
|
Yang S, Penna V, Lavine KJ. Functional diversity of cardiac macrophages in health and disease. Nat Rev Cardiol 2025; 22:431-442. [PMID: 39743564 DOI: 10.1038/s41569-024-01109-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Macrophages make up a substantial portion of the stromal compartment of the heart in health and disease. In the past decade, the origins of these cardiac macrophages have been established as two broad populations derived from either embryonic or definitive haematopoiesis and that can be distinguished by the expression of CC-motif chemokine receptor 2 (CCR2). These cardiac macrophage populations are transcriptionally distinct and have differing cell surface markers and divergent roles in cardiac homeostasis and disease. Embryonic-derived CCR2- macrophages are a tissue-resident population that participates in tissue development, repair and maintenance, whereas CCR2+ macrophages are derived from definitive haematopoiesis and contribute to inflammation and tissue damage. Studies from the past 5 years have leveraged single-cell RNA sequencing technologies to expand our understanding of cardiac macrophage diversity, particularly of the monocyte-derived macrophage populations that reside in the injured and diseased heart. Emerging technologies in spatial transcriptomics have enabled the identification of distinct disease-associated cellular neighbourhoods consisting of macrophages, other immune cells and fibroblasts, highlighting the involvement of macrophages in cell-cell communication. Together, these discoveries lend new insights into the role of specific macrophage populations in the pathogenesis of cardiac disease, which can pave the way for the identification of new therapeutic targets and the development of diagnostic tools. In this Review, we discuss the developmental origin of cardiac macrophages and describe newly identified cell states and associated cellular neighbourhoods in the steady state and injury settings. We also discuss various contributions and effector functions of cardiac macrophages in homeostasis and disease.
Collapse
Affiliation(s)
- Steven Yang
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Vinay Penna
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
2
|
Borges TJ, Lima K, Gassen RB, Liu K, Ganchiku Y, Ribas GT, Liao M, Goncalves JIB, Lape IT, Rosales IA, Zhao Y, Hui E, Fairchild RL, LeGuern C, Bonorino C, Calderwood SK, Madsen JC, Riella LV. The inhibitory receptor Siglec-E controls antigen-presenting cell activation and T cell-mediated transplant rejection. Sci Transl Med 2025; 17:eads2694. [PMID: 40333992 DOI: 10.1126/scitranslmed.ads2694] [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: 08/05/2024] [Revised: 12/16/2024] [Accepted: 03/26/2025] [Indexed: 05/09/2025]
Abstract
After transplantation, inflammation and tissue injury release danger signals that activate myeloid cells, driving adaptive immune responses and acute rejection. Current immunosuppressants primarily target T cells but inadequately control innate immunity. Regulatory signals controlling innate responses in transplantation remain elusive. The sialic acid-binding immunoglobulin-like lectin-E (Siglec-E, or SigE) binds sialylated ligands to suppress inflammation. In mouse heart transplants, SigE is up-regulated in graft-infiltrating myeloid cells, including dendritic cells (DCs). SigE deficiency in recipients, but not donors, accelerates acute rejection by enhancing DC activation, nuclear factor κB (NF-κB) signaling, and tumor necrosis factor-α (TNF-α) production, thereby boosting alloreactive T cell responses. Conversely, SigE overexpression on DCs reduces activation by danger signals and their T cell allostimulatory capacity. The human homologs Siglecs-7 and -9 were up-regulated in rejecting allograft biopsies, and their higher expression correlated with improved allograft survival. Thus, SigE/7/9 is a crucial inhibitory receptor controlling antigen-presenting cell activation and T cell-mediated transplant rejection, offering therapeutic potential.
Collapse
Affiliation(s)
- Thiago J Borges
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Karina Lima
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Rodrigo B Gassen
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Kaifeng Liu
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Yoshikazu Ganchiku
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Guilherme T Ribas
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Minxue Liao
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Joao I B Goncalves
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Isadora T Lape
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Ivy A Rosales
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yunlong Zhao
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Enfu Hui
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert L Fairchild
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH 44196, USA
- Transplant Center, Cleveland Clinic, Cleveland, OH 44196, USA
| | - Christian LeGuern
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Cristina Bonorino
- Immunotherapy Laboratory - (LAIT) - Department of Basic Health Sciences of Federal University of Health Sciences of Porto Alegre, UFCSPA, Porto Alegre 90050-170, Brazil
| | - Stuart K Calderwood
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Joren C Madsen
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Leonardo V Riella
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
3
|
Phi LTH, Cheng Y, Funakoshi Y, Bertucci F, Finetti P, Van Laere SJ, Zou F, Long JP, Ogata S, Krishnamurthy S, Reuben JM, Foulks JM, Warner SL, Rosenbluth JM, Sood AK, Tripathy D, Ueno NT, Wang X. AXL promotes inflammatory breast cancer progression by regulating immunosuppressive macrophage polarization. Breast Cancer Res 2025; 27:70. [PMID: 40329335 PMCID: PMC12057249 DOI: 10.1186/s13058-025-02015-8] [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: 09/24/2024] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are key promoters of inflammatory breast cancer (IBC), the most aggressive form of breast cancer. The receptor tyrosine kinase AXL is highly expressed in various cancer types, including IBC, but its role in TAMs remains unexplored. METHODS We examined the effects of AXL inhibitor TP-0903 on tumor growth and tumor microenvironment (TME) component M2 macrophages (CD206+) in IBC and triple-negative breast cancer mouse models using flow cytometry and immunohistochemical staining. Additionally, we knocked out AXL expression in human THP-1 monocytes and evaluated the effect of AXL signaling on immunosuppressive M2 macrophage polarization and IBC cell growth and migration. We then investigated the underlying mechanisms through RNA sequencing analysis. Last, we performed CIBERSORT deconvolution to analyze the association between AXL expression and tumor-infiltrating immune cell types in tumor samples from the Inflammatory Breast Cancer International Consortium. RESULTS We found that inhibiting the AXL pathway significantly reduced IBC tumor growth and decreased CD206+ macrophage populations within tumors. Mechanistically, our in vitro data showed that AXL promoted M2 macrophage polarization and enhanced the secretion of immunosuppressive chemokines, including CCL20, CCL26, and epiregulin, via the transcription factor STAT6 and thereby accelerated IBC cell growth and migration. RNA sequencing analysis further indicated that AXL signaling in immunosuppressive M2 macrophages regulated the expression of molecules and cytokines, contributing to an immunosuppressive TME in IBC. Moreover, high AXL expression was correlated with larger populations of immunosuppressive immune cells but smaller populations of immunoactive immune cells in tissues from patients with IBC. CONCLUSIONS AXL signaling promotes IBC growth by inducing M2 macrophage polarization and driving the secretion of immunosuppressive molecules and cytokines via STAT6 signaling, thereby contributing to an immunosuppressive TME. Collectively, these findings highlight the potential of targeting AXL signaling as a novel therapeutic approach for IBC that warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Lan T H Phi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Yating Cheng
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yohei Funakoshi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francois Bertucci
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
- Département d'Oncologie Médicale, Institut Paoli-Calmettes, Marseille, France
| | - Pascal Finetti
- Laboratoire d'Oncologie Prédictive, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille Université, Marseille, France
| | - Steven J Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Fang Zou
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James P Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suguru Ogata
- Cancer Biology Program, University of Hawai'i Cancer Center, Honolulu, HI, USA
- University of Hawai'i Inflammatory Breast Cancer Clinic and Research Program, Honolulu, HI, USA
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Jennifer M Rosenbluth
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cancer Biology Program, University of Hawai'i Cancer Center, Honolulu, HI, USA.
- University of Hawai'i Inflammatory Breast Cancer Clinic and Research Program, Honolulu, HI, USA.
| | - Xiaoping Wang
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Cancer Biology Program, University of Hawai'i Cancer Center, Honolulu, HI, USA.
- University of Hawai'i Inflammatory Breast Cancer Clinic and Research Program, Honolulu, HI, USA.
| |
Collapse
|
4
|
Zhu Y, Zheng X, Chen C, Xu Y, Fan Y, Tang Z, Zeng Y, Feng C, Zhang H, Chen X, Qi Z, Li T, Peng L, Lin M, Chen W, Peng F, Jiang X, Dai H. Single-cell RNA sequencing revealed the immunophenotypic features of macrophages in cardiac transplants and uncovered Lgals9 promoted their polarization toward the M2b subtype. J Leukoc Biol 2025; 117:qiaf009. [PMID: 39835675 DOI: 10.1093/jleuko/qiaf009] [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: 08/28/2024] [Revised: 10/30/2024] [Accepted: 01/20/2025] [Indexed: 01/22/2025] Open
Abstract
Macrophages (MAC) play a crucial role in the immune response during allograft rejection in organ transplantation. Therefore, our study aimed to explore the genomic features of MAC in mouse heart transplants and use single-cell RNA sequencing to investigate Galectin-9 (Gal-9 and Lgals9), a lectin that can mediate the activation and differentiation of immune cells through ligand-receptor interactions, and the effects of its regulation in transplantation. We discovered a new subset of MAC called "Myoz2+ MAC," which specifically expressed genes related to myocardial contraction. We identified a distinct differentiation trajectory and process for the Saa3+ macrophage population, representing anti-inflammatory functionality. Also, we observed a significant downregulation of Lgals9 expression in the MAC after mouse heart transplantation. Then, we validated our findings using RT-qPCR and Western blotting and also investigated the impact of Lgals9 on macrophage function through flow cytometry and ELISA. Furthermore, in vitro, we found that rLgals9 (Recombinant Mouse Galectin-9 Protein) treated MAC polarized toward the M2b phenotype at appropriate concentrations.
Collapse
Affiliation(s)
- Yuexing Zhu
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
- Medical College, Guangxi University, 100 University East Road, Nanning, 530004, China
| | - Xinguo Zheng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
- Department of Immunology, School of Basic Medical Science, Central South University, 932 Lushan South Road, Changsha, Hunan 410013, China
| | - Chao Chen
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
- Medical College, Guangxi University, 100 University East Road, Nanning, 530004, China
| | - Ye Xu
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
- Medical College, Guangxi University, 100 University East Road, Nanning, 530004, China
| | - Yuxi Fan
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
- Department of Immunology, School of Basic Medical Science, Central South University, 932 Lushan South Road, Changsha, Hunan 410013, China
| | - Zhouqi Tang
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
| | - Yingqi Zeng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
| | - Chen Feng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
| | - Hedong Zhang
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
| | - Xiaojun Chen
- Department of Nephrology, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Changsha, Hunan 410011, China
| | - Zhongquan Qi
- Fujian Maternity and Child Health Hospital, 18 Daoshan Road, Fuzhou City, Fujian Province 350001, P R. China
| | - Tengfang Li
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
| | - Longkai Peng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
| | - Minjie Lin
- Academic Affairs Department, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Weili Chen
- Department of Analytic Human Pathology, Nippon Medical School, 1-1-5, Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Fenghua Peng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
| | - Xin Jiang
- Department of Organ Transplantation, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), 33 Huanghe Road, Jinshui District, Zhengzhou, Henan 450000, China
| | - Helong Dai
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, 139 Renmin Middle Road, Furong District, Changsha, Hunan 410011, China
- Medical College, Guangxi University, 100 University East Road, Nanning, 530004, China
| |
Collapse
|
5
|
Lafoux B, Fourcaud G, Hortion J, Soyer L, Journeaux A, Germain C, Reynard S, Cousseau H, Larignon C, Pietrosemoli N, Croze S, Lachuer J, Perthame E, Baize S. Expansion of myeloid suppressor cells and suppression of Lassa virus-specific T cells during fatal Lassa fever. PLoS Pathog 2025; 21:e1013111. [PMID: 40245043 PMCID: PMC12040235 DOI: 10.1371/journal.ppat.1013111] [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: 01/16/2025] [Revised: 04/29/2025] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
Lassa fever is a highly lethal hemorrhagic fever endemic to West Africa. In the absence of efficient prophylactic or therapeutic countermeasures, it poses a substantial threat to public health in this region. The pathophysiological mechanisms underlying the severity of the disease are poorly known because Lassa virus (LASV), its causative agent, has to be handled in BSL-4 laboratories and access to clinical samples is difficult. The control of Lassa fever is associated with a rapid and well-balanced immune response and viral clearance. However, severe disease is characterized by uncontrolled innate immune activation and symptoms reminiscent of sepsis and a cytokine storm. In a model of cynomolgus monkeys infected with two different strains of the virus, one causing moderate disease and the other a lethal outcome, we show that the control of LASV infection is characterized by the induction of a LASV-specific T-cell response, whereas severity is associated with the expansion of suppressive myeloid cells, alterations of the stromal network of secondary lymphoid organs, and the anergy of specific T cells. These results suggest that T cells are crucial for the control of LASV and that immunomodulatory therapeutics, such as checkpoint inhibitors, could contribute to new therapeutic strategies to treat Lassa fever. They also highlight how immunosuppressive mechanisms described in sepsis and cancer patients may play a role in the pathogenicity of Lassa fever, as well as in other similar hemorrhagic fevers.
Collapse
Affiliation(s)
- Blaise Lafoux
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1 (UCBL), Institut National de la Santé de la Recherche Médicale (INSERM), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique (CNRS), Lyon, France
| | - Gustave Fourcaud
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1 (UCBL), Institut National de la Santé de la Recherche Médicale (INSERM), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique (CNRS), Lyon, France
| | - Jimmy Hortion
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1 (UCBL), Institut National de la Santé de la Recherche Médicale (INSERM), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique (CNRS), Lyon, France
| | - Laura Soyer
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1 (UCBL), Institut National de la Santé de la Recherche Médicale (INSERM), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique (CNRS), Lyon, France
| | - Alexandra Journeaux
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1 (UCBL), Institut National de la Santé de la Recherche Médicale (INSERM), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique (CNRS), Lyon, France
| | - Clara Germain
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1 (UCBL), Institut National de la Santé de la Recherche Médicale (INSERM), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique (CNRS), Lyon, France
| | - Stéphanie Reynard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1 (UCBL), Institut National de la Santé de la Recherche Médicale (INSERM), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique (CNRS), Lyon, France
| | - Hadrien Cousseau
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1 (UCBL), Institut National de la Santé de la Recherche Médicale (INSERM), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique (CNRS), Lyon, France
| | - Clémentine Larignon
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1 (UCBL), Institut National de la Santé de la Recherche Médicale (INSERM), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique (CNRS), Lyon, France
| | - Natalia Pietrosemoli
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Séverine Croze
- ProfileXpert, SFR Santé Lyon-Est, UCBL, CNRS, INSERM, Lyon, France
| | - Joël Lachuer
- ProfileXpert, SFR Santé Lyon-Est, UCBL, CNRS, INSERM, Lyon, France
| | - Emeline Perthame
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, Paris, France
| | - Sylvain Baize
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie, Université Claude Bernard Lyon 1 (UCBL), Institut National de la Santé de la Recherche Médicale (INSERM), Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique (CNRS), Lyon, France
| |
Collapse
|
6
|
Sepulveda M, Kwan M, Chen L, Cassano A, Cao S, Wang R, Slezak AJ, Hubbell JA, Nagler CR, Alegre ML. Delivery of butyrate to the lower gut by polymeric micelles prolongs survival of distal skin allografts. Am J Transplant 2025; 25:695-705. [PMID: 39566659 PMCID: PMC11972890 DOI: 10.1016/j.ajt.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/14/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
The microbiota composition is known to influence the kinetics of graft rejection, but many questions remain as to whether/how microbiota-derived metabolites affect graft outcome. We investigated the effects of the short-chain fatty acid butyrate, a product of dietary fiber fermentation. Sustained intragastric administration of a micelle-based formulation of butyrate (butyrate micelle [ButM]) that releases its cargo in the lower gastrointestinal tract elevated cecal butyrate content and significantly prolonged minor-mismatched and major-mismatched skin allograft survival in mice. While ButM did not influence regulatory T cells or the adaptive alloimmune responses we tested, it modulated the myeloid cell compartment. At steady state, ButM treatment reduced the number of circulating Ly6ChiCD11b+ monocytes and other myeloid cells in secondary lymphoid organs and skin, altered their expression of genes involved in mitochondrial metabolism and key inflammatory processes, and reduced their ability to produce TNFa, likely via an indirect mechanism. ButM treatment also reduced numbers of graft-infiltrating monocytes but not T cells. Consistent with its critical effect on myeloid cells, ButM's extension of graft survival depended on the presence of CCR2+ cells. These findings imply that cecal ButM improves distal allograft outcomes by quantitatively and qualitatively modulating myeloid cells, thereby inhibiting the innate immune cell-mediated effector phase of alloimmunity.
Collapse
Affiliation(s)
- Martin Sepulveda
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Montserrat Kwan
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Luqiu Chen
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Alexandra Cassano
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Shijie Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| | - Ruyi Wang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA; Department of Chemistry, University of Chicago, Chicago, Illinois, USA
| | - Anna J Slezak
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| | - Cathryn R Nagler
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA; Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Maria-Luisa Alegre
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA.
| |
Collapse
|
7
|
Chen C, Zhen Z, Cui M, Hu X, Yu L, Zhou F, Yu X, Yang D, Wu H, Cui Y, Li X, Cui X, Liang X, Gao Y, Liu Y, Yu Y, Huang Z, Zhang F. The core cellular network modulates immune phenotype switching in hepatitis B. Sci Bull (Beijing) 2025:S2095-9273(25)00289-0. [PMID: 40175178 DOI: 10.1016/j.scib.2025.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Affiliation(s)
- Chuangeng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Ziqi Zhen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Meng Cui
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Xiaoli Hu
- Department of Infectious Diseases, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin 150030, China
| | - Lei Yu
- Department of Infectious Diseases, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Fengxia Zhou
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Xiaorong Yu
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Dehui Yang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - He Wu
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, China
| | - Ying Cui
- Department of Infectious Diseases, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin 150030, China
| | - Xiang Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Xudong Cui
- Department of Infectious Diseases, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xinyue Liang
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Yiyang Gao
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yuchen Liu
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Yang Yu
- Harbin Institute of Technology Hospital, Harbin 150006, China
| | - Zhiwei Huang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China.
| | - Fan Zhang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China.
| |
Collapse
|
8
|
Baek H, Yang SW, Kim S, Lee Y, Park H, Park M, Jeon BJ, Park H, Hwang HS, Kim JY, Kim JH, Kang YS. Development of Anti-Inflammatory Agents Utilizing DC-SIGN Mediated IL-10 Secretion in Autoimmune and Immune-Mediated Disorders: Bridging Veterinary and Human Health. Int J Mol Sci 2025; 26:2329. [PMID: 40076949 PMCID: PMC11901132 DOI: 10.3390/ijms26052329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
DC-SIGN (dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin) is a C-type lectin receptor expressed on dendritic cells and M2 macrophages, playing a key role in immune regulation and pathogen recognition. Its ability to mediate anti-inflammatory effects by interacting with specific ligands triggers pathways that suppress pro-inflammatory responses and promote tissue repair, making it a potential therapeutic target for inflammatory and autoimmune diseases. DC-SIGN homologs in various animal species share structural similarities and perform comparable immune functions, offering valuable insights into its broader application across species. By recognizing carbohydrate ligands on pathogens, DC-SIGN facilitates immune modulation, which can be harnessed for developing therapies aimed at controlling inflammation. In veterinary medicine, autoimmune and inflammatory diseases, such as rheumatoid arthritis and inflammatory bowel disease, represent significant challenges, and the anti-inflammatory properties of DC-SIGN could provide new therapeutic options to improve disease management and enhance animal health. Future investigations should focus on the structural and functional analysis of DC-SIGN homologs in various species, as well as the development of preclinical models to translate these findings into clinical interventions bridging veterinary and human health.
Collapse
Affiliation(s)
- Hayeon Baek
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (H.B.); (M.P.)
| | - Seung-Woo Yang
- Sanford Consortium for Regenerative Medicine, School of Medicine, University of California, San Diego, CA 92037, USA;
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Seulki Kim
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.K.); (Y.L.)
| | - Yunseok Lee
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.K.); (Y.L.)
| | - Hwi Park
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Min Park
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (H.B.); (M.P.)
| | - Byung-Ju Jeon
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Hanwool Park
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Han-Sung Hwang
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Joon-Young Kim
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (H.P.); (B.-J.J.); (H.P.); (J.-Y.K.)
| | - Jung-Hyun Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Young-Sun Kang
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; (H.B.); (M.P.)
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (S.K.); (Y.L.)
| |
Collapse
|
9
|
Costa ADSD, Vadym K, Park K. Engineered endothelium model enables recapitulation of vascular function and early atherosclerosis development. Biomaterials 2025; 314:122889. [PMID: 39423515 DOI: 10.1016/j.biomaterials.2024.122889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Human health relies heavily on the vascular endothelium. Here, we propose a novel engineered endothelium model (EEM), which recapitulated both normal vascular function and pathology. An artificial basement membrane (aBM), where porous polyvinyl alcohol hydrogel was securely integrated with human fibroblast-derived, decellularized extracellular matrix on both sides was fabricated first and followed by endothelial cells (ECs) and pericytes (PCs) adhesion, respectively. Our EEM formed robust adherens junction (VE-cad) and built an impermeable barrier with time, along with the nitric oxide (NO) secretion. In our EEM, ECs and PCs interacted each other via aBM and led to hemoglobin alpha 1 (Hb-α1) development, which was involved in NO control and was strongly interconnected with VE-cad as well. A resilient property of EEM under inflammatory milieu was also confirmed by VE-cad and barrier recovery with time. In particular interest, foam cells formation, a hallmark of atherosclerotic initiation was successfully recapitulated in our EEM, where a series of sequential events were confirmed: human monocytes adhesion, transendothelial migration, and oxidized low-density lipoprotein uptake by macrophages. Collectively, our EEM is excellent in recapitulating not only normal endothelium but early pathologic one, thereby enabling EEM to be a physiologically relevant model for vascular study and disease modeling.
Collapse
Affiliation(s)
| | - Kopych Vadym
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
10
|
Han L, Wu T, Zhang Q, Qi A, Zhou X. Immune Tolerance Regulation Is Critical to Immune Homeostasis. J Immunol Res 2025; 2025:5006201. [PMID: 39950084 PMCID: PMC11824399 DOI: 10.1155/jimr/5006201] [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: 06/12/2024] [Accepted: 12/07/2024] [Indexed: 02/16/2025] Open
Abstract
The body's immune response plays a critical role in defending against external or foreign antigens while also preserving tolerance to self-antigens. This equilibrium, referred to as immune homeostasis, is paramount for overall health. The regulatory mechanisms governing the maintenance of this delicate immune balance are notably complex. It is currently accepted that immune tolerance is a dynamic outcome regulated by multiple factors, including central and peripheral mechanisms. Its induction or elimination plays a significant role in autoimmune diseases, organ transplantation, and cancer therapy, markedly impacting various major diseases in modern clinical practice. Overall, our current understanding of immune tolerance is still very limited. In this review article, we summarized the main mechanisms that have been known to mediate immune tolerance so far, including endogenous immune tolerance, adaptive immune tolerance, other immune tolerance mechanisms, and the homeostasis of immune tolerance, identified the key factors that regulate immune tolerance, and provided new clues for immune system recovery in many autoimmune diseases, organ transplantation, and tumor therapy.
Collapse
Affiliation(s)
- Lei Han
- Department of Pharmacy, Jiangsu Health Vocational College, Nanjing 211800, Jiangsu, China
| | - Tianxiang Wu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Qin Zhang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Anning Qi
- Medical Laboratory, Liuhe People's Hospital of Jiangsu Province, Nanjing, Jiangsu 211500, China
| | - Xiaohui Zhou
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
11
|
Owen MC, Kopecky BJ. Targeting Macrophages in Organ Transplantation: A Step Toward Personalized Medicine. Transplantation 2024; 108:2045-2056. [PMID: 38467591 PMCID: PMC11390981 DOI: 10.1097/tp.0000000000004978] [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] [Indexed: 03/13/2024]
Abstract
Organ transplantation remains the most optimal strategy for patients with end-stage organ failure. However, prevailing methods of immunosuppression are marred by adverse side effects, and allograft rejection remains common. It is imperative to identify and comprehensively characterize the cell types involved in allograft rejection, and develop therapies with greater specificity. There is increasing recognition that processes mediating allograft rejection are the result of interactions between innate and adaptive immune cells. Macrophages are heterogeneous innate immune cells with diverse functions that contribute to ischemia-reperfusion injury, acute rejection, and chronic rejection. Macrophages are inflammatory cells capable of innate allorecognition that strengthen their responses to secondary exposures over time via "trained immunity." However, macrophages also adopt immunoregulatory phenotypes and may promote allograft tolerance. In this review, we discuss the roles of macrophages in rejection and tolerance, and detail how macrophage plasticity and polarization influence transplantation outcomes. A comprehensive understanding of macrophages in transplant will guide future personalized approaches to therapies aimed at facilitating tolerance or mitigating the rejection process.
Collapse
Affiliation(s)
- Macee C Owen
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MI
| | - Benjamin J Kopecky
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MI
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
12
|
Cui Y, Hackett RG, Ascue J, Muralidaran V, Patil D, Kang J, Kaufman SS, Khan K, Kroemer A. Innate and Adaptive Immune Responses in Intestinal Transplant Rejection: Through the Lens of Inflammatory Bowel and Intestinal Graft-Versus-Host Diseases. Gastroenterol Clin North Am 2024; 53:359-382. [PMID: 39068000 DOI: 10.1016/j.gtc.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Intestinal transplantation is a life-saving procedure utilized for patients failing total parenteral nutrition. However, intestinal transplantattion remains plagued with low survival rates and high risk of allograft rejection. The authors explore roles of innate (macrophages, natural killer cells, innate lymphoid cells) and adaptive immune cells (Th1, Th2, Th17, Tregs) in inflammatory responses, particularly inflammatory bowel disease and graft versus host disease, and correlate these findings to intestinal allograft rejection, highlighting which effectors exacerbate or suppress intestinal rejection. Better understanding of this immunology can open further investigation into potential biomolecular targets to develop improved therapeutic treatment options and immunomonitoring techniques to combat allograft rejection and enhance patient lives.
Collapse
Affiliation(s)
- Yuki Cui
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Ryan G Hackett
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Jhalen Ascue
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Vinona Muralidaran
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Digvijay Patil
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Jiman Kang
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Stuart S Kaufman
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Khalid Khan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Alexander Kroemer
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
13
|
Lu X, Xu Z, Shu F, Wang Y, Han Y, Yang X, Shi P, Fan C, Wang L, Yu F, Sun Q, Cheng F, Chen H. Reactive Oxygen Species Responsive Multifunctional Fusion Extracellular Nanovesicles: Prospective Treatments for Acute Heart Transplant Rejection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406758. [PMID: 38949397 DOI: 10.1002/adma.202406758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/19/2024] [Indexed: 07/02/2024]
Abstract
Heart transplantation offers life-saving treatment for patients with end-stage heart failure; however, ischemia-reperfusion injury (IRI) and subsequent immune responses remain significant challenges. Current therapies primarily target adaptive immunity, with limited options available for addressing IRI and innate immune activation. Although plant-derived vesicle-like nanoparticles show promise in managing diseases, their application in organ transplantation complications is unexplored. Here, this work develops a novel reactive oxygen species (ROS)-responsive multifunctional fusion extracellular nanovesicles carrying rapamycin (FNVs@RAPA) to address early IRI and Ly6C+Ly6G- inflammatory macrophage-mediated rejection in heart transplantation. The FNVs comprise Exocarpium Citri grandis-derived extracellular nanovesicles with anti-inflammatory and antioxidant properties, and mesenchymal stem cell membrane-derived nanovesicles expressing calreticulin with macrophage-targeting ability. A novel ROS-responsive bio-orthogonal chemistry approach facilitates the active targeting delivery of FNVs@RAPA to the heart graft site, effectively alleviating IRI and promoting the polarization of Ly6C+Ly6G- inflammatory macrophages toward an anti-inflammatory phenotype. Hence, FNVs@RAPA represents a promising therapeutic approach for mitigating early transplantation complications and immune rejection. The fusion-targeted delivery strategy offers superior heart graft site enrichment and macrophage-specific targeting, promising improved transplant outcomes.
Collapse
Affiliation(s)
- Xingyu Lu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhanxue Xu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Fan Shu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yidan Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yuhang Han
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinrui Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Peilin Shi
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Chuanqiang Fan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Linglu Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Fei Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Qipeng Sun
- Department of Kidney Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
14
|
Chen Y, Wang J, An C, Bao S, Zhang C. The role and research progress of macrophages after heart transplantation. Heliyon 2024; 10:e33844. [PMID: 39027574 PMCID: PMC11255595 DOI: 10.1016/j.heliyon.2024.e33844] [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: 02/25/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
Since the 60s of the 20th century, heart transplantation has been the best treatment for patients with end-stage heart failure. Due to the increasing number of patients, how to expand the number of donor organs and enhance immune compatibility has become an urgent problem to be solved at this stage. Although current immunosuppression is effective, its side effects are also quite obvious, such as opportunistic infections and malignant tumors. In this review, we focus on the important role in macrophages after heart transplantation and their potential targets for achieving allogeneic graft tolerance, in order to improve effective graft survival and reduce infection and the occurrence of malignant tumors.
Collapse
Affiliation(s)
- Yao Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - JianPeng Wang
- School of First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Cheng An
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - ShanQing Bao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - ChengXin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| |
Collapse
|
15
|
DeBerge M, Schroth S, Du F, Yeap XY, Wang JJ, Zhang ZJ, Ansari MJ, Scott EA, Thorp EB. Hypoxia inducible factor 2α promotes tolerogenic macrophage development during cardiac transplantation through transcriptional regulation of colony stimulating factor 1 receptor. Proc Natl Acad Sci U S A 2024; 121:e2319623121. [PMID: 38889142 PMCID: PMC11214057 DOI: 10.1073/pnas.2319623121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Solid organ transplantation mobilizes myeloid cells, including monocytes and macrophages, which are central protagonists of allograft rejection. However, myeloid cells can also be functionally reprogrammed by perioperative costimulatory blockade to promote a state of transplantation tolerance. Transplantation tolerance holds promise to reduce complications from chronic immunosuppression and promote long-term survival in transplant recipients. We sought to identify different mediators of transplantation tolerance by performing single-cell RNA sequencing of acute rejecting or tolerized cardiac allografts. This led to the unbiased identification of the transcription factor, hypoxia inducible factor (HIF)-2α, in a subset of tolerogenic monocytes. Using flow cytometric analyses and mice with conditional loss or gain of function, we uncovered that myeloid cell expression of HIF-2α was required for costimulatory blockade-induced transplantation tolerance. While HIF-2α was dispensable for mobilization of tolerogenic monocytes, which were sourced in part from the spleen, it promoted the expression of colony stimulating factor 1 receptor (CSF1R). CSF1R mediates monocyte differentiation into tolerogenic macrophages and was found to be a direct transcriptional target of HIF-2α in splenic monocytes. Administration of the HIF stabilizer, roxadustat, within micelles to target myeloid cells, increased HIF-2α in splenic monocytes, which was associated with increased CSF1R expression and enhanced cardiac allograft survival. These data support further exploration of HIF-2α activation in myeloid cells as a therapeutic strategy for transplantation tolerance.
Collapse
Affiliation(s)
- Matthew DeBerge
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center, Houston, TX77030
| | - Samantha Schroth
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Fanfan Du
- Department of Biomedical Engineering, Northwestern University, Evanston, IL60208
| | - Xin Yi Yeap
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Jiao-Jing Wang
- Department of Surgery, Comprehensive Transplant Center, Northwestern University, Chicago, IL60611
| | - Zheng Jenny Zhang
- Department of Surgery, Comprehensive Transplant Center, Northwestern University, Chicago, IL60611
| | - Mohammed Javeed Ansari
- Division of Nephrology and Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Evan A. Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL60208
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| |
Collapse
|
16
|
Zhang J, Pei J, Yu C, Luo J, Hong Y, Hua Y, Wei G. CCR7 and CD48 as Predicted Targets in Acute Rejection Related to M1 Macrophage after Pediatric Kidney Transplantation. J Immunol Res 2024; 2024:6908968. [PMID: 38957433 PMCID: PMC11217580 DOI: 10.1155/2024/6908968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/28/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024] Open
Abstract
Background Kidney transplantation (KT) is the best treatment for end-stage renal disease. Although long and short-term survival rates for the graft have improved significantly with the development of immunosuppressants, acute rejection (AR) remains a major risk factor attacking the graft and patients. The innate immune response plays an important role in rejection. Therefore, our objective is to determine the biomarkers of congenital immunity associated with AR after KT and provide support for future research. Materials and Methods A differential expression genes (DEGs) analysis was performed based on the dataset GSE174020 from the NCBI gene Expression Synthesis Database (GEO) and then combined with the GSE5099 M1 macrophage-related gene identified in the Molecular Signatures Database. We then identified genes in DEGs associated with M1 macrophages defined as DEM1Gs and performed gene ontology (GO) and Kyoto Encyclopedia of Genomes (KEGG) enrichment analysis. Cibersort was used to analyze the immune cell infiltration during AR. At the same time, we used the protein-protein interaction (PPI) network and Cytoscape software to determine the key genes. Dataset, GSE14328 derived from pediatric patients, GSE138043 and GSE9493 derived from adult patients, were used to verify Hub genes. Additional verification was the rat KT model, which was used to perform HE staining, immunohistochemical staining, and Western Blot. Hub genes were searched in the HPA database to confirm their expression. Finally, we construct the interaction network of transcription factor (TF)-Hub genes and miRNA-Hub genes. Results Compared to the normal group, 366 genes were upregulated, and 423 genes were downregulated in the AR group. Then, 106 genes related to M1 macrophages were found among these genes. GO and KEGG enrichment analysis showed that these genes are mainly involved in cytokine binding, antigen binding, NK cell-mediated cytotoxicity, activation of immune receptors and immune response, and activation of the inflammatory NF-κB signaling pathway. Two Hub genes, namely CCR7 and CD48, were identified by PPI and Cytoscape analysis. They have been verified in external validation sets, originated from both pediatric patients and adult patients, and animal experiments. In the HPA database, CCR7 and CD48 are mainly expressed in T cells, B cells, macrophages, and tissues where these immune cells are distributed. In addition to immunoinfiltration, CD4+T, CD8+T, NK cells, NKT cells, and monocytes increased significantly in the AR group, which was highly consistent with the results of Hub gene screening. Finally, we predicted that 19 TFs and 32 miRNAs might interact with the Hub gene. Conclusions Through a comprehensive bioinformatic analysis, our findings may provide predictive and therapeutic targets for AR after KT.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Jun Pei
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Chengjun Yu
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Jin Luo
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Yifan Hong
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Yi Hua
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| | - Guanghui Wei
- Department of Urology Children's Hospital of Chongqing Medical UniversityNational Clinical Research Center for Child Health and DisordersMinistry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing, China
| |
Collapse
|
17
|
Iglesias M, Bibicheff D, Komin A, Chicco M, Guinn S, Foley B, Raimondi G. T cell responsiveness to IL-10 defines the immunomodulatory effect of costimulation blockade via anti-CD154 and impacts transplant survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598652. [PMID: 38915537 PMCID: PMC11195256 DOI: 10.1101/2024.06.12.598652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Costimulation blockade (CoB)-based immunotherapy is a promising alternative to immunosuppression for transplant recipients; however, the current limited understanding of the factors that impact its efficacy restrains its clinical applicability. In this context, pro- and anti-inflammatory cytokines are being recognized as having an impact on T cell activation beyond effector differentiation. This study aims at elucidating the impact of direct IL-10 signaling in T cells on CoB outcomes. We used a full-mismatch skin transplantation model where recipients had a T cell-restricted expression of a dominant negative IL-10 receptor (10R-DN), alongside anti-CD154 as CoB therapy. Unlike wild-type recipients, 10R-DN mice failed to benefit from CoB. This accelerated graft rejection correlated with increased accumulation of T cells producing TNF-α, IFN-γ, and IL-17. In vitro experiments indicated that while lack of IL-10 signaling did not change the ability of anti-CD154 to modulate alloreactive T cell proliferation, the absence of this pathway heightened TH1 effector cell differentiation. Furthermore, deficiency of IL-10 signaling in T cells impaired Treg induction, a hallmark of anti-CD154 therapy. Overall, these findings unveil an important and novel role of IL-10 signaling in T cells that defines the success of CoB therapies and identifies a target pathway for obtaining robust immunoregulation.
Collapse
Affiliation(s)
- Marcos Iglesias
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Darrel Bibicheff
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander Komin
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maria Chicco
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samantha Guinn
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brendan Foley
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Giorgio Raimondi
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Lee HJ, Choi YR, Ko JH, Ryu JS, Oh JY. Defining mesenchymal stem/stromal cell-induced myeloid-derived suppressor cells using single-cell transcriptomics. Mol Ther 2024; 32:1970-1983. [PMID: 38627968 PMCID: PMC11184332 DOI: 10.1016/j.ymthe.2024.04.026] [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: 12/30/2023] [Revised: 02/27/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) modulate the immune response through interactions with innate immune cells. We previously demonstrated that MSCs alleviate ocular autoimmune inflammation by directing bone marrow cell differentiation from pro-inflammatory CD11bhiLy6ChiLy6Glo cells into immunosuppressive CD11bmidLy6CmidLy6Glo cells. Herein, we analyzed MSC-induced CD11bmidLy6Cmid cells using single-cell RNA sequencing and compared them with CD11bhiLy6Chi cells. Our investigation revealed seven distinct immune cell types including myeloid-derived suppressor cells (MDSCs) in the CD11bmidLy6Cmid cells, while CD11bhiLy6Chi cells included mostly monocytes/macrophages with a small cluster of neutrophils. These MSC-induced MDSCs highly expressed Retnlg, Cxcl3, Cxcl2, Mmp8, Cd14, and Csf1r as well as Arg1. Comparative analyses of CSF-1RhiCD11bmidLy6Cmid and CSF-1RloCD11bmidLy6Cmid cells demonstrated that the former had a homogeneous monocyte morphology and produced elevated levels of interleukin-10. Functionally, these CSF-1RhiCD11bmidLy6Cmid cells, compared with the CSF-1RloCD11bmidLy6Cmid cells, inhibited CD4+ T cell proliferation and promoted CD4+CD25+Foxp3+ Treg expansion in culture and in a mouse model of experimental autoimmune uveoretinitis. Resistin-like molecule (RELM)-γ encoded by Retnlg, one of the highly upregulated genes in MSC-induced MDSCs, had no direct effects on T cell proliferation, Treg expansion, or splenocyte activation. Together, our study revealed a distinct transcriptional profile of MSC-induced MDSCs and identified CSF-1R as a key cell-surface marker for detection and therapeutic enrichment of MDSCs.
Collapse
Affiliation(s)
- Hyun Ju Lee
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Yoo Rim Choi
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Jung Hwa Ko
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Jin Suk Ryu
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Joo Youn Oh
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea; Department of Ophthalmology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea.
| |
Collapse
|
19
|
Šestan M, Mikašinović S, Benić A, Wueest S, Dimitropoulos C, Mladenić K, Krapić M, Hiršl L, Glantzspiegel Y, Rasteiro A, Aliseychik M, Cekinović Grbeša Đ, Turk Wensveen T, Babić M, Gat-Viks I, Veiga-Fernandes H, Konrad D, Wensveen FM, Polić B. An IFNγ-dependent immune-endocrine circuit lowers blood glucose to potentiate the innate antiviral immune response. Nat Immunol 2024; 25:981-993. [PMID: 38811816 DOI: 10.1038/s41590-024-01848-3] [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: 02/01/2024] [Accepted: 04/18/2024] [Indexed: 05/31/2024]
Abstract
Viral infection makes us feel sick as the immune system alters systemic metabolism to better fight the pathogen. The extent of these changes is relative to the severity of disease. Whether blood glucose is subject to infection-induced modulation is mostly unknown. Here we show that strong, nonlethal infection restricts systemic glucose availability, which promotes the antiviral type I interferon (IFN-I) response. Following viral infection, we find that IFNγ produced by γδ T cells stimulates pancreatic β cells to increase glucose-induced insulin release. Subsequently, hyperinsulinemia lessens hepatic glucose output. Glucose restriction enhances IFN-I production by curtailing lactate-mediated inhibition of IRF3 and NF-κB signaling. Induced hyperglycemia constrained IFN-I production and increased mortality upon infection. Our findings identify glucose restriction as a physiological mechanism to bring the body into a heightened state of responsiveness to viral pathogens. This immune-endocrine circuit is disrupted in hyperglycemia, possibly explaining why patients with diabetes are more susceptible to viral infection.
Collapse
Affiliation(s)
- Marko Šestan
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Sanja Mikašinović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ante Benić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | | | - Karlo Mladenić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Mia Krapić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Lea Hiršl
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Yossef Glantzspiegel
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ana Rasteiro
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Maria Aliseychik
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | | | - Tamara Turk Wensveen
- Center for Diabetes, Endocrinology and Cardiometabolism, Thallassotherapia, Opatija, Croatia
- Department of Internal Medicine, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Marina Babić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Innate Immunity, German Rheumatism Research Centre, Leibniz Institute, Berlin, Germany
| | - Irit Gat-Viks
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Centre, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Felix M Wensveen
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Bojan Polić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
20
|
Liang C, Zhang Y, Wang S, Jiao W, Guo J, Zhang N, Liu X. Nanomaterials in modulating tumor-associated macrophages and enhancing immunotherapy. J Mater Chem B 2024; 12:4809-4823. [PMID: 38695349 DOI: 10.1039/d4tb00230j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Tumor-associated macrophages (TAMs) are predominantly present in the tumor microenvironment (TME) and play a crucial role in shaping the efficacy of tumor immunotherapy. These TAMs primarily exhibit a tumor-promoting M2-like phenotype, which is associated with the suppression of immune responses and facilitation of tumor progression. Interestingly, recent research has highlighted the potential of repolarizing TAMs from an M2 to a pro-inflammatory M1 status-a shift that has shown promise in impeding tumor growth and enhancing immune responsiveness. This concept is particularly intriguing as it offers a new dimension to cancer therapy by targeting the tumor microenvironment, which is a significant departure from traditional approaches that focus solely on tumor cells. However, the clinical application of TAM-modulating agents is often challenged by issues such as insufficient tumor accumulation and off-target effects, limiting their effectiveness and safety. In this regard, nanomaterials have emerged as a novel solution. They serve a dual role: as delivery vehicles that can enhance the accumulation of therapeutic agents in the tumor site and as TAM-modulators. This dual functionality of nanomaterials is a significant advancement as it addresses the key limitations of current TAM-modulating strategies and opens up new avenues for more efficient and targeted therapies. This review provides a comprehensive overview of the latest mechanisms and strategies involving nanomaterials in modulating macrophage polarization within the TME. It delves into the intricate interactions between nanomaterials and macrophages, elucidating how these interactions can be exploited to drive macrophage polarization towards a phenotype that is more conducive to anti-tumor immunity. Additionally, the review explores the burgeoning field of TAM-associated nanomedicines in combination with tumor immunotherapy. This combination approach is particularly promising as it leverages the strengths of both nanomedicine and immunotherapy, potentially leading to synergistic effects in combating cancer.
Collapse
Affiliation(s)
- Chen Liang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
| | - Yihan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
| | - Siyao Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
| | - Wangbo Jiao
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
| | - Jingyi Guo
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
| | - Nan Zhang
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xiaoli Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
21
|
Urie RR, Morris A, Farris D, Hughes E, Xiao C, Chen J, Lombard E, Feng J, Li JZ, Goldstein DR, Shea LD. Biomarkers from subcutaneous engineered tissues predict acute rejection of organ allografts. SCIENCE ADVANCES 2024; 10:eadk6178. [PMID: 38748794 PMCID: PMC11095459 DOI: 10.1126/sciadv.adk6178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/10/2024] [Indexed: 05/19/2024]
Abstract
Invasive graft biopsies assess the efficacy of immunosuppression through lagging indicators of transplant rejection. We report on a microporous scaffold implant as a minimally invasive immunological niche to assay rejection before graft injury. Adoptive transfer of T cells into Rag2-/- mice with mismatched allografts induced acute cellular allograft rejection (ACAR), with subsequent validation in wild-type animals. Following murine heart or skin transplantation, scaffold implants accumulate predominantly innate immune cells. The scaffold enables frequent biopsy, and gene expression analyses identified biomarkers of ACAR before clinical signs of graft injury. This gene signature distinguishes ACAR and immunodeficient respiratory infection before injury onset, indicating the specificity of the biomarkers to differentiate ACAR from other inflammatory insult. Overall, this implantable scaffold enables remote evaluation of the early risk of rejection, which could potentially be used to reduce the frequency of routine graft biopsy, reduce toxicities by personalizing immunosuppression, and prolong transplant life.
Collapse
Affiliation(s)
- Russell R. Urie
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Aaron Morris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Diana Farris
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elizabeth Hughes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chengchuan Xiao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Judy Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elizabeth Lombard
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiane Feng
- Animal Phenotyping Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jun Z. Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel R. Goldstein
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
22
|
Knoedler L, Dean J, Diatta F, Thompson N, Knoedler S, Rhys R, Sherwani K, Ettl T, Mayer S, Falkner F, Kilian K, Panayi AC, Iske J, Safi AF, Tullius SG, Haykal S, Pomahac B, Kauke-Navarro M. Immune modulation in transplant medicine: a comprehensive review of cell therapy applications and future directions. Front Immunol 2024; 15:1372862. [PMID: 38650942 PMCID: PMC11033354 DOI: 10.3389/fimmu.2024.1372862] [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: 01/18/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024] Open
Abstract
Balancing the immune response after solid organ transplantation (SOT) and vascularized composite allotransplantation (VCA) remains an ongoing clinical challenge. While immunosuppressants can effectively reduce acute rejection rates following transplant surgery, some patients still experience recurrent acute rejection episodes, which in turn may progress to chronic rejection. Furthermore, these immunosuppressive regimens are associated with an increased risk of malignancies and metabolic disorders. Despite significant advancements in the field, these IS related side effects persist as clinical hurdles, emphasizing the need for innovative therapeutic strategies to improve transplant survival and longevity. Cellular therapy, a novel therapeutic approach, has emerged as a potential pathway to promote immune tolerance while minimizing systemic side-effects of standard IS regiments. Various cell types, including chimeric antigen receptor T cells (CAR-T), mesenchymal stromal cells (MSCs), regulatory myeloid cells (RMCs) and regulatory T cells (Tregs), offer unique immunomodulatory properties that may help achieve improved outcomes in transplant patients. This review aims to elucidate the role of cellular therapies, particularly MSCs, T cells, Tregs, RMCs, macrophages, and dendritic cells in SOT and VCA. We explore the immunological features of each cell type, their capacity for immune regulation, and the prospective advantages and obstacles linked to their application in transplant patients. An in-depth outline of the current state of the technology may help SOT and VCA providers refine their perioperative treatment strategies while laying the foundation for further trials that investigate cellular therapeutics in transplantation surgery.
Collapse
Affiliation(s)
- Leonard Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Jillian Dean
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fortunay Diatta
- Division of Plastic Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Noelle Thompson
- University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Samuel Knoedler
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Richmond Rhys
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Khalil Sherwani
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Tobias Ettl
- Department of Dental, Oral and Maxillofacial Surgery, Regensburg, Germany
| | - Simon Mayer
- University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Florian Falkner
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Katja Kilian
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Adriana C. Panayi
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, Berufsgenossenschaft (BG) Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Jasper Iske
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ali-Farid Safi
- Faculty of Medicine, University of Bern, Bern, Switzerland
- Craniologicum, Center for Cranio-Maxillo-Facial Surgery, Bern, Switzerland
| | - Stefan G. Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Siba Haykal
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Bohdan Pomahac
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Martin Kauke-Navarro
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
23
|
Aiello S, Benigni A, Remuzzi G. Tissue-Resident Macrophages in Solid Organ Transplantation: Harmful or Protective? JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1051-1061. [PMID: 38498808 DOI: 10.4049/jimmunol.2300625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/27/2023] [Indexed: 03/20/2024]
Abstract
Transplanted organs carry donor immune cells into the recipient, the majority of which are tissue-resident macrophages (TRMs). The role they play in guiding the fate of the transplanted organ toward acceptance or rejection remains elusive. TRMs originate from both embryonic and bone marrow-derived precursors. Embryo-derived TRMs retain the embryonic capability to proliferate, so they are able to self-renew and, theoretically, persist for extended periods of time after transplantation. Bone marrow-derived TRMs do not proliferate and must constantly be replenished by adult circulating monocytes. Recent studies have aimed to clarify the different roles and interactions between donor TRMs, recipient monocytes, and monocyte-derived macrophages (MFs) after organ transplantation. This review aims to shed light on how MFs affect the fate of a transplanted organ by differentiating between the role of donor TRMs and that of MFs derived from graft infiltrating monocytes.
Collapse
Affiliation(s)
- Sistiana Aiello
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
24
|
Elesela S, Arzola-Martínez L, Rasky A, Ptaschinski C, Hogan SP, Lukacs NW. Mucosal IgA immune complex induces immunomodulatory responses in allergic airway and intestinal T H2 disease. J Allergy Clin Immunol 2023; 152:1607-1618.e1. [PMID: 37604310 DOI: 10.1016/j.jaci.2023.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND IgA is the most abundant immunoglobulin at the mucosal surface and although its role in regulating mucosal immunity is not fully understood, its presence is associated with protection from developing allergic disease. OBJECTIVE We sought to determine the role of IgA immune complexes for therapeutic application to mucosal allergic responses. METHODS Trinitrophenol (TNP)-specific IgA immune complexes were applied, using TNP-coupled ovalbumin (OVA), to airway and gut mucosal surfaces in systemically sensitized allergic animals to regulate allergen challenge responses. Animals were assessed for both pathologic and immune-mediated responses in the lung and gut, respectively, using established mouse models. RESULTS The mucosal application of IgA immune complexes in the lung and gut with TNP-OVA regulated TH2-driven allergic response in the lung and gut, reducing TH2 cytokines and mucus (lung) as well as diarrhea and temperature loss (gut), but increasing IL-10 and the number of regulatory T cells. The IgA-OVA immune complex did not alter peanut-induced anaphylaxis, indicating antigen specificity. Using OVA-specific DO.11-green fluorescent protein IL-4 reporter mouse-derived TH2-skewed cells in a transfer model demonstrated that mucosal IgA immune complex treatment reduced TH2-cell expansion and increased the number of regulatory T cells. To address a potential mechanism of action, TGF-β and IL-10 were induced in bone marrow-derived dendritic cells when they were exposed to IgA immune complex, suggesting a regulatory phenotype induced in dendritic cells that also led to an altered primary T-cell-mediated response in in vitro OVA-specific assays. CONCLUSIONS These studies highlight one possible mechanism of how allergen-specific IgA may provide a regulatory signal to reduce the development of allergic responses in the lung and gut.
Collapse
Affiliation(s)
- Srikanth Elesela
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Llilian Arzola-Martínez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Andrew Rasky
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Simon P Hogan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich.
| |
Collapse
|
25
|
Guinn MT, Szuter ES, Yokose T, Ge J, Rosales IA, Chetal K, Sadreyev RI, Cuenca AG, Kreisel D, Sage PT, Russell PS, Madsen JC, Colvin RB, Alessandrini A. Intragraft B cell differentiation during the development of tolerance to kidney allografts is associated with a regulatory B cell signature revealed by single cell transcriptomics. Am J Transplant 2023; 23:1319-1330. [PMID: 37295719 PMCID: PMC11232115 DOI: 10.1016/j.ajt.2023.05.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/23/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023]
Abstract
Mouse kidney allografts are spontaneously accepted in select, fully mismatched donor-recipient strain combinations, like DBA/2J to C57BL/6 (B6), by natural tolerance. We previously showed accepted renal grafts form aggregates containing various immune cells within 2 weeks posttransplant, referred to as regulatory T cell-rich organized lymphoid structures, which are a novel regulatory tertiary lymphoid organ. To characterize the cells within T cell-rich organized lymphoid structures, we performed single-cell RNA sequencing on CD45+ sorted cells from accepted and rejected renal grafts from 1-week to 6-months posttransplant. Analysis of single-cell RNA sequencing data revealed a shifting from a T cell-dominant to a B cell-rich population by 6 months with an increased regulatory B cell signature. Furthermore, B cells were a greater proportion of the early infiltrating cells in accepted vs rejecting grafts. Flow cytometry of B cells at 20 weeks posttransplant revealed T cell, immunoglobulin domain and mucin domain-1+ B cells, potentially implicating a regulatory role in the maintenance of allograft tolerance. Lastly, B cell trajectory analysis revealed intragraft differentiation from precursor B cells to memory B cells in accepted allografts. In summary, we show a shifting T cell- to B cell-rich environment and a differential cellular pattern among accepted vs rejecting kidney allografts, possibly implicating B cells in the maintenance of kidney allograft acceptance.
Collapse
Affiliation(s)
- Michael Tyler Guinn
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA; Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Edward S Szuter
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Takahiro Yokose
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jifu Ge
- Boston's Children Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivy A Rosales
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kashish Chetal
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ruslan I Sadreyev
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alex G Cuenca
- Boston's Children Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Kreisel
- Departments of Surgery, Pathology, and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Peter T Sage
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paul S Russell
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joren C Madsen
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA; Division of Cardiac Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robert B Colvin
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alessandro Alessandrini
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
26
|
Strizova Z, Benesova I, Bartolini R, Novysedlak R, Cecrdlova E, Foley L, Striz I. M1/M2 macrophages and their overlaps - myth or reality? Clin Sci (Lond) 2023; 137:1067-1093. [PMID: 37530555 PMCID: PMC10407193 DOI: 10.1042/cs20220531] [Citation(s) in RCA: 164] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 08/03/2023]
Abstract
Macrophages represent heterogeneous cell population with important roles in defence mechanisms and in homoeostasis. Tissue macrophages from diverse anatomical locations adopt distinct activation states. M1 and M2 macrophages are two polarized forms of mononuclear phagocyte in vitro differentiation with distinct phenotypic patterns and functional properties, but in vivo, there is a wide range of different macrophage phenotypes in between depending on the microenvironment and natural signals they receive. In human infections, pathogens use different strategies to combat macrophages and these strategies include shaping the macrophage polarization towards one or another phenotype. Macrophages infiltrating the tumours can affect the patient's prognosis. M2 macrophages have been shown to promote tumour growth, while M1 macrophages provide both tumour-promoting and anti-tumour properties. In autoimmune diseases, both prolonged M1 activation, as well as altered M2 function can contribute to their onset and activity. In human atherosclerotic lesions, macrophages expressing both M1 and M2 profiles have been detected as one of the potential factors affecting occurrence of cardiovascular diseases. In allergic inflammation, T2 cytokines drive macrophage polarization towards M2 profiles, which promote airway inflammation and remodelling. M1 macrophages in transplantations seem to contribute to acute rejection, while M2 macrophages promote the fibrosis of the graft. The view of pro-inflammatory M1 macrophages and M2 macrophages suppressing inflammation seems to be an oversimplification because these cells exploit very high level of plasticity and represent a large scale of different immunophenotypes with overlapping properties. In this respect, it would be more precise to describe macrophages as M1-like and M2-like.
Collapse
Affiliation(s)
- Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Robin Bartolini
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, U.K
| | - Rene Novysedlak
- Third Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Eva Cecrdlova
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Lily Koumbas Foley
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, U.K
| | - Ilja Striz
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
27
|
Dart SJ, Prosser AC, Huang WH, Liu L, Lucas AD, Delriviere L, Gaudieri S, Jeffrey GP, Lucas M. Subset-specific Retention of Donor Myeloid Cells After Major Histocompatibility Complex-matched and Mismatched Liver Transplantation. Transplantation 2023; 107:1502-1512. [PMID: 36584373 PMCID: PMC10508270 DOI: 10.1097/tp.0000000000004481] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 10/14/2022] [Accepted: 11/03/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND During solid organ transplantation, donor leukocytes, including myeloid cells, are transferred within the organ to the recipient. Both tolerogenic and alloreactive roles have been attributed to donor myeloid cells; however, their subset-specific retention posttransplantation has not been investigated in detail. METHODS Major histocompatibility complex (MHC)-matched and mismatched liver transplants were performed in mice, and the fate of donor and recipient myeloid cells was assessed. RESULTS Following MHC-matched transplantation, a proportion of donor myeloid cells was retained in the graft, whereas others egressed and persisted in the blood, spleen, and bone marrow but not the lymph nodes. In contrast, after MHC-mismatched transplantation, all donor myeloid cells, except Kupffer cells, were depleted. This depletion was caused by recipient T and B cells because all donor myeloid subsets were retained in MHC-mismatched grafts when recipients lacked T and B cells. Recipient myeloid cells rapidly infiltrated MHC-matched and, to a greater extent, MHC-mismatched liver grafts. MHC-mismatched grafts underwent a transient rejection episode on day 7, coinciding with a transition in macrophages to a regulatory phenotype, after which rejection resolved. CONCLUSIONS Phenotypic and kinetic differences in the myeloid cell responses between MHC-matched and mismatched grafts were identified. A detailed understanding of the dynamics of immune responses to transplantation is critical to improving graft outcomes.
Collapse
Affiliation(s)
- Sarah J. Dart
- Medical School, The University of Western Australia, Perth, WA, Australia
| | - Amy C. Prosser
- Medical School, The University of Western Australia, Perth, WA, Australia
| | - Wen Hua Huang
- Medical School, The University of Western Australia, Perth, WA, Australia
- Western Australian Liver Transplant Service, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Liu Liu
- Medical School, The University of Western Australia, Perth, WA, Australia
| | - Andrew D. Lucas
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Luc Delriviere
- Medical School, The University of Western Australia, Perth, WA, Australia
- Western Australian Liver Transplant Service, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Silvana Gaudieri
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Gary P. Jeffrey
- Medical School, The University of Western Australia, Perth, WA, Australia
- Western Australian Liver Transplant Service, Sir Charles Gairdner Hospital, Perth, WA, Australia
- Department of Hepatology, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Michaela Lucas
- Medical School, The University of Western Australia, Perth, WA, Australia
- Department of Immunology, Sir Charles Gairdner Hospital and PathWest Laboratory Medicine, Perth, WA, Australia
| |
Collapse
|
28
|
Pham HL, Hoang TX, Kim JY. Human Regulatory Macrophages Derived from THP-1 Cells Using Arginylglycylaspartic Acid and Vitamin D3. Biomedicines 2023; 11:1740. [PMID: 37371835 DOI: 10.3390/biomedicines11061740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/15/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Regulatory macrophages (Mregs) are unique in that they have anti-inflammatory and immunosuppressive properties. Thus, treating inflammatory diseases using Mregs is an area of active research. Human Mregs are usually generated by culturing peripheral blood monocytes stimulated using a macrophage colony-stimulating factor with interferon (IFN)-γ. Herein, we generated Mregs with an elongated cell morphology from THP-1 cells that were stimulated with phorbol 12-myristate 13-acetate and cultured with both arginylglycylaspartic acid and vitamin D3. These Mregs regulated macrophage function, and respectively downregulated and upregulated the expression of pro-inflammatory and immunosuppressive mediators. They also expressed Mregs-specific markers, such as dehydrogenase/reductase 9, even when exposed to such inflammatory stimulants as IFN-γ, lipopolysaccharide, purified xenogeneic antigen, and xenogeneic cells. The Mregs also exerted anti-inflammatory and anticoagulatory actions in response to xenogeneic cells, as well as exerting immunosuppressive effects on mitogen-induced Jurkat T-cell proliferation. Our method of generating functional Mregs in vitro without cytokines is simple and cost-effective.
Collapse
Affiliation(s)
- Hoang Lan Pham
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Thi Xoan Hoang
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Jae Young Kim
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| |
Collapse
|
29
|
Lackner K, Ebner S, Watschinger K, Maglione M. Multiple Shades of Gray-Macrophages in Acute Allograft Rejection. Int J Mol Sci 2023; 24:8257. [PMID: 37175964 PMCID: PMC10179242 DOI: 10.3390/ijms24098257] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/27/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Long-term results following solid organ transplantation do not mirror the excellent short-term results achieved in recent decades. It is therefore clear that current immunosuppressive maintenance protocols primarily addressing the adaptive immune system no longer meet the required clinical need. Identification of novel targets addressing this shortcoming is urgently needed. There is a growing interest in better understanding the role of the innate immune system in this context. In this review, we focus on macrophages, which are known to prominently infiltrate allografts and, during allograft rejection, to be involved in the surge of the adaptive immune response by expression of pro-inflammatory cytokines and direct cytotoxicity. However, this active participation is janus-faced and unspecific targeting of macrophages may not consider the different subtypes involved. Under this premise, we give an overview on macrophages, including their origins, plasticity, and important markers. We then briefly describe their role in acute allograft rejection, which ranges from sustaining injury to promoting tolerance, as well as the impact of maintenance immunosuppressants on macrophages. Finally, we discuss the observed immunosuppressive role of the vitamin-like compound tetrahydrobiopterin and the recent findings that suggest the innate immune system, particularly macrophages, as its target.
Collapse
Affiliation(s)
- Katharina Lackner
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.L.); (S.E.)
| | - Susanne Ebner
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.L.); (S.E.)
| | - Katrin Watschinger
- Institute of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Manuel Maglione
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.L.); (S.E.)
- Department of Visceral, Transplant, and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
30
|
Ott LC, Cuenca AG. Innate immune cellular therapeutics in transplantation. FRONTIERS IN TRANSPLANTATION 2023; 2:1067512. [PMID: 37994308 PMCID: PMC10664839 DOI: 10.3389/frtra.2023.1067512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Successful organ transplantation provides an opportunity to extend the lives of patients with end-stage organ failure. Selectively suppressing the donor-specific alloimmune response, however, remains challenging without the continuous use of non-specific immunosuppressive medications, which have multiple adverse effects including elevated risks of infection, chronic kidney injury, cardiovascular disease, and cancer. Efforts to promote allograft tolerance have focused on manipulating the adaptive immune response, but long-term allograft survival rates remain disappointing. In recent years, the innate immune system has become an attractive therapeutic target for the prevention and treatment of transplant organ rejection. Indeed, contemporary studies demonstrate that innate immune cells participate in both the initial alloimmune response and chronic allograft rejection and undergo non-permanent functional reprogramming in a phenomenon termed "trained immunity." Several types of innate immune cells are currently under investigation as potential therapeutics in transplantation, including myeloid-derived suppressor cells, dendritic cells, regulatory macrophages, natural killer cells, and innate lymphoid cells. In this review, we discuss the features and functions of these cell types, with a focus on their role in the alloimmune response. We examine their potential application as therapeutics to prevent or treat allograft rejection, as well as challenges in their clinical translation and future directions for investigation.
Collapse
Affiliation(s)
- Leah C Ott
- Department of General Surgery, Boston Children's Hospital, Boston, MA, United States
| | - Alex G Cuenca
- Department of General Surgery, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
31
|
Pham HL, Yang DH, Chae WR, Jung JH, Hoang TX, Lee NY, Kim JY. PDMS Micropatterns Coated with PDA and RGD Induce a Regulatory Macrophage-like Phenotype. MICROMACHINES 2023; 14:673. [PMID: 36985080 PMCID: PMC10052727 DOI: 10.3390/mi14030673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 06/18/2023]
Abstract
Regulatory macrophages (Mreg) are a special cell type that present a potential therapeutic strategy for various inflammatory diseases. In vitro, Mreg generation mainly takes 7-10 days of treatment with chemicals, including cytokines. In the present study, we established a new approach for Mreg generation using a three-dimensional (3D) micropatterned polydimethylsiloxane (PDMS) surface coated with a natural biopolymer adhesive polydopamine (PDA) and the common cell adhesion peptide motif arginylglycylaspartic acid (RGD). The 3D PDMS surfaces were fabricated by photolithography and soft lithography techniques and were subsequently coated with an RGD+PDA mixture to form a surface that facilitates cell adhesion. Human monocytes (THP-1 cells) were cultured on different types of 2D or 3D micropatterns for four days, and the cell morphology, elongation, and Mreg marker expression were assessed using microscopic and flow cytometric analyses. The cells grown on the PDA+RGD-coated 3D micropatterns (20-µm width/20-µm space) exhibited the most elongated morphology and strongest expression levels of Mreg markers, such as CD163, CD206, CD209, CD274, MER-TK, TREM2, and DHRS9. The present study demonstrated that PDA+RGD-coated 3D PDMS micropatterns successfully induced Mreg-like cells from THP-1 cells within four days without the use of cytokines, suggesting a time- and cost-effective method to generate Mreg-like cells in vitro.
Collapse
Affiliation(s)
- Hoang Lan Pham
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Da Hyun Yang
- Department of BioNano Technology, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Woo Ri Chae
- Department of BioNano Technology, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Jong Hyeok Jung
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Thi Xoan Hoang
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Nae Yoon Lee
- Department of BioNano Technology, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| | - Jae Young Kim
- Department of Life Science, Gachon University, Seongnam 13120, Gyeonggi-Do, Republic of Korea
| |
Collapse
|
32
|
Mistarz A, Winkler M, Battaglia S, Liu S, Hutson A, Rokita H, Gambotto A, Odunsi KO, Singh PK, McGray AR, Wang J, Kozbor D. Reprogramming the tumor microenvironment leverages CD8 + T cell responses to a shared tumor/self antigen in ovarian cancer. Mol Ther Oncolytics 2023; 28:230-248. [PMID: 36875325 PMCID: PMC9982455 DOI: 10.1016/j.omto.2023.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Tumor antigen-driven responses to weakly immunogenic self-antigens and neoantigens directly affect treatment efficacy following immunotherapy. Using orthotopically grown SV40 T antigen+ ovarian carcinoma in antigen-naive wild-type or TgMISIIR-TAg-Low transgenic mice expressing SV40 T antigen as a self-antigen, we investigated the impact of CXCR4-antagonist-armed oncolytic virotherapy on tumor progression and antitumor immunity. Immunostaining and single-cell RNA sequencing analyses of the peritoneal tumor microenvironment of untreated tumors in syngeneic wild-type mice revealed the presence of SV40 T antigen-specific CD8+ T cells, a balanced M1/M2 transcriptomic signature of tumor-associated macrophages, and immunostimulatory cancer-associated fibroblasts. This contrasted with polarized M2 tumor-associated macrophages, immunosuppressive cancer-associated fibroblasts, and poor immune activation in TgMISIIR-TAg-Low mice. Intraperitoneal delivery of CXCR4-antagonist-armed oncolytic vaccinia virus led to nearly complete depletion of cancer-associated fibroblasts, M1 polarization of macrophages, and generation of SV40 T antigen-specific CD8+ T cells in transgenic mice. Cell depletion studies revealed that the therapeutic effect of armed oncolytic virotherapy was dependent primarily on CD8+ cells. These results demonstrate that targeting the interaction between immunosuppressive cancer-associated fibroblasts and macrophages in the tolerogenic tumor microenvironment by CXCR4-A-armed oncolytic virotherapy induces tumor/self-specific CD8+ T cell responses and consequently increases therapeutic efficacy in an immunocompetent ovarian cancer model.
Collapse
Affiliation(s)
- Anna Mistarz
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Marta Winkler
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Sebastiano Battaglia
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Alan Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hanna Rokita
- Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Kunle O. Odunsi
- University of Chicago Comprehensive Cancer Center, Chicago, IL 60637, USA
| | - Prashant K. Singh
- Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - A.J. Robert McGray
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Danuta Kozbor
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
33
|
Kervella D, Mesnard B, Prudhomme T, Bruneau S, Masset C, Cantarovich D, Blancho G, Branchereau J. Sterile Pancreas Inflammation during Preservation and after Transplantation. Int J Mol Sci 2023; 24:ijms24054636. [PMID: 36902067 PMCID: PMC10003374 DOI: 10.3390/ijms24054636] [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: 12/24/2022] [Revised: 02/23/2023] [Accepted: 02/26/2023] [Indexed: 03/04/2023] Open
Abstract
The pancreas is very susceptible to ischemia-reperfusion injury. Early graft losses due to pancreatitis and thrombosis represent a major issue after pancreas transplantation. Sterile inflammation during organ procurement (during brain death and ischemia-reperfusion) and after transplantation affects organ outcomes. Sterile inflammation of the pancreas linked to ischemia-reperfusion injury involves the activation of innate immune cell subsets such as macrophages and neutrophils, following tissue damage and release of damage-associated molecular patterns and pro-inflammatory cytokines. Macrophages and neutrophils favor tissue invasion by other immune cells, have deleterious effects or functions, and promote tissue fibrosis. However, some innate cell subsets may promote tissue repair. This outburst of sterile inflammation promotes adaptive immunity activation via antigen exposure and activation of antigen-presenting cells. Better controlling sterile inflammation during pancreas preservation and after transplantation is of utmost interest in order to decrease early allograft loss (in particular thrombosis) and increase long-term allograft survival. In this regard, perfusion techniques that are currently being implemented represent a promising tool to decrease global inflammation and modulate the immune response.
Collapse
Affiliation(s)
- Delphine Kervella
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Néphrologie et Immunologie Clinique, ITUN, F-44000 Nantes, France
- Correspondence:
| | - Benoît Mesnard
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Service d’Urologie, ITUN, F-44000 Nantes, France
| | - Thomas Prudhomme
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Sarah Bruneau
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
| | - Christophe Masset
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Néphrologie et Immunologie Clinique, ITUN, F-44000 Nantes, France
| | - Diego Cantarovich
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Néphrologie et Immunologie Clinique, ITUN, F-44000 Nantes, France
| | - Gilles Blancho
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Néphrologie et Immunologie Clinique, ITUN, F-44000 Nantes, France
| | - Julien Branchereau
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France
- Centre Hospitalier Universitaire de Nantes, Nantes Université, Service d’Urologie, ITUN, F-44000 Nantes, France
| |
Collapse
|
34
|
McGovern KE, Sonar SA, Watanabe M, Coplen CP, Bradshaw CM, Nikolich JŽ. The aging of the immune system and its implications for transplantation. GeroScience 2023:10.1007/s11357-022-00720-2. [PMID: 36626019 PMCID: PMC9838392 DOI: 10.1007/s11357-022-00720-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
By the last third of life, most mammals, including humans, exhibit a decline in immune cell numbers, immune organ structure, and immune defense of the organism, commonly known as immunosenescence. This decline leads to clinical manifestations of increased susceptibility to infections, particularly those caused by emerging and reemerging microorganisms, which can reach staggering levels-infection with SARS-CoV-2 has been 270-fold more lethal to older adults over 80 years of age, compared to their 18-39-year-old counterparts. However, while this would be expected to be beneficial to situations where hyporeactivity of the immune system may be desirable, this is not always the case. Here, we discuss the cellular and molecular underpinnings of immunosenescence as they pertain to outcomes of solid organ and hematopoietic transplantation.
Collapse
Affiliation(s)
- Kathryn E McGovern
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Sandip A Sonar
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Makiko Watanabe
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Christopher P Coplen
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Christine M Bradshaw
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA.
- Arizona Center On Aging, The University of Arizona, University of Arizona College of Medicine-Tucson, Tucson, AZ, 85724, USA.
- BIO5 Institute, University of Arizona, Tucson, AZ, USA.
- The Aegis Consortium for Pandemic-free Future, University of Arizona Health Sciences, University of Arizona, Tucson, 85719, USA.
| |
Collapse
|
35
|
Choi H, Yang SW, Joo JS, Park M, Jin Y, Kim JW, Lee SY, Lee SV, Yun TJ, Cho ML, Hwang HS, Kang YS. Sialylated IVIg binding to DC-SIGN + Hofbauer cells induces immune tolerance through the caveolin-1/NF-kB pathway and IL-10 secretion. Clin Immunol 2023; 246:109215. [PMID: 36581222 DOI: 10.1016/j.clim.2022.109215] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/15/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Although the use of IVIg has increased in various immune-driven diseases and even in pregnancy, the exact action mechanisms of IVIg are not fully understood. Dendritic cell-specific intercellular adhesion molecule-3 grabbing non-integrin (DC-SIGN) is a known receptor for α-2,6-sialylated IgG (sIVIg), which is responsible for the anti-inflammatory effect of IVIg. DC-SIGN is expressed on Hofbauer cells (HBCs) of the fetal villi of the placenta which act as an innate immune modulator at the maternal-fetal interface. Preeclampsia is a major complication in pregnancy and is related to IL-10, a cytokine with an important role in immune tolerance. DC-SIGN interaction with sIVIg in HBCs promoted IL-10 secretion through the activation of the caveolin-1/NF-κB pathway, especially in plasma lipid rafts. Consistent results were obtained for HBCs from patients with preeclampsia. Collectively, the stimulation of DC-SIGN+ HBCs with sIVIg enhanced immune tolerance in the feto-maternal environment, suggesting the therapeutic application of sIVIg to prevent preeclampsia.
Collapse
Affiliation(s)
- Hyeongjwa Choi
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Seung-Woo Yang
- Department of Obstetrics and Gynecology, Sang-Gye Paik Hospital, Inje University School of Medicine; Seoul 01757, Republic of Korea
| | - Jin-Soo Joo
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University; 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Min Park
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Yihua Jin
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Ji-Woon Kim
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Seon-Yeong Lee
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Vin Lee
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University; 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Tae-Jin Yun
- Department of Pathology, New York University Grossman School of Medicine; New York, NY 10016, USA
| | - Mi-La Cho
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, South Korea
| | - Han-Sung Hwang
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine; Seoul, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Young-Sun Kang
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University; 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; KU Research Center for Zoonosis, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea.
| |
Collapse
|
36
|
Muacevic A, Adler JR. Classic and Current Opinions in Human Organ and Tissue Transplantation. Cureus 2022; 14:e30982. [PMID: 36337306 PMCID: PMC9624478 DOI: 10.7759/cureus.30982] [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] [Accepted: 11/01/2022] [Indexed: 11/30/2022] Open
Abstract
Graft tolerance is a pathophysiological condition heavily reliant on the dynamic interaction of the innate and adaptive immune systems. Genetic polymorphism determines immune responses to tissue/organ transplantation, and intricate humoral and cell-mediated mechanisms control these responses. In transplantation, the clinician's goal is to achieve a delicate equilibrium between the allogeneic immune response, undesired effects of the immunosuppressive drugs, and the existing morbidities that are potentially life-threatening. Transplant immunopathology involves sensitization, effector, and apoptosis phases which recruit and engages immunological cells like natural killer cells, lymphocytes, neutrophils, and monocytes. Similarly, these cells are involved in the transfer of normal or genetically engineered T cells. Advances in tissue transplantation would involve a profound knowledge of the molecular mechanisms that underpin the respective immunopathology involved and the design of precision medicines that are safe and effective.
Collapse
|
37
|
Zhang X, Hong R, Bei L, Hu Z, Yang X, Song T, Chen L, Meng H, Niu G, Ke C. SELENBP1 inhibits progression of colorectal cancer by suppressing epithelial–mesenchymal transition. Open Med (Wars) 2022; 17:1390-1404. [PMID: 36117772 PMCID: PMC9438969 DOI: 10.1515/med-2022-0532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 06/19/2022] [Accepted: 07/11/2022] [Indexed: 01/13/2023] Open
Abstract
Selenium-binding protein 1 (SELENBP1) is frequently dysregulated in various malignancies including colorectal cancer (CRC); however, its roles in progression of CRCs and the underlying mechanism remain to be elucidated. In this study, we compared the expression of SELENBP1 between CRCs and colorectal normal tissues (NTs), as well as between primary and metastatic CRCs; we determined the association between SELENBP1 expression and CRC patient prognoses; we conducted both in vitro and in vivo experiments to explore the functional roles of SELENBP1 in CRC progression; and we characterized the potential underlying mechanisms associated with SELENBP1 activities. We found that the expression of SELENBP1 was significantly and consistently decreased in CRCs than that in adjacent NTs, while significantly and frequently decreased in metastatic than primary CRCs. High expression of SELENBP1 was an independent predictor of favorable prognoses in CRC patients. Overexpression of SELENBP1 suppressed, while silencing of SELENBP1 promoted cell proliferation, migration and invasion, and in vivo tumorigenesis of CRC. Mechanically, SELENBP1 may suppress CRC progression by inhibiting the epithelial–mesenchymal transition.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, P.R. China
| | - Runqi Hong
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, P.R. China
| | - Lanxin Bei
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhiqing Hu
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, P.R. China
| | - Ximin Yang
- Department of Radiology, Dongying New District Hospital, Dongying, Shandong Province, 257000, P.R. China
| | - Tao Song
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Liang Chen
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, 200240, P.R. China
| | - He Meng
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Gengming Niu
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| | - Chongwei Ke
- Department of General Surgery, Shanghai Fifth People’s Hospital, Fudan University, 801 Heqing Road, Minhang District, Shanghai, 200240, P.R. China
| |
Collapse
|
38
|
Kopecky BJ, Dun H, Amrute JM, Lin CY, Bredemeyer AL, Terada Y, Bayguinov PO, Koenig AL, Frye CC, Fitzpatrick JAJ, Kreisel D, Lavine KJ. Donor Macrophages Modulate Rejection After Heart Transplantation. Circulation 2022; 146:623-638. [PMID: 35880523 PMCID: PMC9398940 DOI: 10.1161/circulationaha.121.057400] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 06/07/2022] [Indexed: 01/23/2023]
Abstract
BACKGROUND Cellular rejection after heart transplantation imparts significant morbidity and mortality. Current immunosuppressive strategies are imperfect, target recipient T cells, and have adverse effects. The innate immune response plays an essential role in the recruitment and activation of T cells. Targeting the donor innate immune response would represent the earliest interventional opportunity within the immune response cascade. There is limited knowledge about donor immune cell types and functions in the setting of cardiac transplantation, and no current therapeutics exist for targeting these cell populations. METHODS Using genetic lineage tracing, cell ablation, and conditional gene deletion, we examined donor mononuclear phagocyte diversity and macrophage function during acute cellular rejection of transplanted hearts in mice. We performed single-cell RNA sequencing on donor and recipient macrophages and monocytes at multiple time points after transplantation. On the basis of our imaging and single-cell RNA sequencing data, we evaluated the functional relevance of donor CCR2+ (C-C chemokine receptor 2) and CCR2- macrophages using selective cell ablation strategies in donor grafts before transplant. Last, we performed functional validation that donor macrophages signal through MYD88 (myeloid differentiation primary response protein 88) to facilitate cellular rejection. RESULTS Donor macrophages persisted in the rejecting transplanted heart and coexisted with recipient monocyte-derived macrophages. Single-cell RNA sequencing identified donor CCR2+ and CCR2- macrophage populations and revealed remarkable diversity among recipient monocytes, macrophages, and dendritic cells. Temporal analysis demonstrated that donor CCR2+ and CCR2- macrophages were transcriptionally distinct, underwent significant morphologic changes, and displayed unique activation signatures after transplantation. Although selective depletion of donor CCR2- macrophages reduced allograft survival, depletion of donor CCR2+ macrophages prolonged allograft survival. Pathway analysis revealed that donor CCR2+ macrophages are activated through MYD88/nuclear factor kappa light chain enhancer of activated B cells signaling. Deletion of MYD88 in donor macrophages resulted in reduced antigen-presenting cell recruitment, reduced ability of antigen-presenting cells to present antigen to T cells, decreased emergence of allograft-reactive T cells, and extended allograft survival. CONCLUSIONS Distinct populations of donor and recipient macrophages coexist within the transplanted heart. Donor CCR2+ macrophages are key mediators of allograft rejection, and deletion of MYD88 signaling in donor macrophages is sufficient to suppress rejection and extend allograft survival. This highlights the therapeutic potential of donor heart-based interventions.
Collapse
Affiliation(s)
- Benjamin J Kopecky
- Cardiovascular Division, Department of Medicine, Washington
University School of Medicine, St. Louis, Missouri, USA
| | - Hao Dun
- Department of Surgery, Washington University School of
Medicine, Saint Louis, Missouri, USA
| | - Junedh M Amrute
- Cardiovascular Division, Department of Medicine, Washington
University School of Medicine, St. Louis, Missouri, USA
| | - Chieh-Yu Lin
- Department of Pathology and Immunology, Washington
University School of Medicine, Saint Louis, Missouri, USA
| | - Andrea L Bredemeyer
- Cardiovascular Division, Department of Medicine, Washington
University School of Medicine, St. Louis, Missouri, USA
| | - Yuriko Terada
- Department of Surgery, Washington University School of
Medicine, Saint Louis, Missouri, USA
| | - Peter O Bayguinov
- Washington University Center for Cellular Imaging,
Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrew L Koenig
- Cardiovascular Division, Department of Medicine, Washington
University School of Medicine, St. Louis, Missouri, USA
| | - Christian C Frye
- Department of Surgery, Washington University School of
Medicine, Saint Louis, Missouri, USA
| | - James AJ Fitzpatrick
- Washington University Center for Cellular Imaging,
Washington University School of Medicine, St. Louis, Missouri, USA
- Departments of Neuroscience and Cell Biology &
Physiology, Washington University School of Medicine, Saint Louis, Missouri,
USA
| | - Daniel Kreisel
- Department of Surgery, Washington University School of
Medicine, Saint Louis, Missouri, USA
- Department of Pathology and Immunology, Washington
University School of Medicine, Saint Louis, Missouri, USA
| | - Kory J Lavine
- Cardiovascular Division, Department of Medicine, Washington
University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington
University School of Medicine, Saint Louis, Missouri, USA
- Department of Developmental Biology, Washington University
School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
39
|
Arnold JN, Mitchell DA. Tinker, tailor, soldier, cell: the role of C-type lectins in the defense and promotion of disease. Protein Cell 2022; 14:4-16. [PMID: 36726757 PMCID: PMC9871964 DOI: 10.1093/procel/pwac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
C-type lectins (CTLs) represent a large family of soluble and membrane-bound proteins which bind calcium dependently via carbohydrate recognition domains (CRDs) to glycan residues presented on the surface of a variety of pathogens. The deconvolution of a cell's glycan code by CTLs underpins several important physiological processes in mammals such as pathogen neutralization and opsonization, leukocyte trafficking, and the inflammatory response. However, as our knowledge of CTLs has developed it has become apparent that the role of this innate immune family of proteins can be double-edged, where some pathogens have developed approaches to subvert and exploit CTL interactions to promote infection and sustain the pathological state. Equally, CTL interactions with host glycoproteins can contribute to inflammatory diseases such as arthritis and cancer whereby, in certain contexts, they exacerbate inflammation and drive malignant progression. This review discusses the 'dual agent' roles of some of the major mammalian CTLs in both resolving and promoting infection, inflammation and inflammatory disease and highlights opportunities and emerging approaches for their therapeutic modulation.
Collapse
|
40
|
Campos‐Mora M, De Solminihac J, Rojas C, Padilla C, Kurte M, Pacheco R, Kaehne T, Wyneken Ú, Pino‐Lagos K. Neuropilin-1 is present on Foxp3+ T regulatory cell-derived small extracellular vesicles and mediates immunity against skin transplantation. J Extracell Vesicles 2022; 11:e12237. [PMID: 35676234 PMCID: PMC9177693 DOI: 10.1002/jev2.12237] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/16/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022] Open
Abstract
Among the mechanisms of suppression that T regulatory (Treg) cells exert to control the immune responses, the secretion of small extracellular vesicles (sEV) has been recently proposed as a novel contact-independent immunomodulatory mechanism. Previous studies have demonstrated that Treg cells produce sEV, including exosomes, able to modulate the effector function of CD4+ T cells, and antigen presenting cells (APCs) such as dendritic cells (DCs) through the transfer of microRNA, cytokines, the production of adenosine, among others. Previously, we have demonstrated that Neuropilin-1 (Nrp1) is required for Tregs-mediated immunosuppression mainly by impacting on the phenotype and function of effector CD4+ T cells. Here, we show that Foxp3+ Treg cells secrete sEV, which bear Nrp1 in their membrane. These sEV modulate effector CD4+ T cell phenotype and proliferation in vitro in a Nrp1-dependent manner. Proteomic analysis indicated that sEV obtained from wild type (wt) and Nrp1KO Treg cells differed in proteins related to immune tolerance, finding less representation of CD73 and Granzyme B in sEV obtained from Nrp1KO Treg cells. Likewise, we show that Nrp1 is required in Treg cell-derived sEV for inducing skin transplantation tolerance, since a reduction in graft survival and an increase on M1/M2 ratio were found in animals treated with Nrp1KO Treg cell-derived sEV. Altogether, this study describes for the first time that Treg cells secrete sEV containing Nrp1 and that this protein, among others, is necessary to promote transplantation tolerance in vivo via sEV local administration.
Collapse
Affiliation(s)
- Mauricio Campos‐Mora
- Centro de Investigación e Innovación BiomédicaFacultad de MedicinaUniversidad de los AndesSantiagoChile
| | - Javiera De Solminihac
- Centro de Investigación e Innovación BiomédicaFacultad de MedicinaUniversidad de los AndesSantiagoChile
| | - Carolina Rojas
- Periodontal Biology LaboratoryFaculty of DentistryUniversidad de ChileSantiagoChile
| | - Cristina Padilla
- Centro de Investigación e Innovación BiomédicaFacultad de MedicinaUniversidad de los AndesSantiagoChile
| | - Mónica Kurte
- Centro de Investigación e Innovación BiomédicaFacultad de MedicinaUniversidad de los AndesSantiagoChile
| | - Rodrigo Pacheco
- Laboratorio de NeuroinmunologíaCentro Ciencia & VidaFundación Ciencia & VidaSantiagoChile
| | - Thilo Kaehne
- Institute of Experimental MedicineMedical FacultyOtto von Guericke UniversityMagdeburgGermany
| | - Úrsula Wyneken
- Centro de Investigación e Innovación BiomédicaFacultad de MedicinaUniversidad de los AndesSantiagoChile
| | - Karina Pino‐Lagos
- Centro de Investigación e Innovación BiomédicaFacultad de MedicinaUniversidad de los AndesSantiagoChile
| |
Collapse
|
41
|
Zhu Y, Dun H, Ye L, Terada Y, Shriver LP, Patti GJ, Kreisel D, Gelman AE, Wong BW. Targeting fatty acid β-oxidation impairs monocyte differentiation and prolongs heart allograft survival. JCI Insight 2022; 7:e151596. [PMID: 35239515 PMCID: PMC9057610 DOI: 10.1172/jci.insight.151596] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 03/02/2022] [Indexed: 11/17/2022] Open
Abstract
Monocytes play an important role in the regulation of alloimmune responses after heart transplantation (HTx). Recent studies have highlighted the importance of immunometabolism in the differentiation and function of myeloid cells. While the importance of glucose metabolism in monocyte differentiation and function has been reported, a role for fatty acid β-oxidation (FAO) has not been explored. Heterotopic HTx was performed using hearts from BALB/c donor mice implanted into C57BL/6 recipient mice and treated with etomoxir (eto), an irreversible inhibitor of carnitine palmitoyltransferase 1 (Cpt1), a rate-limiting step of FAO, or vehicle control. FAO inhibition prolonged HTx survival, reduced early T cell infiltration/activation, and reduced DC and macrophage infiltration to heart allografts of eto-treated recipients. ELISPOT demonstrated that splenocytes from eto-treated HTx recipients were less reactive to activated donor antigen-presenting cells. FAO inhibition reduced monocyte-to-DC and monocyte-to-macrophage differentiation in vitro and in vivo. FAO inhibition did not alter the survival of heart allografts when transplanted into Ccr2-deficient recipients, suggesting that the effects of FAO inhibition were dependent on monocyte mobilization. Finally, we confirmed the importance of FAO on monocyte differentiation in vivo using conditional deletion of Cpt1a. Our findings demonstrate that targeting FAO attenuates alloimmunity after HTx, in part through impairing monocyte differentiation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daniel Kreisel
- Department of Surgery
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrew E. Gelman
- Department of Surgery
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
42
|
Brilland B, Laplante P, Thebault P, Geoffroy K, Brissette MJ, Latour M, Chassé M, Qi S, Hébert MJ, Cardinal H, Cailhier JF. MFG-E8 Reduces Aortic Intimal Proliferation in a Murine Model of Transplant Vasculopathy. Int J Mol Sci 2022; 23:ijms23084094. [PMID: 35456911 PMCID: PMC9027378 DOI: 10.3390/ijms23084094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 12/10/2022] Open
Abstract
Transplant vasculopathy is characterized by endothelial apoptosis, which modulates the local microenvironment. Milk fat globule epidermal growth factor 8 (MFG-E8), which is released by apoptotic endothelial cells, limits tissue damage and inflammation by promoting anti-inflammatory macrophages. We aimed to study its role in transplant vasculopathy using the murine aortic allotransplantation model. BALB/c mice were transplanted with fully mismatched aortic transplants from MFG-E8 knockout (KO) or wild type (WT) C57BL/6J mice. Thereafter, mice received MFG-E8 (or vehicle) injections for 9 weeks prior to histopathological analysis of allografts for intimal proliferation (hematoxylin and eosin staining) and leukocyte infiltration assessment (immunofluorescence). Phenotypes of blood leukocytes and humoral responses were also evaluated (flow cytometry and ELISA). Mice receiving MFG-E8 KO aortas without MFG-E8 injections had the most severe intimal proliferation (p < 0.001). Administration of MFG-E8 decreased intimal proliferation, especially in mice receiving MFG-E8 KO aortas. Administration of MFG-E8 also increased the proportion of anti-inflammatory macrophages among graft-infiltrating macrophages (p = 0.003) and decreased systemic CD4+ and CD8+ T-cell activation (p < 0.001). An increase in regulatory T cells occurred in both groups of mice receiving WT aortas (p < 0.01). Thus, the analarmin MFG-E8 appears to be an important protein for reducing intimal proliferation in this murine model of transplant vasculopathy. MFG-E8 effects are associated with intra-allograft macrophage reprogramming and systemic T-cell activation dampening.
Collapse
Affiliation(s)
- Benoit Brilland
- Service de Néphrologie-Dialyse-Transplantation, CHU d’Angers, F-49000 Angers, France;
- University of Angers, Université de Nantes, CHU Angers, INSERM, CRCINA, SFR ICAT, F-49000 Angers, France
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
| | - Patrick Laplante
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Pamela Thebault
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Karen Geoffroy
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Marie-Joëlle Brissette
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
| | - Mathieu Latour
- Department of Pathology, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 3J4, Canada;
| | - Michaël Chassé
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Department of Medicine, Critical Care Division, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 3J4, Canada
| | - Shijie Qi
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
| | - Marie-Josée Hébert
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Canadian National Transplant Research Program, Edmonton, AB T6G 2E1, Canada
- Department of Medicine, Renal Division, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 3J4, Canada
| | - Héloïse Cardinal
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Canadian National Transplant Research Program, Edmonton, AB T6G 2E1, Canada
- Department of Medicine, Renal Division, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 3J4, Canada
| | - Jean-François Cailhier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada; (P.L.); (P.T.); (K.G.); (M.-J.B.); (M.C.); (S.Q.); (M.-J.H.); (H.C.)
- Institut du Cancer de Montréal, Montréal, QC H2X 0A9, Canada
- Canadian National Transplant Research Program, Edmonton, AB T6G 2E1, Canada
- Department of Medicine, Renal Division, Centre Hospitalier de l’Université de Montréal, Montreal, QC H2X 3J4, Canada
- Correspondence: ; Tel.: +514-890-8000 (ext. 25971); Fax: +514-412-7938
| |
Collapse
|
43
|
Horwitz JK, Bin S, Fairchild RL, Keslar KS, Yi Z, Zhang W, Pavlov VI, Li Y, Madsen JC, Cravedi P, Heeger PS. Linking erythropoietin to regulatory T-cell-dependent allograft survival through myeloid cells. JCI Insight 2022; 7:158856. [PMID: 35389892 PMCID: PMC9220923 DOI: 10.1172/jci.insight.158856] [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: 02/01/2022] [Accepted: 04/06/2022] [Indexed: 12/03/2022] Open
Abstract
Erythropoietin (EPO) has multiple nonerythropoietic functions, including immune modulation, but EPO’s effects in transplantation remain incompletely understood. We tested the mechanisms linking EPO administration to prolongation of murine heterotopic heart transplantation using WT and conditional EPO receptor–knockout (EPOR-knockout) mice as recipients. In WT controls, peritransplant administration of EPO synergized with CTLA4-Ig to prolong allograft survival (P < 0.001), reduce frequencies of donor-reactive effector CD8+ T cells in the spleen (P < 0.001) and in the graft (P < 0.05), and increase frequencies and total numbers of donor-reactive Tregs (P < 0.01 for each) versus CTLA4-Ig alone. Studies performed in conditional EPOR-knockout recipients showed that each of these differences required EPOR expression in myeloid cells but not in T cells. Analysis of mRNA isolated from spleen monocytes showed that EPO/EPOR ligation upregulated macrophage-expressed, antiinflammatory, regulatory, and pro-efferocytosis genes and downregulated selected proinflammatory genes. Taken together, the data support the conclusion that EPO promotes Treg-dependent murine cardiac allograft survival by crucially altering the phenotype and function of macrophages. Coupled with our previous documentation that EPO promotes Treg expansion in humans, the data support the need for testing the addition of EPO to costimulatory blockade-containing immunosuppression regimens in an effort to prolong human transplant survival.
Collapse
Affiliation(s)
- Julian K Horwitz
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Sofia Bin
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Robert L Fairchild
- Department of Immunology, Cleveland Clinic, Cleveland, United States of America
| | - Karen S Keslar
- Department of Immunology, Cleveland Clinic, Cleveland, United States of America
| | - Zhengzi Yi
- Translational Transplant Research Center, Icahn School of medicine at Mount Sinai, New York, United States of America
| | - Weijia Zhang
- Translational Transplant Research Center, Icahn school of Medicine at Mount Sinai, New York, United States of America
| | - Vasile I Pavlov
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Yansui Li
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Joren C Madsen
- Department of Surgery, Massachusetts General Hospital, Boston, United States of America
| | - Paolo Cravedi
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Peter S Heeger
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| |
Collapse
|
44
|
Single-cell transcriptional profiling of murine conjunctival immune cells reveals distinct populations expressing homeostatic and regulatory genes. Mucosal Immunol 2022; 15:620-628. [PMID: 35361907 PMCID: PMC9262780 DOI: 10.1038/s41385-022-00507-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/15/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023]
Abstract
Immune cells in the exposed conjunctiva mucosa defend against environmental and microbial stresses. Expression profiling by single-cell RNA sequencing was performed to identify conjunctival immune cell populations expressing homeostatic and regulatory genes. Fourteen distinct clusters were identified, including myeloid cells (neutrophils, monocytes, macrophages), dendritic cells (DC), and lymphoid cells (B, T, γδT, ILC2, and NK) lineages. Novel neutrophil [lipocalin (Lcn2) high and low), and MHCIIlo macrophage (MP) clusters were identified. More than half of the cells map to myeloid and dendritic cell populations with differential expression profiles that include genes with homeostatic and regulatory functions: Serpinb2 (MHCIIlo macrophage), Apoe (monocyte), Cd209a (macrophage), Cst3 (cDC1), and IL4i1 in migratory DC (mDC). ILC2 expresses the goblet cell trophic factor IL-13. Suppressed inflammatory and activated anti-inflammatory/regulatory pathways were observed in certain myeloid and DC populations. Confocal immunolocalization of identity markers showed mDC (CCR7, FASCIN1) located on or within the conjunctival epithelium. Monocyte, macrophage, cDC1 and IL-13/IL-5+ ILC2 were located below the conjunctival epithelium and goblet cells. This study found distinct immune cell populations in the conjunctiva and identified cells expressing genes with known homeostatic and immunoregulatory functions.
Collapse
|
45
|
Fueyo-González F, McGinty M, Ningoo M, Anderson L, Cantarelli C, Andrea Angeletti, Demir M, Llaudó I, Purroy C, Marjanovic N, Heja D, Sealfon SC, Heeger PS, Cravedi P, Fribourg M. Interferon-β acts directly on T cells to prolong allograft survival by enhancing regulatory T cell induction through Foxp3 acetylation. Immunity 2022; 55:459-474.e7. [PMID: 35148827 PMCID: PMC8917088 DOI: 10.1016/j.immuni.2022.01.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 06/18/2021] [Accepted: 01/13/2022] [Indexed: 12/19/2022]
Abstract
Type I interferons (IFNs) are pleiotropic cytokines with potent antiviral properties that also promote protective T cell and humoral immunity. Paradoxically, type I IFNs, including the widely expressed IFNβ, also have immunosuppressive properties, including promoting persistent viral infections and treating T-cell-driven, remitting-relapsing multiple sclerosis. Although associative evidence suggests that IFNβ mediates these immunosuppressive effects by impacting regulatory T (Treg) cells, mechanistic links remain elusive. Here, we found that IFNβ enhanced graft survival in a Treg-cell-dependent murine transplant model. Genetic conditional deletion models revealed that the extended allograft survival was Treg cell-mediated and required IFNβ signaling on T cells. Using an in silico computational model and analysis of human immune cells, we found that IFNβ directly promoted Treg cell induction via STAT1- and P300-dependent Foxp3 acetylation. These findings identify a mechanistic connection between the immunosuppressive effects of IFNβ and Treg cells, with therapeutic implications for transplantation, autoimmunity, and malignancy.
Collapse
Affiliation(s)
- Francisco Fueyo-González
- Division of Nephrology, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Mitchell McGinty
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22903, USA
| | - Mehek Ningoo
- Division of Nephrology, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Lisa Anderson
- Division of Nephrology, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Chiara Cantarelli
- UO Nefrologia, Azienda Ospedaliero-Universitaria Parma, Parma, Italy
| | - Andrea Angeletti
- Division of Nephrology, Dialysis, Transplantation, IRCCS Giannina Gaslini, Genoa, Italy
| | - Markus Demir
- Department of Anesthesiology, University of Cologne, Cologne, Germany
| | - Inés Llaudó
- Division of Nephrology, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Carolina Purroy
- Department of Nephrology, Complejo Hospitalario de Navarra, Navarra, Spain
| | - Nada Marjanovic
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - David Heja
- Division of Nephrology, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Stuart C Sealfon
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Peter S Heeger
- Division of Nephrology, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Paolo Cravedi
- Division of Nephrology, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Miguel Fribourg
- Division of Nephrology, Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York City, NY, USA; Immunology Institute Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| |
Collapse
|
46
|
Xiaocui L, Wei H, Yunlang C, Zhenzhen Z, Min A. CSF-1-induced DC-SIGN + macrophages are present in the ovarian endometriosis. Reprod Biol Endocrinol 2022; 20:48. [PMID: 35260161 PMCID: PMC8903642 DOI: 10.1186/s12958-022-00901-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Researchers have found that macrophages are the predominant cells in the peritoneal fluid (PF) of endometriosis patients. CSF-1 has been found to accumulate in the lesions and PF of endometriosis patients, and CSF-1 induces THP-1-derived macrophages to polarize toward a CD169+ DC-SIGN+ phenotype. Does the cytokine CSF-1 induce monocytes to differentiate into macrophages with a DC-SIGN+ phenotype in endometriosis? METHODS The level of CSF-1 in the endometrium of control subjects, and the eutopic, and ectopic endometrium of endometriosis patients was evaluated by real-time polymerase chain reaction (qRT-PCR) and was determined by enzyme-linked immunosorbent assay (ELISA) in the PF of control and endometriosis patients. CSF-1 expression was examined with a MILLIPLEX MAP Mouse Cytokine/Chemokine Magnetic Bead Panel. DC-SIGN+ macrophages were detected by immunohistochemical staining of tissues and flow cytometric analysis of the PF of control subjects (N = 25) and endometriosis (N = 35) patients. The phenotypes and biological activities of CSF-1 -induced macrophages were compared in an in vitro coculture system with peripheral blood lymphocytes from control subjects. RESULTS In this study, we found that the proportion of DC-SIGN+ CD169+ macrophages was higher in the abdominal immune microenvironment of endometriosis patients. CSF-1 was primarily secreted from ectopic lesions and peritoneum in mice with endometriosis. In addition, CSF-1 induced the polarization of macrophages toward a DC-SIGN+ CD169+ phenotype; this effect was abolished by the addition of an anti-CSF-1R antibody. CSF-1 induced the generation of DC-SIGN+ macrophages, leading to a depressed status of peripheral blood lymphocytes, including a high percentage of Treg cells and a low percentage of CD8+ T cells. Similarly, blockade with the anti-CSF-1R antibody abrogated this biological effect. CONCLUSIONS This is the first study on the role of DC-SIGN+ macrophages in the immune microenvironment of endometriosis. Further study of the mechanism and biological activities of CSF-1-induced DC-SIGN+ macrophages will enhance our understanding of the physiology of endometriosis.
Collapse
Affiliation(s)
- Li Xiaocui
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, P.R. China
| | - Hong Wei
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, P.R. China
| | - Cai Yunlang
- Department of Obstetrics and Gynecology, Medical School, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Zheng Zhenzhen
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, P.R. China
| | - An Min
- Department of Obstetrics and Gynecology, Medical School, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
47
|
Yang H, Hu B. Immunological Perspective: Helicobacter pylori Infection and Gastritis. Mediators Inflamm 2022; 2022:2944156. [PMID: 35300405 PMCID: PMC8923794 DOI: 10.1155/2022/2944156] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori is a spiral-shaped gram-negative bacterium. Its infection is mainly transmitted via oral-oral and fecal-oral routes usually during early childhood. It can achieve persistent colonization by manipulating the host immune responses, which also causes mucosal damage and inflammation. H. pylori gastritis is an infectious disease and results in chronic gastritis of different severity in near all patients with infection. It may develop from acute/chronic inflammation, chronic atrophic gastritis, intestinal metaplasia, dysplasia, and intraepithelial neoplasia, eventually to gastric cancer. This review attempts to cover recent studies which provide important insights into how H. pylori causes chronic inflammation and what the characteristic is, which will immunologically explain H. pylori gastritis.
Collapse
Affiliation(s)
- Hang Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
48
|
Gan X, Gu J, Ju Z, Lu L. Diverse Roles of Immune Cells in Transplant Rejection and Immune Tolerance. ENGINEERING 2022; 10:44-56. [DOI: 10.1016/j.eng.2021.03.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
49
|
Peckert-Maier K, Schönberg A, Wild AB, Royzman D, Braun G, Stich L, Hadrian K, Tripal P, Cursiefen C, Steinkasserer A, Zinser E, Bock F. Pre-incubation of corneal donor tissue with sCD83 improves graft survival via the induction of alternatively activated macrophages and tolerogenic dendritic cells. Am J Transplant 2022; 22:438-454. [PMID: 34467638 DOI: 10.1111/ajt.16824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/02/2021] [Accepted: 08/22/2021] [Indexed: 01/25/2023]
Abstract
Immune responses reflect a complex interplay of cellular and extracellular components which define the microenvironment of a tissue. Therefore, factors that locally influence the microenvironment and re-establish tolerance might be beneficial to mitigate immune-mediated reactions, including the rejection of a transplant. In this study, we demonstrate that pre-incubation of donor tissue with the immune modulator soluble CD83 (sCD83) significantly improves graft survival using a high-risk corneal transplantation model. The induction of tolerogenic mechanisms in graft recipients was achieved by a significant upregulation of Tgfb, Foxp3, Il27, and Il10 in the transplant and an increase of regulatory dendritic cells (DCs), macrophages (Mφ), and T cells (Tregs) in eye-draining lymph nodes. The presence of sCD83 during in vitro DC and Mφ generation directed these cells toward a tolerogenic phenotype leading to reduced proliferation-stimulating activity in MLRs. Mechanistically, sCD83 induced a tolerogenic Mφ and DC phenotype, which favors Treg induction and significantly increased transplant survival after adoptive cell transfer. Conclusively, pre-incubation of corneal grafts with sCD83 significantly prolongs graft survival by modulating recipient Mφ and DCs toward tolerance and thereby establishing a tolerogenic microenvironment. This functional strategy of donor graft pre-treatment paves the way for new therapeutic options in the field of transplantation.
Collapse
Affiliation(s)
- Katrin Peckert-Maier
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Alfrun Schönberg
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Andreas B Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Dmytro Royzman
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gabriele Braun
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lena Stich
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Karina Hadrian
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Tripal
- Optical Imaging Centre, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Claus Cursiefen
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - Elisabeth Zinser
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Felix Bock
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
50
|
Abstract
Macrophages have emerged at the forefront of research in immunology and transplantation because of recent advances in basic science. New findings have illuminated macrophage populations not identified previously, expanded upon traditional macrophage phenotypes, and overhauled macrophage ontogeny. These advances have major implications for the field of transplant immunology. Macrophages are known to prime adaptive immune responses, perpetuate T-cell-mediated rejection and antibody-mediated rejection, and promote allograft fibrosis. In this review, macrophage phenotypes and their role in allograft injury of solid organ transplants will be discussed with an emphasis on kidney transplantation. Additionally, consideration will be given to the prospect of manipulating macrophage phenotypes as cell-based therapy. Innate immunity and macrophages represent important players in allograft injury and a promising target to improve transplant outcomes.
Collapse
Affiliation(s)
- Sarah E. Panzer
- Department of Medicine, Division of Nephrology, University of Wisconsin, Madison, WI, United States
| |
Collapse
|