1
|
Cheng HY, Anggelia MR, Lin CH. Unraveling the Roles of Macrophages in Vascularized Composite Allotransplantation. Biomedicines 2025; 13:1425. [PMID: 40564144 DOI: 10.3390/biomedicines13061425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 06/06/2025] [Accepted: 06/08/2025] [Indexed: 06/28/2025] Open
Abstract
The phenotypic heterogeneity and functional diversity of macrophages have been increasingly appreciated, particularly regarding their roles as innate immune cells in shaping transplantation outcomes. However, their functions in vascularized composite allotransplantation (VCA) remain underexplored. In this review, we first describe the development of macrophages and the heterogeneity of macrophage differentiation, then present current insights into macrophages' involvement across key stages of VCA, including ischemia-reperfusion injury at the peri-transplantation stage, and the outcomes following transplantation, including acute rejection, chronic rejection, and development of transplantation tolerance. The existing evidence supports that macrophages significantly influence both short- and long-term VCA graft survival. The presence of vascularized bone marrow within some VCA grafts further suggests the involvement of donor bone marrow-derived macrophage population and adds another layer of complexity to immune dynamics. Collectively, current understanding highlights the macrophage as a promising target for therapeutic intervention and warrants continued investigation into their diverse functions and potential for improving VCA outcomes.
Collapse
Affiliation(s)
- Hui-Yun Cheng
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Kweishan, Taoyuan 333, Taiwan
| | - Madonna Rica Anggelia
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Kweishan, Taoyuan 333, Taiwan
| | - Cheng-Hung Lin
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Kweishan, Taoyuan 333, Taiwan
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Kweishan, Taoyuan 333, Taiwan
| |
Collapse
|
2
|
Kronenberg K, Dormann F, Brosig A, Pamler I, Geissler EK, Burkhardt R, Offner R, Hutchinson JA, Haehnel V. Platelet apheresis with additive solution and plasma rinseback affects the cellular composition of LRS chamber products. Sci Rep 2025; 15:19923. [PMID: 40481081 PMCID: PMC12144167 DOI: 10.1038/s41598-025-04350-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
Human leukocyte concentrates recovered from leukocyte reduction system (LRS) chambers of the Trima Accel automated blood collection device are a by-product of platelet apheresis that is often used in research. The Trima Accel software was obligatorily updated in 2023 from version 6 to 7. Here, we investigated software-dependent differences in the cellular composition of LRS concentrates when performing apheresis with either plasma or platelet additive solution. When using plasma as suspension medium, the software update to Trima 7 led to higher fraction of B cells as revealed by flow cytometry. Compared to platelets-in-plasma collection, the total recovered volume and leukocyte density was significantly reduced when running Trima 7 software with platelet additive solution including a plasma rinseback. Moreover, the proportion of lymphocytes and monocytes in these products was lower, whereas the proportion of neutrophils and eosinophils was higher. Researchers working with leukocytes isolated from LRS chambers should be aware that performing apheresis with platelet additive solution alters the composition of their starting material, which could affect the interpretation of some experiments.
Collapse
Affiliation(s)
- Katharina Kronenberg
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.
| | - Frauke Dormann
- Institute of Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Andreas Brosig
- Institute of Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Irene Pamler
- Institute of Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Robert Offner
- Institute of Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| | - James A Hutchinson
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Viola Haehnel
- Institute of Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
3
|
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
|
4
|
Ma L, Fink J, Yao K, McDonald-Hyman C, Dougherty P, Koehn B, Blazar BR. Immunoregulatory iPSC-derived non-lymphoid progeny in autoimmunity and GVHD alloimmunity. Stem Cells 2025; 43:sxaf011. [PMID: 40103180 DOI: 10.1093/stmcls/sxaf011] [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: 12/19/2024] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
Non-lymphoid immunoregulatory cells, including mesenchymal stem cells (MSCs), myeloid-derived suppressor cells (MDSCs), regulatory macrophages (Mregs), and tolerogenic dendritic cells (Tol-DCs), play critical roles in maintaining immune homeostasis. However, their therapeutic application in autoimmune diseases and graft-versus-host disease (GVHD) has received comparatively less attention. Induced pluripotent stem cells (iPSCs) offer a promising platform for cell engineering, enabling superior quality control, scalable production, and large-scale in vitro expansion of iPSC-derived non-lymphoid immunoregulatory cells. These advances pave the way for their broader application in autoimmune disease and GVHD therapy. Recent innovations in iPSC differentiation protocols have facilitated the generation of these cell types with functional characteristics akin to their primary counterparts. This review explores the unique features and generation processes of iPSC-derived non-lymphoid immunoregulatory cells, their therapeutic potential in GVHD and autoimmune disease, and their progress toward clinical translation. It emphasizes the phenotypic and functional diversity within each cell type and their distinct effects on disease modulation. Despite these advancements, challenges persist in optimizing differentiation efficiency, ensuring functional stability, and bridging the gap to clinical application. By synthesizing current methodologies, preclinical findings, and translational efforts, this review underscores the transformative potential of iPSC-derived non-lymphoid immunoregulatory cells in advancing cell-based therapies for alloimmune and autoimmune diseases.
Collapse
Affiliation(s)
- Lie Ma
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN, United States
| | - Jordan Fink
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN, United States
| | - Ke Yao
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN, United States
| | - Cameron McDonald-Hyman
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Phillip Dougherty
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN, United States
| | - Brent Koehn
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN, United States
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, MN, United States
| |
Collapse
|
5
|
Saris J, Li Yim AYF, Bootsma S, Lenos KJ, Franco Fernandez R, Khan HN, Verhoeff J, Poel D, Mrzlikar NM, Xiong L, Schijven MP, van Grieken NCT, Kranenburg O, Wildenberg ME, Logiantara A, Jongerius C, Garcia Vallejo JJ, Gisbertz SS, Derks S, Tuynman JB, D'Haens GRAM, Vermeulen L, Grootjans J. Peritoneal resident macrophages constitute an immunosuppressive environment in peritoneal metastasized colorectal cancer. Nat Commun 2025; 16:3669. [PMID: 40246872 PMCID: PMC12006467 DOI: 10.1038/s41467-025-58999-6] [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: 04/22/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Patients with peritoneal metastasized colorectal cancer (PM-CRC) have a dismal prognosis. We hypothesized that an immunosuppressive environment in the peritoneal cavity underlies poor prognosis. We define the composition of the human peritoneal immune system (PerIS) using single-cell technologies in 18 patients with- and without PM-CRC, as well as in matched peritoneal metastases (n = 8). Here we show that the PerIS contains abundant immunosuppressive C1Q+VSIG4+ and SPP1+VSIG4+ peritoneal-resident macrophages (PRMs), as well as monocyte-like cavity macrophages (mono-CMs), which share features with tumor-associated macrophages, even in homeostasis. In PM-CRC, expression of immunosuppressive cytokines IL10 and VEGF increases, while simultaneously expression of antigen-presenting molecules decreases in PRMs. These intratumoral suppressive PRMs originate from the PerIS, and intraperitoneal depletion of PRMs in vivo using anti-CSF1R combined with anti-PD1 significantly reduces tumor burden and improves survival. Thus, PRMs define a metastatic site-specific immunosuppressive niche, and targeting PRMs is a promising treatment strategy for PM-CRC.
Collapse
Affiliation(s)
- J Saris
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - A Y F Li Yim
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands
| | - S Bootsma
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - K J Lenos
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - R Franco Fernandez
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - H N Khan
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - J Verhoeff
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands
- Molecular Cell Biology & Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - D Poel
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - N M Mrzlikar
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - L Xiong
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - M P Schijven
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Digital Health, Amsterdam, The Netherlands
| | - N C T van Grieken
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - O Kranenburg
- Laboratory Translational Oncology, Division of Imaging and Cancer, University Medical Center Utrecht, Utrecht, The Netherlands
- Utrecht Platform for Organoid Technology, Utrecht University, Utrecht, The Netherlands
| | - M E Wildenberg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - A Logiantara
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - C Jongerius
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - J J Garcia Vallejo
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands
- Molecular Cell Biology & Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - S S Gisbertz
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - S Derks
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J B Tuynman
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - G R A M D'Haens
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - L Vermeulen
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
- Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - J Grootjans
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Amsterdam, The Netherlands.
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Oncode Institute, Amsterdam, The Netherlands.
- Amsterdam Infection & Immunity Institute, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Ho QY, Hester J, Issa F. Regulatory cell therapy for kidney transplantation and autoimmune kidney diseases. Pediatr Nephrol 2025; 40:39-52. [PMID: 39278988 PMCID: PMC11584488 DOI: 10.1007/s00467-024-06514-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 09/18/2024]
Abstract
Regulatory cell therapies, including regulatory T cells and mesenchymal stromal cells, have shown promise in early clinical trials for reducing immunosuppression burden in transplantation. While regulatory cell therapies may also offer potential for treating autoimmune kidney diseases, data remains sparse, limited mainly to preclinical studies. This review synthesises current literature on the application of regulatory cell therapies in these fields, highlighting the safety and efficacy shown in existing clinical trials. We discuss the need for further clinical validation, optimisation of clinical and immune monitoring protocols, and the challenges of manufacturing and quality control under Good Manufacturing Practice conditions, particularly for investigator-led trials. Additionally, we explore the potential for expanding clinical indications and the unique challenges posed in paediatric applications. Future directions include scaling up production, refining protocols to ensure consistent quality across manufacturing sites, and extending applications to other immune-mediated diseases.
Collapse
Affiliation(s)
- Quan Yao Ho
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, UK
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, UK
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, UK.
| |
Collapse
|
7
|
Bellos E, Santillo D, Vantourout P, Jackson HR, Duret A, Hearn H, Seeleuthner Y, Talouarn E, Hodeib S, Patel H, Powell O, Yeoh S, Mustafa S, Habgood-Coote D, Nichols S, Estramiana Elorrieta L, D’Souza G, Wright VJ, Estrada-Rivadeneyra D, Tremoulet AH, Dummer KB, Netea SA, Condino-Neto A, Lau YL, Núñez Cuadros E, Toubiana J, Holanda Pena M, Rieux-Laucat F, Luyt CE, Haerynck F, Mège JL, Chakravorty S, Haddad E, Morin MP, Metin Akcan Ö, Keles S, Emiroglu M, Alkan G, Tüter Öz SK, Elmas Bozdemir S, Morelle G, Volokha A, Kendir-Demirkol Y, Sözeri B, Coskuner T, Yahsi A, Gulhan B, Kanik-Yuksek S, Bayhan GI, Ozkaya-Parlakay A, Yesilbas O, Hatipoglu N, Ozcelik T, Belot A, Chopin E, Barlogis V, Sevketoglu E, Menentoglu E, Gayretli Aydin ZG, Bloomfield M, AlKhater SA, Cyrus C, Stepanovskiy Y, Bondarenko A, Öz FN, Polat M, Fremuth J, Lebl J, Geraldo A, Jouanguy E, Carter MJ, Wellman P, Peters M, Pérez de Diego R, Edwards LA, Chiu C, Noursadeghi M, Bolze A, Shimizu C, Kaforou M, Hamilton MS, Herberg JA, Schmitt EG, Rodriguez-Palmero A, Pujol A, Kim J, Cobat A, Abel L, Zhang SY, Casanova JL, Kuijpers TW, Burns JC, Levin M, Hayday AC, Sancho-Shimizu V. Heterozygous BTNL8 variants in individuals with multisystem inflammatory syndrome in children (MIS-C). J Exp Med 2024; 221:e920240699. [PMID: 39576310 PMCID: PMC11586762 DOI: 10.1084/jem.20240699] [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: 04/22/2024] [Revised: 08/27/2024] [Accepted: 09/27/2024] [Indexed: 11/24/2024] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a rare condition following SARS-CoV-2 infection associated with intestinal manifestations. Genetic predisposition, including inborn errors of the OAS-RNAseL pathway, has been reported. We sequenced 154 MIS-C patients and utilized a novel statistical framework of gene burden analysis, "burdenMC," which identified an enrichment for rare predicted-deleterious variants in BTNL8 (OR = 4.2, 95% CI: 3.5-5.3, P < 10-6). BTNL8 encodes an intestinal epithelial regulator of Vγ4+γδ T cells implicated in regulating gut homeostasis. Enrichment was exclusive to MIS-C, being absent in patients with COVID-19 or bacterial disease. Using an available functional test for BTNL8, rare variants from a larger cohort of MIS-C patients (n = 835) were tested which identified eight variants in 18 patients (2.2%) with impaired engagement of Vγ4+γδ T cells. Most of these variants were in the B30.2 domain of BTNL8 implicated in sensing epithelial cell status. These findings were associated with altered intestinal permeability, suggesting a possible link between disrupted gut homeostasis and MIS-C-associated enteropathy triggered by SARS-CoV-2.
Collapse
Affiliation(s)
- Evangelos Bellos
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
| | - Dilys Santillo
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Section of Virology, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Pierre Vantourout
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King’s College London, London, UK
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Heather R. Jackson
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Amedine Duret
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Henry Hearn
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163 Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
| | - Estelle Talouarn
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163 Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
| | - Stephanie Hodeib
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Section of Virology, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Harsita Patel
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Oliver Powell
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Sophya Yeoh
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Sobia Mustafa
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Dominic Habgood-Coote
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Samuel Nichols
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Leire Estramiana Elorrieta
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Giselle D’Souza
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Victoria J. Wright
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Diego Estrada-Rivadeneyra
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Adriana H. Tremoulet
- Department of Pediatrics, Kawasaki Disease Research Center, University of California San Diego, La Jolla, CA, USA
- Rady Children’s Hospital-San Diego, San Diego, CA, USA
| | - Kirsten B. Dummer
- Department of Pediatrics, Kawasaki Disease Research Center, University of California San Diego, La Jolla, CA, USA
- Rady Children’s Hospital-San Diego, San Diego, CA, USA
| | - Stejara A. Netea
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Emma Children’s Hospital, Amsterdam University Medical Center (AmsterdamUMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Antonio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Yu Lung Lau
- Department of Paediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Esmeralda Núñez Cuadros
- Department of Pediatrics, Regional University Hospital of Málaga, IBIMA Research Institute, Málaga, Spain
| | - Julie Toubiana
- Department of General Pediatrics and Infectious Diseases, Necker-Enfants Malades University Hospital, AP-HP, Université Paris Cité, Paris, France
| | | | - Frédéric Rieux-Laucat
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163-Institut Imagine, Paris, France
- Imagine Institute, Paris Descartes-Sorbonne Université Paris Cité, Paris, France
| | - Charles-Edouard Luyt
- Intensive Care Unit, AP-HP Pitié-Salpêtrière Hospital, Paris University, Paris, France
| | | | | | - Samya Chakravorty
- Biocon Bristol Myers Squibb Research and Development Center, Syngene Intl. Ltd., Bengaluru, India
- Bristol Myers Squibb, Lawrenceville, NJ, USA
- Emory University Department of Pediatrics and Human Genetics, Atlanta GA, USA
| | - Elie Haddad
- CHU Sainte-Justine Azrieli Research Center, Montreal, Canada
- Department of Microbiology, Infectious Diseases and Immunology, University of Montreal, Montreal, Canada
- Department of Pediatrics, University of Montreal, Montreal, Canada
| | | | - Özge Metin Akcan
- Division of Pediatric Infectious Diseases, Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Sevgi Keles
- Division of Pediatric Allergy and Immunology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Melike Emiroglu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Gulsum Alkan
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Sadiye Kübra Tüter Öz
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Sefika Elmas Bozdemir
- Division of Pediatric Allergy and Immunology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Guillaume Morelle
- Department of General Paediatrics, Hôpital Bicêtre, AP-HP, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Alla Volokha
- Pediatric Infectious Disease and Pediatric Immunology Department, Shupyk National Healthcare University, Kyiv, Ukraine
| | - Yasemin Kendir-Demirkol
- Department of Pediatric Genetics, Umraniye Education and Research Hospital, Health Sciences University, İstanbul, Turkey
| | - Betul Sözeri
- Division of Pediatric Rheumatology, Umraniye Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Taner Coskuner
- Division of Pediatric Rheumatology, Umraniye Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Aysun Yahsi
- Department of Pediatric Infectious Diseases, Ankara City Hospital, Ankara, Turkey
| | - Belgin Gulhan
- Department of Pediatric Infectious Diseases, Ankara City Hospital, Ankara, Turkey
| | - Saliha Kanik-Yuksek
- Department of Pediatric Infectious Diseases, Ankara City Hospital, Ankara, Turkey
| | | | | | - Osman Yesilbas
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Nevin Hatipoglu
- Pediatric Infectious Diseases Unit, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | - Tayfun Ozcelik
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Alexandre Belot
- Service de Rhumatologie Pédiatrique, Hôpital Femme-Mère-Enfant, Groupement Hospitalier Est – Bâtiment “Pinel”, Bron, France
| | - Emilie Chopin
- CBC BIOTEC Biobank, GHE, Hospices Civils de Lyon, Lyon, France
| | - Vincent Barlogis
- La Timone Children Hospital, Aix-Marseille University, APHM, Marseille, France
| | - Esra Sevketoglu
- Univeristy of Health Sciences Turkiye Bakirkoy Dr. Sadi Konuk Research and Training Hospital Pediatirc Intensive Care Department, Istanbul, Türkiye
| | - Emin Menentoglu
- Univeristy of Health Sciences Turkiye Bakirkoy Dr. Sadi Konuk Research and Training Hospital Pediatirc Intensive Care Department, Istanbul, Türkiye
| | - Zeynep Gokce Gayretli Aydin
- Division of Pediatric Infectious Disease, Department of Pediatrics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Marketa Bloomfield
- Department of Immunology, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
- Department of Paediatrics, 1st Faculty of Medicine, Charles University in Prague and Thomayer University Hospital, Prague, Czech Republic
| | - Suzan A. AlKhater
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Department of Pediatrics, King Fahad Hospital of the University, Al-Khobar, Saudi Arabia
| | - Cyril Cyrus
- Department of Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Yuriy Stepanovskiy
- Department of Pediatrics, Immunology, Infectious, and Rare Diseases at the International European University, Kyiv, Ukraine
| | - Anastasiia Bondarenko
- Department of Pediatrics, Immunology, Infectious, and Rare Diseases at the International European University, Kyiv, Ukraine
| | - Fatma Nur Öz
- Department of Pediatric Infectious Disease, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Meltem Polat
- Department of Pediatric Infectious Diseases, Gazi University School of Medicine, Ankara, Turkey
| | - Jiří Fremuth
- Department of Pediatrics - PICU, Faculty of Medicine in Pilsen, Charles University in Prague, Prague, Czech Republic
| | - Jan Lebl
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czech Republic
| | - Amyrath Geraldo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163 Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
- Department of Pediatircs, Germans Trias i Pujol Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, Rockefeller University, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163 Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
- Department of Pediatircs, Germans Trias i Pujol Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, Rockefeller University, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Michael J. Carter
- Paediatric Intensive Care, Evelina London Children’s Hospital, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College London, St Thomas’ Hospital, London, UK
| | - Paul Wellman
- Paediatric Intensive Care, Evelina London Children’s Hospital, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Mark Peters
- Paediatric Intensive Care Unit, Great Ormond Street Hospital for Children NHS Foundation Trust and NIHR Biomedical Research Centre, London, UK
- University College London Great Ormond St Institute of Child Health, London, UK
| | - Rebeca Pérez de Diego
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ Institute for Health Research, University Hospital “La Paz”, Madrid, Spain
| | - Lindsey Ann Edwards
- Centre Host Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s Tower, Guy’s Hospital, London, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, UK
| | | | - Chisato Shimizu
- Department of Pediatrics, Kawasaki Disease Research Center, University of California San Diego, La Jolla, CA, USA
- Rady Children’s Hospital-San Diego, San Diego, CA, USA
| | - Myrsini Kaforou
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Melissa Shea Hamilton
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
| | - Jethro A. Herberg
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Erica G. Schmitt
- Division of Rheumatology and Immunology, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Agusti Rodriguez-Palmero
- Department of Pediatircs, Germans Trias i Pujol Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
- Catalan Institution of Research and Advanced Studies, Barcelona, Spain
| | - Jihoon Kim
- Department of Biomedical Informatics, University of California, San Diego, CA, USA
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT, USA
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163 Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163 Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163 Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163 Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Université Paris Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, Rockefeller University, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Taco W. Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Emma Children’s Hospital, Amsterdam University Medical Center (AmsterdamUMC), University of Amsterdam, Amsterdam, The Netherlands
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory at the AmsterdamUMC, Amsterdam Institute for Infection and Immunity, AmsterdamUMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jane C. Burns
- Department of Pediatrics, Kawasaki Disease Research Center, University of California San Diego, La Jolla, CA, USA
- Rady Children’s Hospital-San Diego, San Diego, CA, USA
| | - Michael Levin
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Adrian C. Hayday
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King’s College London, London, UK
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Vanessa Sancho-Shimizu
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, UK
- Section of Virology, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
8
|
Liu S, Zahorchak AF, Dobrowolski SF, Metes DM, Thomson AW, Abdelsamed HA. Epigenetic signature of human vitamin D3 and IL-10 conditioned regulatory DCs. Sci Rep 2024; 14:28748. [PMID: 39567586 PMCID: PMC11579388 DOI: 10.1038/s41598-024-79299-x] [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: 05/07/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024] Open
Abstract
During differentiation of precursor cells into their destination cell type, cell fate decisions are enforced by a broad array of epigenetic modifications, including DNA methylation, which is reflected by the transcriptome. Thus, regulatory dendritic cells (DCregs) acquire specific epigenetic programs and immunomodulatory functions during their differentiation from monocytes. To define the epigenetic signature of human DCregs generated in vitamin D3 (vitD3) and IL-10 compared to immune stimulatory DCs (sDCs), we measured levels of DNA methylation by whole genome bisulfite sequencing (WGBS). Distinct DNA methylation patterns were acquired by DCregs compared to sDCs. These patterns were located mainly in transcriptional regulatory regions. Associated genes were enriched in STAT3-signaling and valine catabolism in DCregs; conversely, pro-inflammatory pathways, e.g. pattern recognition receptor signaling, were enriched in sDCs. Further, DCreg differentially-methylated regions (DMRs) were enriched in binding motifs specific to the immunomodulatory transcription factor Krueppel-like factor 11 (KLF11), while activator protein-1 (AP-1) (Fos:Jun) transcription factor-binding motifs were enriched in sDC DMRs. Using publicly-available data-sets, we defined a common epigenetic signature shared between DCregs generated in vitD3 and IL-10, or dexamethasone or vitD3 alone. These insights may help pave the way for design of epigenetic-based approaches to enhance the production of DCregs as effective therapeutic agents.
Collapse
Affiliation(s)
- Silvia Liu
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Alan F Zahorchak
- Department of Surgery, University of Pittsburgh, Pittsburgh, USA
- Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, USA
| | | | - Diana M Metes
- Department of Surgery, University of Pittsburgh, Pittsburgh, USA
- Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, USA
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Angus W Thomson
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, USA.
- Department of Surgery, University of Pittsburgh, Pittsburgh, USA.
- Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, USA.
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, USA.
| | - Hossam A Abdelsamed
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, USA.
- Department of Surgery, University of Pittsburgh, Pittsburgh, USA.
- Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, USA.
- Immunology Center of Georgia, Augusta University, Augusta, USA.
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, USA.
| |
Collapse
|
9
|
Migliorini A, Ge S, Atkins MH, Oakie A, Sambathkumar R, Kent G, Huang H, Sing A, Chua C, Gehring AJ, Keller GM, Notta F, Nostro MC. Embryonic macrophages support endocrine commitment during human pancreatic differentiation. Cell Stem Cell 2024; 31:1591-1611.e8. [PMID: 39406230 DOI: 10.1016/j.stem.2024.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/02/2024] [Accepted: 09/12/2024] [Indexed: 11/10/2024]
Abstract
Organogenesis is a complex process that relies on a dynamic interplay between extrinsic factors originating from the microenvironment and tissue-specific intrinsic factors. For pancreatic endocrine cells, the local niche consists of acinar and ductal cells as well as neuronal, immune, endothelial, and stromal cells. Hematopoietic cells have been detected in human pancreas as early as 6 post-conception weeks, but whether they play a role during human endocrinogenesis remains unknown. To investigate this, we performed single-nucleus RNA sequencing (snRNA-seq) of the second-trimester human pancreas and identified a wide range of hematopoietic cells, including two distinct subsets of tissue-resident macrophages. Leveraging this discovery, we developed a co-culture system of human embryonic stem cell-derived endocrine-macrophage organoids to model their interaction in vitro. Here, we show that macrophages support the differentiation and viability of endocrine cells in vitro and enhance tissue engraftment, highlighting their potential role in tissue engineering strategies for diabetes.
Collapse
Affiliation(s)
- Adriana Migliorini
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada.
| | - Sabrina Ge
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Michael H Atkins
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | | | - Gregory Kent
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Haiyang Huang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Angel Sing
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Conan Chua
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Adam J Gehring
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gordon M Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Faiyaz Notta
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada.
| |
Collapse
|
10
|
Liu L, Yao W, Wang M, Wang B, Kong F, Fan Z, Fan G. A systematic review of cardiovascular toxicities induced by cancer immune therapies: Underlying mechanisms, clinical manifestations and therapeutic approaches. Semin Cancer Biol 2024; 106-107:179-191. [PMID: 39442678 DOI: 10.1016/j.semcancer.2024.10.004] [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: 04/29/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Immunotherapy has revolutionized the management of various types of cancers, even those previously deemed untreatable. Nonetheless, these medications have been associated with inflammation and damage across various organs. These challenges are exemplified by the adverse cardiovascular impacts of cancer immunotherapy, which need comprehensive understanding, clarification, and management integrated into the overall care of cancer patients. Numerous anticancer immunotherapies have been linked to the prevalence and severity of cardiovascular toxicity. These challenges emphasize the importance of conducting fundamental and applied research to elucidate disease causes, discover prognostic indicators, enhance diagnostic methods, and create successful therapies. Despite the acknowledged importance of T cells, there remains a knowledge gap regarding the inciting antigens, the reasons for their recognition, and the mechanisms of how they contribute to cardiac cell injury. In this review, we summarize the molecular mechanism, epidemiology, diagnosis, pathophysiology and corresponding treatment of cardiovascular toxicity induced by immunotherapy, including immune checkpoint inhibitors (ICIs), adoptive cell therapies (ACT), and bi-specific T-cell engagers (BiTEs) among others. By elucidating these aspects, we aim to provide a better understanding of immunotherapies in cancer treatment and offer guidance for their clinical application.
Collapse
Affiliation(s)
- Li Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Wentao Yao
- Department of Urology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Mi Wang
- Department of Cardiology Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Baohui Wang
- Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fanming Kong
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Zhongguo Fan
- Department of Cardiology Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 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
|
Huang CH, Chen WY, Chen RF, Ramachandran S, Liu KF, Kuo YR. Cell therapies and its derivatives as immunomodulators in vascularized composite allotransplantation. Asian J Surg 2024; 47:4251-4259. [PMID: 38704267 DOI: 10.1016/j.asjsur.2024.04.094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024] Open
Abstract
The adverse effects of traditional pharmaceutical immunosuppressive regimens have been a major obstacle to successful allograft survival in vascularized composite tissue allotransplantation (VCA) cases. Consequently, there is a pressing need to explore alternative approaches to reduce reliance on conventional immunotherapy. Cell therapy, encompassing immune-cell-based and stem-cell-based regimens, has emerged as a promising avenue of research. Immune cells can be categorized into two main systems: innate immunity and adaptive immunity. Innate immunity comprises tolerogenic dendritic cells, regulatory macrophages, and invariant natural killer T cells, while adaptive immunity includes T regulatory cells and B regulatory cells. Investigations are currently underway to assess the potential of these immune cell populations in inducing immune tolerance. Furthermore, mixed chimerism therapy, involving the transplantation of hematopoietic stem and progenitor cells and mesenchymal stem cells (MSC), shows promise in promoting allograft tolerance. Additionally, extracellular vesicles (EVs) derived from MSCs offer a novel avenue for extending allograft survival. This review provides a comprehensive summary of cutting-edge research on immune cell therapies, mixed chimerism therapies, and MSCs-derived EVs in the context of VCAs. Findings from preclinical and clinical studies demonstrate the tremendous potential of these alternative therapies in optimizing allograft survival in VCAs.
Collapse
Affiliation(s)
- Chao-Hsin Huang
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| | - Wei Yu Chen
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| | - Rong-Fu Chen
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| | - Savitha Ramachandran
- Department of Plastic and Reconstructive Surgery, Singapore General Hospital, Singapore.
| | - Keng-Fan Liu
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| | - Yur-Ren Kuo
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Faculty of Medicine, College of Medicine, Orthopaedic Research Center, Regenerative Medicine, Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Academic Clinical Programme for Musculoskeletal Sciences, Duke-NUS Graduate Medical School, Singapore; Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan.
| |
Collapse
|
13
|
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
|
14
|
Zitta K, Hummitzsch L, Lichte F, Fändrich F, Steinfath M, Eimer C, Kapahnke S, Buerger M, Hess K, Rusch M, Rusch R, Berndt R, Albrecht M. Effects of temporal IFNγ exposure on macrophage phenotype and secretory profile: exploring GMP-Compliant production of a novel subtype of regulatory macrophages (Mreg IFNγ0) for potential cell therapeutic applications. J Transl Med 2024; 22:534. [PMID: 38835045 PMCID: PMC11151567 DOI: 10.1186/s12967-024-05336-y] [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: 04/17/2024] [Accepted: 05/18/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Macrophages are involved in tissue homeostasis, angiogenesis and immunomodulation. Proangiogenic and anti-inflammatory macrophages (regulatory macrophages, Mreg) can be differentiated in-vitro from CD14+ monocytes by using a defined cell culture medium and a stimulus of IFNγ. AIM OF THE STUDY To scrutinize the potential impact of temporal IFNγ exposure on macrophage differentiation as such exposure may lead to the emergence of a distinct and novel macrophage subtype. METHODS Differentiation of human CD14+ monocytes to Mreg was performed using a GMP compliant protocol and administration of IFNγ on day 6. Monocytes from the same donor were in parallel differentiated to MregIFNγ0 using the identical protocol but with administration of IFNγ on day 0. Cell characterization was performed using brightfield microscopy, automated and metabolic cell analysis, transmission electron microscopy, flow cytometry, qPCR and secretome profiling. RESULTS Mreg and MregIFNγ0 showed no differences in cell size and volume. However, phenotypically MregIFNγ0 exhibited fewer intracellular vesicles/vacuoles but larger pseudopodia-like extensions. MregIFNγ0 revealed reduced expression of IDO and PD-L1 (P < 0.01 for both). They were positive for CD80, CD14, CD16 and CD38 (P < 0.0001vs. Mreg for all), while the majority of MregIFNγ0 did not express CD206, CD56, and CD103 on their cell surface (P < 0.01 vs. Mreg for all). In terms of their secretomes, MregIFNγ0 differed significantly from Mreg. MregIFNγ0 media exhibited reduced levels of ENA-78, Osteopontin and Serpin E1, while the amounts of MIG (CXCL9) and IP10 were increased. CONCLUSION Exposing CD14+ monocytes to an alternatively timed IFNγ stimulation results in a novel macrophage subtype which possess additional M1-like features (MregIFNγ0). MregIFNγ0 may therefore have the potential to serve as cellular therapeutics for clinical applications beyond those covered by M2-like Mreg, including immunomodulation and tumor treatment.
Collapse
Affiliation(s)
- Karina Zitta
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein (UKSH), Kiel, Germany.
| | - Lars Hummitzsch
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Frank Lichte
- Department of Anatomy, University of Kiel, Kiel, Germany
| | - Fred Fändrich
- Clinic for Applied Cell Therapy, UKSH, Kiel, Germany
| | - Markus Steinfath
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Christine Eimer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | | | - Matthias Buerger
- Clinic for Vascular and Endovascular Surgery, UKSH, Kiel, Germany
| | | | - Melanie Rusch
- Clinic for Vascular and Endovascular Surgery, UKSH, Kiel, Germany
| | - Rene Rusch
- Clinic for Vascular and Endovascular Surgery, UKSH, Kiel, Germany
| | - Rouven Berndt
- Clinic for Vascular and Endovascular Surgery, UKSH, Kiel, Germany
| | - Martin Albrecht
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| |
Collapse
|
15
|
Yin N, Li X, Zhang X, Xue S, Cao Y, Niedermann G, Lu Y, Xue J. Development of pharmacological immunoregulatory anti-cancer therapeutics: current mechanistic studies and clinical opportunities. Signal Transduct Target Ther 2024; 9:126. [PMID: 38773064 PMCID: PMC11109181 DOI: 10.1038/s41392-024-01826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 05/23/2024] Open
Abstract
Immunotherapy represented by anti-PD-(L)1 and anti-CTLA-4 inhibitors has revolutionized cancer treatment, but challenges related to resistance and toxicity still remain. Due to the advancement of immuno-oncology, an increasing number of novel immunoregulatory targets and mechanisms are being revealed, with relevant therapies promising to improve clinical immunotherapy in the foreseeable future. Therefore, comprehending the larger picture is important. In this review, we analyze and summarize the current landscape of preclinical and translational mechanistic research, drug development, and clinical trials that brought about next-generation pharmacological immunoregulatory anti-cancer agents and drug candidates beyond classical immune checkpoint inhibitors. Along with further clarification of cancer immunobiology and advances in antibody engineering, agents targeting additional inhibitory immune checkpoints, including LAG-3, TIM-3, TIGIT, CD47, and B7 family members are becoming an important part of cancer immunotherapy research and discovery, as are structurally and functionally optimized novel anti-PD-(L)1 and anti-CTLA-4 agents and agonists of co-stimulatory molecules of T cells. Exemplified by bispecific T cell engagers, newly emerging bi-specific and multi-specific antibodies targeting immunoregulatory molecules can provide considerable clinical benefits. Next-generation agents also include immune epigenetic drugs and cytokine-based therapeutics. Cell therapies, cancer vaccines, and oncolytic viruses are not covered in this review. This comprehensive review might aid in further development and the fastest possible clinical adoption of effective immuno-oncology modalities for the benefit of patients.
Collapse
Affiliation(s)
- Nanhao Yin
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xintong Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xuanwei Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Shaolong Xue
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, No. 20, Section 3, South Renmin Road, Chengdu, 610041, Sichuan, PR China
| | - Yu Cao
- Department of Emergency Medicine, Laboratory of Emergency Medicine, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
- Institute of Disaster Medicine & Institute of Emergency Medicine, Sichuan University, No. 17, Gaopeng Avenue, Chengdu, 610041, Sichuan, PR China
| | - Gabriele Niedermann
- Department of Radiation Oncology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site DKTK-Freiburg, Robert-Koch-Strasse 3, 79106, Freiburg, Germany.
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| |
Collapse
|
16
|
Pei J, Zhang J, Yu C, Luo J, Wen S, Hua Y, Wei G. Transcriptomics-based identification of TYROBP and TLR8 as novel macrophage-related biomarkers for the diagnosis of acute rejection after kidney transplantation. Biochem Biophys Res Commun 2024; 709:149790. [PMID: 38564938 DOI: 10.1016/j.bbrc.2024.149790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024]
Abstract
Macrophages play an important role in the development and progression of acute rejection after kidney transplantation. The study aims to investigate the biological role and significance of macrophage-associated genes (MAG) in acute rejection after kidney transplantation. We utilized transcriptome sequencing results from public databases related to acute rejection of kidney transplantation for comprehensive analysis and validation in animal experiments. We found that a large number of immune-related signaling pathways are activated in acute rejection. PPI protein interaction networks and machine learning were used to establish a Hub gene consisting of TYROBP and TLR8 for the diagnosis of acute rejection. The single-gene GSEA enrichment analysis and immune cell correlation analysis revealed a close correlation between the expression of Hub genes and immune-related biological pathways as well as the expression of multiple immune cells. In addition, the study of TF, miRNAs, and drugs provided a theoretical basis for regulating and treating the Hub genes in acute rejection. Finally, the animal experiments demonstrated once again that acute rejection can aggravate kidney tissue damage, apoptosis level, and increase the release of inflammatory factors. We established and validated a macrophage-associated diagnostic model for acute rejection after kidney transplantation, which can accurately diagnose the biological alterations in acute rejection after kidney transplantation.
Collapse
Affiliation(s)
- Jun Pei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jie Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Chengjun Yu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jin Luo
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Sheng Wen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Yi Hua
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China.
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China.
| |
Collapse
|
17
|
Mani S, Garifallou J, Kim SJ, Simoni MK, Huh DD, Gordon SM, Mainigi M. Uterine macrophages and NK cells exhibit population and gene-level changes after implantation but maintain pro-invasive properties. Front Immunol 2024; 15:1364036. [PMID: 38566989 PMCID: PMC10985329 DOI: 10.3389/fimmu.2024.1364036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Prior to pregnancy, hormonal changes lead to cellular adaptations in the endometrium allowing for embryo implantation. Critical for successful pregnancy establishment, innate immune cells constitute a significant proportion of uterine cells prior to arrival of the embryo and throughout the first trimester in humans and animal models. Abnormal uterine immune cell function during implantation is believed to play a role in multiple adverse pregnancy outcomes. Current work in humans has focused on uterine immune cells present after pregnancy establishment, and limited in vitro models exist to explore unique functions of these cells. Methods With single-cell RNA-sequencing (scRNAseq), we comprehensively compared the human uterine immune landscape of the endometrium during the window of implantation and the decidua during the first trimester of pregnancy. Results We uncovered global and cell-type-specific gene signatures for each timepoint. Immune cells in the endometrium prior to implantation expressed genes associated with immune metabolism, division, and activation. In contrast, we observed widespread interferon signaling during the first trimester of pregnancy. We also provide evidence of specific inflammatory pathways enriched in pre- and post-implantation macrophages and natural killer (NK) cells in the uterine lining. Using our novel implantation-on-a-chip (IOC) to model human implantation ex vivo, we demonstrate for the first time that uterine macrophages strongly promote invasion of extravillous trophoblasts (EVTs), a process essential for pregnancy establishment. Pre- and post-implantation uterine macrophages promoted EVT invasion to a similar degree as pre- and post-implantation NK cells on the IOC. Conclusions This work provides a foundation for further investigation of the individual roles of uterine immune cell subtypes present prior to embryo implantation and during early pregnancy, which will be critical for our understanding of pregnancy complications associated with abnormal trophoblast invasion and placentation.
Collapse
Affiliation(s)
- Sneha Mani
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | - James Garifallou
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Se-jeong Kim
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael K. Simoni
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | - Dan Dongeun Huh
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
- National Science Foundation (NSF) Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Scott M. Gordon
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | - Monica Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
18
|
Mao K, Wang J, Xie Q, Yang YG, Shen S, Sun T, Wang J. Cationic nanoparticles-based approaches for immune tolerance induction in vivo. J Control Release 2024; 366:425-447. [PMID: 38154540 DOI: 10.1016/j.jconrel.2023.12.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/04/2023] [Accepted: 12/25/2023] [Indexed: 12/30/2023]
Abstract
The development of autoimmune diseases and the rejection of transplanted organs are primarily caused by an exaggerated immune response to autoantigens or graft antigens. Achieving immune tolerance is crucial for the effective treatment of these conditions. However, traditional therapies often have limited therapeutic efficacy and can result in systemic toxic effects. The emergence of nanomedicine offers a promising avenue for addressing immune-related diseases. Among the various nanoparticle formulations, cationic nanoparticles have demonstrated significant potential in inducing immune tolerance. In this review, we provide an overview of the underlying mechanism of autoimmune disease and organ transplantation rejection. We then highlight the recent advancements and advantages of utilizing cationic nanoparticles for inducing immune tolerance in the treatment of autoimmune diseases and the prevention of transplant rejection.
Collapse
Affiliation(s)
- Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Qianyue Xie
- Huafu International Department, Affiliated High School of South China Normal University, Guangzhou, Guangdong, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China; Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovatiion Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
19
|
Hoang TX, Kim JY. Regulatory macrophages in solid organ xenotransplantation. KOREAN JOURNAL OF TRANSPLANTATION 2023; 37:229-240. [PMID: 38115165 PMCID: PMC10772277 DOI: 10.4285/kjt.23.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/10/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023] Open
Abstract
Due to a critical organ shortage, pig organs are being explored for use in transplantation. Differences between species, particularly in cell surface glycans, can trigger elevated immune responses in xenotransplantation. To mitigate the risk of hyperacute rejection, genetically modified pigs have been developed that lack certain glycans and express human complement inhibitors. Nevertheless, organs from these pigs may still provoke stronger inflammatory and innate immune reactions than allotransplants. Dysregulation of coagulation and persistent inflammation remain obstacles in the transplantation of pig organs into primates. Regulatory macrophages (Mregs), known for their anti-inflammatory properties, could offer a potential solution. Mregs secrete interleukin 10 and transforming growth factor beta, thereby suppressing immune responses and promoting the development of regulatory T cells. These Mregs are typically induced via the stimulation of monocytes or macrophages with macrophage colony-stimulating factor and interferon gamma, and they conspicuously express the stable marker dehydrogenase/reductase 9. Consequently, understanding the precise mechanisms governing Mreg generation, stability, and immunomodulation could pave the way for the therapeutic use of Mregs generated in vitro. This approach has the potential to reduce the required dosages and durations of anti-inflammatory and immunosuppressive medications in preclinical and clinical settings.
Collapse
Affiliation(s)
- Thi Xoan Hoang
- Department of Life Science, Gachon University, Seongnam, Korea
| | - Jae Young Kim
- Department of Life Science, Gachon University, Seongnam, Korea
| |
Collapse
|
20
|
Kleber J, Yang Zhou J, Weber F, Bitterer F, Hauer P, Kupke P, Kronenberg K, Geissler EK, Schlitt HJ, Hornung M, Hutchinson JA, Werner JM. Immune profile of patients with peritoneal carcinomatosis selected for CRS-HIPEC therapy. Cancer Immunol Immunother 2023; 72:3867-3873. [PMID: 37580610 PMCID: PMC10576707 DOI: 10.1007/s00262-023-03515-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023]
Abstract
Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is a treatment option for peritoneal carcinomatosis (PC) from colorectal cancer (CRC), which is otherwise a terminal stage of disease. Nevertheless, survival outcomes are only marginally superior to other treatments. This fact highlights the need for better strategies to control intra-abdominal disease recurrence after CRS-HIPEC, including the complementary use of immunotherapies. The aim of this study was therefore to investigate the immune phenotype of T cells in patients with PC. Fifty three patients with CRC (34 patients with PC and 19 patients without PC) were enrolled in a prospective study (clinicaltrials.gov: NCT04108936). Peripheral blood and omental fat were collected to isolate peripheral blood mononuclear cells (PBMCs) and adipose tissue mononuclear cells (ATMCs). These cells were analysed by flow cytometry using a panel focused upon T cell memory differentiation and exhaustion markers. We found a more naïve profile for CD8+ T cells in peripheral blood and intra-abdominal fat of PC patients compared to comparator group (CG) patients. Furthermore, there was an over-representation of CD4+ T cells expressing inhibitory receptors in adipose tissue of PC patients, but not in blood. Our description of intraperitoneal T cell subsets gives us a better understanding of how peritoneal carcinomatosis shapes local immune responses.
Collapse
Affiliation(s)
- Julia Kleber
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Jordi Yang Zhou
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Florian Weber
- Institute for Pathology, University of Regensburg, Regensburg, Germany
| | - Florian Bitterer
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Patricia Hauer
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Paul Kupke
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Katharina Kronenberg
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Hans J Schlitt
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Matthias Hornung
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - James A Hutchinson
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Jens M Werner
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
21
|
Unar A, Bertolino L, Patauner F, Gallo R, Durante-Mangoni E. Decoding Sepsis-Induced Disseminated Intravascular Coagulation: A Comprehensive Review of Existing and Emerging Therapies. J Clin Med 2023; 12:6128. [PMID: 37834771 PMCID: PMC10573475 DOI: 10.3390/jcm12196128] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/09/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
Disseminated intravascular coagulation (DIC) is a recurrent complication of sepsis. Since DIC not only promotes organ dysfunction but also represents a strong prognostic factor, it is important to diagnose DIC as early as possible. When coagulation is activated, fibrinolysis is inhibited, blood thinners are consumed, and a condition is created that promotes blood clotting, making it more difficult for the body to remove fibrin or prevent it from being deposited in the blood vessels. This leads to microvascular thrombosis, which plays a role in organ dysfunction. Despite efforts to understand the underlying mechanisms of sepsis-induced DIC, healthcare providers worldwide still face challenges in effectively treating this condition. In this review, we provide an in-depth analysis of the available strategies for sepsis-induced DIC, considering their effectiveness, limitations, and potential for future advances. Corticosteroids (CS), recombinant thrombomodulin (rTM), vitamin C, fibrinolytic therapy, and platelet transfusion are among the treatments discussed in the review. In addition, we are specifically addressing immunomodulatory therapy (IMT) by investigating treatments such as granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), interferon gamma (IFN-γ), and mesenchymal stem cell therapy (MSC). Finally, we also examined how these therapies might affect COVID-19 cases, which often present with sepsis-induced DIC. The review suggests that targeted experiments with randomization are needed to verify the effectiveness of these treatments and to discover novel approaches to treat sepsis-induced DIC. By increasing our knowledge of sepsis-induced DIC, we can develop targeted treatments that have the potential to save lives and improve outcomes.
Collapse
Affiliation(s)
- Ahsanullah Unar
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, 80138 Naples, Italy; (A.U.); (L.B.); (F.P.); (R.G.)
| | - Lorenzo Bertolino
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, 80138 Naples, Italy; (A.U.); (L.B.); (F.P.); (R.G.)
| | - Fabian Patauner
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, 80138 Naples, Italy; (A.U.); (L.B.); (F.P.); (R.G.)
| | - Raffaella Gallo
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, 80138 Naples, Italy; (A.U.); (L.B.); (F.P.); (R.G.)
| | - Emanuele Durante-Mangoni
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, 80138 Naples, Italy; (A.U.); (L.B.); (F.P.); (R.G.)
- Unit of Infectious and Transplant Medicine, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy
| |
Collapse
|
22
|
Aghbash PS, Rasizadeh R, Arefi V, Nahand JS, Baghi HB. Immune-checkpoint expression in antigen-presenting cells (APCs) of cytomegaloviruses infection after transplantation: as a diagnostic biomarker. Arch Microbiol 2023; 205:280. [PMID: 37430000 DOI: 10.1007/s00203-023-03623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/12/2023]
Abstract
Cytomegalovirus (CMV), a member of the Herpesviridae family, mostly causes only slight feverish symptoms or can be asymptomatic in immunocompetent individuals. However, it is known to be particularly a significant cause of morbidity in immunocompromised patients, including transplant recipients, whose immune system has been weakened due to the consumption of immunosuppressor drugs. Therefore, the diagnosis of CMV infection after transplantation is crucial. New diagnostic methods for the quick detection of CMV have been developed as a result of understanding the clinical importance of invasive CMV. Antigen-presenting cells (APCs) and T cells are important components of the immune system and it may be possible to diagnose viral infections using immunological markers, such as lymphocytosis, cytotoxic T lymphocytes (CTL), and serum cytokine levels. Moreover, PD-1, CTLA 4, and TIGIT, which are expressed on certain T cells and antigen-presenting cells, are over-expressed during the infection. The assessment of CMV infection based on T cell and APC activity, and the expression of immunological checkpoints, can be helpful for the diagnosis of transplant patients at risk for CMV infection. In this review, we will investigate how immune checkpoints affect immune cells and how they impair organ transplantation after CMV infection.
Collapse
Affiliation(s)
- Parisa Shiri Aghbash
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reyhaneh Rasizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Arefi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran.
| |
Collapse
|
23
|
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
|
24
|
Malard F, Holler E, Sandmaier BM, Huang H, Mohty M. Acute graft-versus-host disease. Nat Rev Dis Primers 2023; 9:27. [PMID: 37291149 DOI: 10.1038/s41572-023-00438-1] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 06/10/2023]
Abstract
Acute graft-versus-host disease (GVHD) is a common immune complication that can occur after allogeneic haematopoietic cell transplantation (alloHCT). Acute GVHD is a major health problem in these patients, and is associated with high morbidity and mortality. Acute GVHD is caused by the recognition and the destruction of the recipient tissues and organs by the donor immune effector cells. This condition usually occurs within the first 3 months after alloHCT, but later onset is possible. Targeted organs include the skin, the lower and upper gastrointestinal tract and the liver. Diagnosis is mainly based on clinical examination, and complementary examinations are performed to exclude differential diagnoses. Preventive treatment for acute GVHD is administered to all patients who receive alloHCT, although it is not always effective. Steroids are used for first-line treatment, and the Janus kinase 2 (JAK2) inhibitor ruxolitinib is second-line treatment. No validated treatments are available for acute GVHD that is refractory to steroids and ruxolitinib, and therefore it remains an unmet medical need.
Collapse
Affiliation(s)
- Florent Malard
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France.
| | - Ernst Holler
- University Hospital of Regensburg, Department of Internal Medicine 3, Regensburg, Germany
| | - Brenda M Sandmaier
- Fred Hutchinson Cancer Center, Translational Science and Therapeutics Division, Seattle, WA, USA
- University of Washington School of Medicine, Division of Medical Oncology, Seattle, WA, USA
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Province, Hangzhou, China
- Engineering Laboratory for Stem Cell and Immunity Therapy, Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, China
| | - Mohamad Mohty
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France.
| |
Collapse
|
25
|
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
|
26
|
Zhang D, Liu Y, Ma J, Xu Z, Duan C, Wang Y, Li X, Han J, Zhuang R. Competitive binding of CD226/TIGIT with PVR regulates macrophage polarization and is involved in vascularized skin graft rejection. Am J Transplant 2023:S1600-6135(23)00404-5. [PMID: 37054890 DOI: 10.1016/j.ajt.2023.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/15/2023]
Abstract
End-stage organ failure often requires solid organ transplantation. Nevertheless, transplant rejection remains an unresolved issue. The induction of donor-specific tolerance is the ultimate goal in transplantation research. Here, an allograft vascularized skin rejection model using BALB/c-C57/BL6 mice was established to evaluate the regulation of the poliovirus receptor signaling pathway via CD226 knockout (KO) or TIGIT-Fc recombinant protein treatment. In the TIGIT-Fc-treated and CD226KO groups, graft survival time was significantly prolonged, with a Treg cell proportion increase and M2-type macrophage polarization. Donor-reactive recipient T cells became hyporesponsive while responding normally after a third-party antigen challenge. In both groups, serum IL-1β, IL-6, IL-12p70, IL-17A, TNF-α, IFN-γ, and monocyte chemoattractant protein-1 levels decreased, and the IL-10 level increased. In vitro, M2 markers, such as Arg1 and IL-10, were markedly increased by TIGIT-Fc, whereas iNOS, IL-1β, IL-6, IL-12p70, TNF-α, and IFN-γ levels decreased. CD226-Fc had the opposite effect. TIGIT suppressed Th1 and Th17 differentiation by inhibiting macrophage SHP-1 phosphorylation and enhanced ERK1/2-MSK1 phosphorylation and nuclear translocation of CREB. In conclusion, CD226 and TIGIT competitively bind to PVR with activating and inhibitory functions, respectively. Mechanistically, TIGIT promotes IL-10 transcription from macrophages by activating the ERK1/2-MSK1-CREB pathway and enhancing M2-type polarization. CD226/TIGIT-PVR are crucial regulatory molecules of allograft rejection.
Collapse
Affiliation(s)
- Dongliang Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China; Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yitian Liu
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jingchang Ma
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhigang Xu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chujun Duan
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yuling Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xuemei Li
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Juntao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
27
|
Zahorchak AF, DeRiggi ML, Muzzio JL, Sutherland V, Humar A, Lakkis FG, Hsu YMS, Thomson AW. Manufacturing and validation of Good Manufacturing Practice-compliant regulatory dendritic cells for infusion into organ transplant recipients. Cytotherapy 2023; 25:432-441. [PMID: 36639251 DOI: 10.1016/j.jcyt.2022.11.005] [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: 07/27/2022] [Revised: 10/28/2022] [Accepted: 11/18/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND AIMS Regulatory (or "tolerogenic") dendritic cells (DCregs) are a highly promising, innovative cell therapy for the induction or restoration of antigen-specific tolerance in immune-mediated inflammatory disorders. These conditions include organ allograft rejection, graft-versus-host disease following bone marrow transplantation and various autoimmune disorders. DCregs generated for adoptive transfer have potential to reduce patients' dependence on non-specific immunosuppressive drugs that can induce serious side effects and enhance the risk of infection and certain types of cancer. Here, our aim was to provide a detailed account of our experience manufacturing and validating comparatively large numbers of Good Manufacturing Practice-grade DCregs for systemic (intravenous) infusion into 28 organ (liver) transplant recipients and to discuss factors that influence the satisfaction of release criteria and attainment of target cell numbers. RESULTS DCregs were generated in granulocyte-macrophage colony stimulating factor and interleukin (IL)-4 from elutriated monocyte fractions isolated from non-mobilized leukapheresis products of consenting healthy adult prospective liver transplant donors. Vitamin D3 was added on day 0 and 4 and IL-10 on day 4 during the 7-day culture period. Release and post-release criteria included cell viability, purity, phenotype, sterility and functional assessment. The overall conversion rate of monocytes to DCregs was 28 ± 8.2%, with 94 ± 5.1% product viability. The mean cell surface T-cell co-inhibitory to co-stimulatory molecule (programmed death ligand-1:CD86) mean fluorescence intensity ratio was 3.9 ± 2.2, and the mean ratio of anti-inflammatory:pro-inflammatory cytokine product (IL-10:IL-12p70) secreted upon CD40 ligation was 60 ± 63 (median = 40). The mean total number of DCregs generated from a single leukapheresis product (n = 25 donors) and from two leukapheresis products (n = 3 donors) was 489 ± 223 × 106 (n = 28). The mean total number of DCregs infused was 5.9 ± 2.8 × 106 per kg body weight. DCreg numbers within a target cell range of 2.5-10 × 106/kg were achieved for 25 of 27 (92.6%) of products generated. CONCLUSIONS High-purity DCregs meeting a range of quality criteria were readily generated from circulating blood monocytes under Good Manufacturing Practice conditions to meet target cell numbers for infusion into prospective organ transplant recipients.
Collapse
Affiliation(s)
- Alan F Zahorchak
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Misty L DeRiggi
- Immunologic Monitoring & Cellular Products Laboratory, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jennifer L Muzzio
- Immunologic Monitoring & Cellular Products Laboratory, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Veronica Sutherland
- Immunologic Monitoring & Cellular Products Laboratory, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Abhinav Humar
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Fadi G Lakkis
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yen-Michael S Hsu
- Immunologic Monitoring & Cellular Products Laboratory, University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania, USA; Department of Medicine, Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | - Angus W Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
The Potential Clinical Application of Induced Tolerogenic Macrophages. Transplantation 2023; 107:23-24. [PMID: 35876367 DOI: 10.1097/tp.0000000000004246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
31
|
Sheikholeslami A, Fazaeli H, Kalhor N, Khoshandam M, Eshagh Hoseini SJ, Sheykhhasan M. Use of Mesenchymal Stem Cells in Crohn's Disease and Perianal Fistulas: A Narrative Review. Curr Stem Cell Res Ther 2023; 18:76-92. [PMID: 34530720 DOI: 10.2174/1574888x16666210916145717] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/08/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Crohn's Disease (CD), which usually leads to anal fistulas among patients, is the most important inflammatory bowel disease that causes morbidity in many people around the world. This review article proposes using MSCs as a hopeful therapeutic strategy for CD and anal fistula treatment in both preclinical and clinical conditions. Finally, darvadstrocel, a cell-based medication to treat complex anal fistulas in adults, as the only European Medicines Agency (EMA)-approved product for the treatment of anal fistulas in CD is addressed. Although several common therapies, such as surgery and anti-tumor necrosis factor-alpha (TNF-α) drugs as well as a combination of these methods is used to improve this disease, however, due to the low effectiveness of these treatments, the use of new strategies with higher efficiency is still recommended. Cell therapy is among the new emerging therapeutic strategies that have attracted great attention from clinicians due to its unique capabilities. One of the most widely used cell sources administrated in cell therapy is mesenchymal stem cell (MSC). This review article will discuss preclinical and clinical studies about MSCs as a potent and promising therapeutic option in the treatment of CD and anal fistula.
Collapse
Affiliation(s)
- Azar Sheikholeslami
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
| | - Hoda Fazaeli
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom,Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
| | - Mohadeseh Khoshandam
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
| | | | - Mohsen Sheykhhasan
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research (ACECR), Qom Branch, Qom, Iran
- Department of Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
32
|
Yang Zhou J. Innate immunity and early liver inflammation. Front Immunol 2023; 14:1175147. [PMID: 37205101 PMCID: PMC10187146 DOI: 10.3389/fimmu.2023.1175147] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/30/2023] [Indexed: 05/21/2023] Open
Abstract
The innate system constitutes a first-line defence mechanism against pathogens. 80% of the blood supply entering the human liver arrives from the splanchnic circulation through the portal vein, so it is constantly exposed to immunologically active substances and pathogens from the gastrointestinal tract. Rapid neutralization of pathogens and toxins is an essential function of the liver, but so too is avoidance of harmful and unnecessary immune reactions. This delicate balance of reactivity and tolerance is orchestrated by a diverse repertoire of hepatic immune cells. In particular, the human liver is enriched in many innate immune cell subsets, including Kupffer cells (KCs), innate lymphoid cells (ILCs) like Natural Killer (NK) cells and ILC-like unconventional T cells - namely Natural Killer T cells (NKT), γδ T cells and Mucosal-associated Invariant T cells (MAIT). These cells reside in the liver in a memory-effector state, so they respond quickly to trigger appropriate responses. The contribution of aberrant innate immunity to inflammatory liver diseases is now being better understood. In particular, we are beginning to understand how specific innate immune subsets trigger chronic liver inflammation, which ultimately results in hepatic fibrosis. In this review, we consider the roles of specific innate immune cell subsets in early inflammation in human liver disease.
Collapse
Affiliation(s)
- Jordi Yang Zhou
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- *Correspondence: Jordi Yang Zhou,
| |
Collapse
|
33
|
Yang Zhou J, Werner JM, Glehr G, Geissler EK, Hutchinson JA, Kronenberg K. Identification and Isolation of Type II NKT Cell Subsets in Human Blood and Liver. Front Immunol 2022; 13:898473. [PMID: 35720369 PMCID: PMC9202826 DOI: 10.3389/fimmu.2022.898473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Background Steatotic livers are more prone to rejection, but are often transplanted owing to the shortage of available organs. Type II NKT (T2NKT) cells are liver-resident lymphocytes that react to lipids presented by CD1d. The role of T2NKT cells in rejection of fatty liver transplants is unclear, partly because of a lack of T2NKT cell markers and their very low frequency in blood. Here, we quantify human T2NKT cells in blood and liver tissue by flow cytometry and provide a strategy for their enrichment and expansion. Methods Human T2NKT cells were identified as CD3+ CD56+ CD161+ TCR-γᵹ- TCRVα7.2- and TCRVα24- cells. T2NKT cells were enriched from blood by sequential positive selection using CD56 and CD3 microbeads. These were subsequently FACS-sorted to purity then expanded in vitro for 3 weeks using anti-CD3/CD28 beads and TGF-β1. Results The frequency of human T2NKT cells in blood was very low (0.8 ± 0.4% of CD3+ T cells) but they were a more abundant population in liver (6.3 ± 0.9%). Enriched T2NKT cells expressed the transcription factor PLZF. A novel subset of FoxP3+ T2NKT cells was discovered in blood and liver tissue. T2NKT cells were expanded in culture by 15- to 28-fold over 3 weeks, during which time they maintained expression of all identifying markers, including PLZF and FoxP3. Conclusions Our work defines new strategies for identifying and isolating T2NKT cells from human blood and liver tissue. We showed that this rare population can be expanded in vitro in order to obtain experimentally amenable cell numbers. Further, we identified a novel T2NKT cell subset that stably expresses FoxP3, which might play a role in regulating innate-like lymphocyte responses in steatotic liver transplants.
Collapse
Affiliation(s)
- Jordi Yang Zhou
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany.,Leibniz Institute for Immunotherapy, Regensburg, Germany
| | - Jens M Werner
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Gunther Glehr
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany.,Leibniz Institute for Immunotherapy, Regensburg, Germany.,Fraunhofer-Institute for Toxicology and Experimental Medicine Institute for Toxicology and Experimental Medicine-Regensburg (ITEM-R), Regensburg, Germany.,Regensburg International Graduate School of Life Sciences, University of Regensburg, Regensburg, Germany
| | - James A Hutchinson
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | | |
Collapse
|
34
|
Yue C, Gao S, Li S, Xing Z, Qian H, Hu Y, Wang W, Hua C. TIGIT as a Promising Therapeutic Target in Autoimmune Diseases. Front Immunol 2022; 13:911919. [PMID: 35720417 PMCID: PMC9203892 DOI: 10.3389/fimmu.2022.911919] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/13/2022] [Indexed: 12/19/2022] Open
Abstract
Co-inhibitory receptors (IRs) are molecules that protect host against autoimmune reactions and maintain peripheral self-tolerance, playing an essential role in maintaining immune homeostasis. In view of the substantial clinical progresses of negative immune checkpoint blockade in cancer treatment, the role of IRs in autoimmune diseases is also obvious. Several advances highlighted the substantial impacts of T cell immunoglobulin and ITIM domain (TIGIT), a novel IR, in autoimmunity. Blockade of TIGIT pathway exacerbates multiple autoimmune diseases, whereas enhancement of TIGIT function has been shown to alleviate autoimmune settings in mice. These data suggested that TIGIT pathway can be manipulated to achieve durable tolerance to treat autoimmune disorders. In this review, we provide an overview of characteristics of TIGIT and its role in autoimmunity. We then discuss recent approaches and future directions to leverage our knowledge of TIGIT as therapeutic target in autoimmune diseases.
Collapse
Affiliation(s)
- Chenran Yue
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou, China
| | - Shuting Li
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhouhang Xing
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hengrong Qian
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ying Hu
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenqian Wang
- Department of Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
35
|
Lin Z, Rao Z, Chen J, Chu H, Zhou J, Yang L, Quan D, Bai Y. Bioactive Decellularized Extracellular Matrix Hydrogel Microspheres Fabricated Using a Temperature-Controlling Microfluidic System. ACS Biomater Sci Eng 2022; 8:1644-1655. [PMID: 35357124 DOI: 10.1021/acsbiomaterials.1c01474] [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] [Indexed: 02/07/2023]
Abstract
Hydrogel microspheres have drawn great attention as functional three-dimensional (3D) microcarriers for cell attachment and growth, which have shown great potential in cell-based therapies and biomedical research. Hydrogels derived from a decellularized extracellular matrix (dECM) retain the intrinsic physical and biological cues from the native tissues, which often exhibit high bioactivity and tissue-specificity in promoting tissue regeneration. Herein, a novel two-stage temperature-controlling microfluidic system was developed which enabled production of pristine dECM hydrogel microspheres in a high-throughput manner. Porcine decellularized peripheral nerve matrix (pDNM) was used as the model raw dECM material for continuous generation of pDNM microgels without additional supporting materials or chemical crosslinking. The sizes of the microspheres were well-controlled by tuning the feed ratios of water/oil phases into the microfluidic device. The resulting pDNM microspheres (pDNM-MSs) were relatively stable, which maintained a spherical shape and a nanofibrous ultrastructure for at least 14 days. Schwann cells and PC12 cells preseeded on the pDNM-MSs not only showed excellent viability and an adhesive property, but also promoted cell extension compared to the commercially available gelatin microspheres. Moreover, primary neural stem/progenitor cells attached well to the pDNM-MSs, which further facilitated their proliferation. The successfully fabricated dECM hydrogel microspheres provided a highly bioactive microenvironment for 3D cell culture and functionalization, which showed promising potential in versatile biomedical applications.
Collapse
Affiliation(s)
- Zudong Lin
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Zilong Rao
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Jiaxin Chen
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Hanyu Chu
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Jing Zhou
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Liqun Yang
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Daping Quan
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China.,Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Ying Bai
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| |
Collapse
|
36
|
Liver ischaemia-reperfusion injury: a new understanding of the role of innate immunity. Nat Rev Gastroenterol Hepatol 2022; 19:239-256. [PMID: 34837066 DOI: 10.1038/s41575-021-00549-8] [Citation(s) in RCA: 194] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/29/2021] [Indexed: 02/08/2023]
Abstract
Liver ischaemia-reperfusion injury (LIRI), a local sterile inflammatory response driven by innate immunity, is one of the primary causes of early organ dysfunction and failure after liver transplantation. Cellular damage resulting from LIRI is an important risk factor not only for graft dysfunction but also for acute and even chronic rejection and exacerbates the shortage of donor organs for life-saving liver transplantation. Hepatocytes, liver sinusoidal endothelial cells and Kupffer cells, along with extrahepatic monocyte-derived macrophages, neutrophils and platelets, are all involved in LIRI. However, the mechanisms underlying the responses of these cells in the acute phase of LIRI and how these responses are orchestrated to control and resolve inflammation and achieve homeostatic tissue repair are not well understood. Technological advances allow the tracking of cells to better appreciate the role of hepatic macrophages and platelets (such as their origin and immunomodulatory and tissue-remodelling functions) and hepatic neutrophils (such as their selective recruitment, anti-inflammatory and tissue-repairing functions, and formation of extracellular traps and reverse migration) in LIRI. In this Review, we summarize the role of macrophages, platelets and neutrophils in LIRI, highlight unanswered questions, and discuss prospects for innovative therapeutic regimens against LIRI in transplant recipients.
Collapse
|
37
|
Moreau JM, Velegraki M, Bolyard C, Rosenblum MD, Li Z. Transforming growth factor-β1 in regulatory T cell biology. Sci Immunol 2022; 7:eabi4613. [PMID: 35302863 PMCID: PMC10552796 DOI: 10.1126/sciimmunol.abi4613] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transforming growth factor-β1 (TGF-β1) is inextricably linked to regulatory T cell (Treg) biology. However, precisely untangling the role for TGF-β1 in Treg differentiation and function is complicated by the pleiotropic and context-dependent activity of this cytokine and the multifaceted biology of Tregs. Among CD4+ T cells, Tregs are the major producers of latent TGF-β1 and are uniquely able to activate this cytokine via expression of cell surface docking receptor glycoprotein A repetitions predominant (GARP) and αv integrins. Although a preponderance of evidence indicates no essential roles for Treg-derived TGF-β1 in Treg immunosuppression, TGF-β1 signaling is crucial for Treg development in the thymus and periphery. Furthermore, active TGF-β1 instructs the differentiation of other T cell subsets, including TH17 cells. Here, we will review TGF-β1 signaling in Treg development and function and discuss knowledge gaps, future research, and the TGF-β1/Treg axis in the context of cancer immunotherapy and fibrosis.
Collapse
Affiliation(s)
- Joshua M. Moreau
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Maria Velegraki
- Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center—James Cancer Hospital, Columbus, OH, USA
| | - Chelsea Bolyard
- Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center—James Cancer Hospital, Columbus, OH, USA
| | - Michael D. Rosenblum
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, the Ohio State University Comprehensive Cancer Center—James Cancer Hospital, Columbus, OH, USA
| |
Collapse
|
38
|
Dendritic cell-based cancer immunotherapy in the era of immune checkpoint inhibitors: From bench to bedside. Life Sci 2022; 297:120466. [PMID: 35271882 DOI: 10.1016/j.lfs.2022.120466] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) can present tumoral antigens to T-cells and stimulate T-cell-mediated anti-tumoral immune responses. In addition to uptaking, processing, and presenting tumoral antigens to T-cells, co-stimulatory signals have to be established between DCs with T-cells to develop anti-tumoral immune responses. However, most of the tumor-infiltrated immune cells are immunosuppressive in the tumor microenvironment (TME), paving the way for immune evasion of tumor cells. This immunosuppressive TME has also been implicated in suppressing the DC-mediated anti-tumoral immune responses, as well. Various factors, i.e., immunoregulatory cells, metabolic factors, tumor-derived immunosuppressive factors, and inhibitory immune checkpoint molecules, have been implicated in developing the immunosuppressive TME. Herein, we aimed to review the biology of DCs in developing T-cell-mediated anti-tumoral immune responses, the significance of immunoregulatory cells in the TME, metabolic barriers contributing to DCs dysfunction in the TME, tumor-derived immunosuppressive factors, and inhibitory immune checkpoint molecules in DC-based cell therapy outcomes. With reviewing the ongoing clinical trials, we also proposed a novel therapeutic strategy to increase the efficacy of DC-based cell therapy. Indeed, the combination of DC-based cell therapy with monoclonal antibodies against novel immune checkpoint molecules can be a promising strategy to increase the response rate of patients with cancers.
Collapse
|
39
|
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]
|
40
|
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
|
41
|
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
|
42
|
Yu S, Lu J. Macrophages in transplant rejection. Transpl Immunol 2022; 71:101536. [PMID: 35017096 DOI: 10.1016/j.trim.2022.101536] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 11/17/2022]
Abstract
Transplant rejection is one of the primary factors leading to loss of allograft function, and macrophages are involved in allograft rejection. Macrophages polarize into different phenotypes according to stimulation by different external factors. Different types of macrophages play distinct roles in inflammation, tumors, and autoimmune diseases and are involved in transplant rejection. In this review, we introduce the origin and migration of macrophages, outline the classification of macrophages and their polarization mechanisms, and review the currently understood mechanisms of their involvement in transplant rejection. Finally, we discuss the regulation of macrophage polarization and miRNA expression with respect to transplant rejection, which is important for the development of new anti-rejection therapies.
Collapse
Affiliation(s)
- Shaochen Yu
- Department of Emergency and Critical Care Medicine, Guangdong Second Provincial General Hospital, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, Guangdong 510317, China.
| | - Jian Lu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui 230022, China.
| |
Collapse
|
43
|
Bottomley MJ, Brook MO, Shankar S, Hester J, Issa F. Towards regulatory cellular therapies in solid organ transplantation. Trends Immunol 2022; 43:8-21. [PMID: 34844848 DOI: 10.1016/j.it.2021.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/10/2021] [Accepted: 11/01/2021] [Indexed: 01/03/2023]
Abstract
Organ transplantation is a modern medical success story. However, since its inception it has been limited by the need for pharmacological immunosuppression. Regulatory cellular therapies offer an attractive solution to these challenges by controlling transplant alloresponses through multiple parallel suppressive mechanisms. A number of cell types have seen an accelerated development into human trials and are now on the threshold of a long-awaited breakthrough in personalized transplant therapeutics. Here we assess recent developments with a focus on the most likely candidates, some of which have already facilitated successful immunosuppression withdrawal in early clinical trials. We propose that this may constitute a promising approach in clinical transplantation but also evaluate outstanding issues in the field, providing cause for cautious optimism.
Collapse
Affiliation(s)
- Matthew J Bottomley
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford Transplant Centre, Churchill Hospital, Oxford, UK
| | - Matthew O Brook
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford Transplant Centre, Churchill Hospital, Oxford, UK
| | - Sushma Shankar
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford Transplant Centre, Churchill Hospital, Oxford, UK
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
44
|
Li J, Thomson AW, Rogers NM. Myeloid and Mesenchymal Stem Cell Therapies for Solid Organ Transplant Tolerance. Transplantation 2021; 105:e303-e321. [PMID: 33756544 PMCID: PMC8455706 DOI: 10.1097/tp.0000000000003765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Transplantation is now performed globally as a routine procedure. However, the increased demand for donor organs and consequent expansion of donor criteria has created an imperative to maximize the quality of these gains. The goal is to balance preservation of allograft function against patient quality-of-life, despite exposure to long-term immunosuppression. Elimination of immunosuppressive therapy to avoid drug toxicity, with concurrent acceptance of the allograft-so-called operational tolerance-has proven elusive. The lack of recent advances in immunomodulatory drug development, together with advances in immunotherapy in oncology, has prompted interest in cell-based therapies to control the alloimmune response. Extensive experimental work in animals has characterized regulatory immune cell populations that can induce and maintain tolerance, demonstrating that their adoptive transfer can promote donor-specific tolerance. An extension of this large body of work has resulted in protocols for manufacture, as well as early-phase safety and feasibility trials for many regulatory cell types. Despite the excitement generated by early clinical trials in autoimmune diseases and organ transplantation, there is as yet no clinically validated, approved regulatory cell therapy for transplantation. In this review, we summarize recent advances in this field, with a focus on myeloid and mesenchymal cell therapies, including current understanding of the mechanisms of action of regulatory immune cells, and clinical trials in organ transplantation using these cells as therapeutics.
Collapse
Affiliation(s)
- Jennifer Li
- Center of Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Angus W Thomson
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Natasha M Rogers
- Center of Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
45
|
Hosaka Y, Itoh K, Matsutani S, Kawate S, Miura A, Mizoura Y, Yamada S, Konno H, Grave E, Nagata K, Wakui H, Itoh H. Fermented food Tempeh induces interleukin 12 and enhances macrophage phagocytosis. J Food Biochem 2021; 45:e13958. [PMID: 34611901 DOI: 10.1111/jfbc.13958] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/08/2021] [Accepted: 09/20/2021] [Indexed: 01/20/2023]
Abstract
It is known that lactic acid bacteria induce the IL-12. The IL-12 activates NK cells and promotes the production of IFN-γ. The IFN-γ activates macrophages resulting in enhanced phagocytosis and bactericidal activity. We have been investigating fermented foods that activate the immune function. In this study, we investigated the IL-12 inducibility of fermented foods using the specific antibody. Fermented soybean foods such as Tempeh and Natto are attracting attention in terms of nutrition, functionality, and food problems. In this study, Tempeh induced 1,080 µg/ml of IL-12, and IFN-γ associated with the induction of IL-12 was also induced at 682 µg/ml. This was more than twice the induced intensity of PBS. On the contrary, Natto hardly induced IL-12 and IFN-γ. Tempeh also accelerated phagocytosis of the macrophage THP-1 cells. In this study, it was found that the fermented soybean-derived food, Tempeh, has a function of activating the immune function. This is the first report that Tempeh activates innate immunity. PRACTICAL APPLICATIONS: Tempeh, a fermented soybean food induced the IL-12 and IFN-γ production and the increase of macrophage phagocytosis in this study suggested a new function to enhance immunity. Tempeh is also expected to be effective in preventing lifestyle diseases. Fermented soybean products of Tempeh was considered to be a very useful health food for the problems of modern society such as maintaining health by eating, improving immunity, and ingesting vegetable protein due to diversifying food.
Collapse
Affiliation(s)
- Yoshihito Hosaka
- Department of Life Science, Graduate School and Faculty of Engineering Science, Akita University, Akita, Japan.,Akita Konno Co., Ltd., Akita, Japan
| | - Kei Itoh
- Department of Life Science, Graduate School and Faculty of Engineering Science, Akita University, Akita, Japan
| | - Shun Matsutani
- Department of Life Science, Graduate School and Faculty of Engineering Science, Akita University, Akita, Japan
| | - Shinya Kawate
- Department of Life Science, Graduate School and Faculty of Engineering Science, Akita University, Akita, Japan
| | - Atsuko Miura
- Department of Life Science, Graduate School and Faculty of Engineering Science, Akita University, Akita, Japan
| | - Yukaze Mizoura
- Department of Life Science, Graduate School and Faculty of Engineering Science, Akita University, Akita, Japan
| | - Sayumi Yamada
- Department of Life Science, Graduate School and Faculty of Engineering Science, Akita University, Akita, Japan
| | | | - Ewa Grave
- Department of Life Science, Graduate School and Faculty of Engineering Science, Akita University, Akita, Japan
| | - Koji Nagata
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Hideki Wakui
- Department of Life Science, Graduate School and Faculty of Engineering Science, Akita University, Akita, Japan
| | - Hideaki Itoh
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
46
|
Suuring M, Moreau A. Regulatory Macrophages and Tolerogenic Dendritic Cells in Myeloid Regulatory Cell-Based Therapies. Int J Mol Sci 2021; 22:7970. [PMID: 34360736 PMCID: PMC8348814 DOI: 10.3390/ijms22157970] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022] Open
Abstract
Myeloid regulatory cell-based therapy has been shown to be a promising cell-based medicinal approach in organ transplantation and for the treatment of autoimmune diseases, such as type 1 diabetes, rheumatoid arthritis, Crohn's disease and multiple sclerosis. Dendritic cells (DCs) are the most efficient antigen-presenting cells and can naturally acquire tolerogenic properties through a variety of differentiation signals and stimuli. Several subtypes of DCs have been generated using additional agents, including vitamin D3, rapamycin and dexamethasone, or immunosuppressive cytokines, such as interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β). These cells have been extensively studied in animals and humans to develop clinical-grade tolerogenic (tol)DCs. Regulatory macrophages (Mregs) are another type of protective myeloid cell that provide a tolerogenic environment, and have mainly been studied within the context of research on organ transplantation. This review aims to thoroughly describe the ex vivo generation of tolDCs and Mregs, their mechanism of action, as well as their therapeutic application and assessment in human clinical trials.
Collapse
Affiliation(s)
| | - Aurélie Moreau
- Centre de Recherche en Transplantation et Immunologie—UMR1064, INSERM—ITUN, Nantes Université, CHU Nantes, 44000 Nantes, France;
| |
Collapse
|
47
|
Preclinical Assessment of Autologous Tolerogenic Dendritic Cells From End-stage Renal Disease Patients. Transplantation 2021; 105:832-841. [PMID: 32433241 DOI: 10.1097/tp.0000000000003315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Kidney transplantation is the therapeutic of choice for patients with kidney failure. While immunosuppressive drugs can control graft rejection, their use is associated with increased infections and cancer, and they do not effectively control chronic graft rejection. Cell therapy is an attractive strategy to minimize the use of pharmacological drugs. METHODS We recently developed a protocol to generate human monocyte-derived autologous tolerogenic dendritic cells (ATDCs) from healthy volunteers. Herein, we transferred the ATDC manufacturing protocol to a Good Manufacturing Practice (GMP)-compliant facility. Furthermore, we compared the phenotype and in vitro functions of ATDCs generated from patients with end-stage renal disease to those generated from healthy volunteers. RESULTS We describe the critical steps for GMP-compliant production of ATDCs and define the quality criteria required to allow release of the cell products. Furthermore, we showed that ATDCs generated from healthy volunteers and patients with kidney failure display the same tolerogenic profile based on their phenotype, resistance to maturation, and ability to modulate T-cell responses. CONCLUSIONS Together, these results allowed us to define the production process and the quality criteria for the release of ATDCs before their administration in patients receiving a kidney transplant.
Collapse
|
48
|
Macedo C, Tran LM, Zahorchak AF, Dai H, Gu X, Ravichandran R, Mohanakumar T, Elinoff B, Zeevi A, Styn MA, Humar A, Lakkis FG, Metes DM, Thomson AW. Donor-derived regulatory dendritic cell infusion results in host cell cross-dressing and T cell subset changes in prospective living donor liver transplant recipients. Am J Transplant 2021; 21:2372-2386. [PMID: 33171019 PMCID: PMC8215622 DOI: 10.1111/ajt.16393] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/13/2020] [Accepted: 11/01/2020] [Indexed: 01/25/2023]
Abstract
Regulatory dendritic cells (DCreg) promote transplant tolerance following their adoptive transfer in experimental animals. We investigated the feasibility, safety, fate, and impact on host T cells of donor monocyte-derived DCreg infused into prospective, living donor liver transplant patients, 7 days before transplantation. The DCreg expressed a tolerogenic gene transcriptional profile, high cell surface programed death ligand-1 (PD-L1):CD86 ratios, high IL-10/no IL-12 productivity and poor ability to stimulate allogeneic T cell proliferation. Target DCreg doses (range 2.5-10 × 106 cells/kg) were achieved in all but 1 of 15 recipients, with no infusion reactions. Following DCreg infusion, transiently elevated levels of donor HLA and immunoregulatory PD-L1, CD39, and CD73 were detected in circulating small extracellular vesicles. At the same time, flow and advanced image stream analysis revealed intact DCreg and "cross-dressing" of host DCs in blood and lymph nodes. PD-L1 co-localization with donor HLA was observed at higher levels than with recipient HLA. Between DCreg infusion and transplantation, T-bethi Eomeshi memory CD8+ T cells decreased, whereas regulatory (CD25hi CD127- Foxp3+ ): T-bethi Eomeshi CD8+ T cell ratios increased. Thus, donor-derived DCreg infusion may induce systemic changes in host antigen-presenting cells and T cells potentially conducive to modulated anti-donor immune reactivity at the time of transplant.
Collapse
Affiliation(s)
- Camila Macedo
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lillian M. Tran
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Alan F. Zahorchak
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Helong Dai
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xinyan Gu
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | | | - Beth Elinoff
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Adriana Zeevi
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Department of Pathology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania,Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Mindi A. Styn
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Abhinav Humar
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Fadi G. Lakkis
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Diana M. Metes
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Angus W. Thomson
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania,Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
49
|
Yu K, Hammerschmidt SI, Permanyer M, Galla M, Rothe M, Zheng X, Werth K, Martens R, Lueder Y, Janssen A, Friedrichsen M, Bernhardt G, Förster R. Targeted delivery of regulatory macrophages to lymph nodes interferes with T cell priming by preventing the formation of stable immune synapses. Cell Rep 2021; 35:109273. [PMID: 34161766 DOI: 10.1016/j.celrep.2021.109273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 02/05/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Immunosuppressive myeloid cells are frequently induced in tumors and attenuate anti-tumor effector functions. In this study, we differentiate immunosuppressive regulatory macrophages (Mregs) from hematopoietic progenitors and test their potential to suppress adaptive immune responses in lymph nodes. Targeted delivery of Mregs to lymph nodes is facilitated by retroviral overexpression of the chemokine receptor CCR7 and intra-lymphatic cell application. Delivery of Mregs completely abolishes the priming of cognate CD8 cells and strongly reduces delayed-type hypersensitivity reactions. Mreg-mediated T cell suppression requires cell-cell contact-regulated nitric oxide production. Two-photon microscopy reveals that nitric oxide produced by Mregs reduces the interaction duration between dendritic cells and T cells. Exposure of activated T cells to nitric oxide strongly reduces their binding to ICAM-1, indicating that nitrosylation of proteins involved in cell adhesion affects synapse formation. Thus, this study identifies a mechanism of myeloid cell-mediated immune suppression and provides an approach for its therapeutic use.
Collapse
Affiliation(s)
- Kai Yu
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Marc Permanyer
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Rothe
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Xiang Zheng
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Kathrin Werth
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Rieke Martens
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Yvonne Lueder
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Anika Janssen
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Günter Bernhardt
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany; German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 30625 Hannover, Germany.
| |
Collapse
|
50
|
Ostroumov D, Duong S, Wingerath J, Woller N, Manns MP, Timrott K, Kleine M, Ramackers W, Roessler S, Nahnsen S, Czemmel S, Dittrich-Breiholz O, Eggert T, Kühnel F, Wirth TC. Transcriptome Profiling Identifies TIGIT as a Marker of T-Cell Exhaustion in Liver Cancer. Hepatology 2021; 73:1399-1418. [PMID: 32716559 DOI: 10.1002/hep.31466] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/09/2020] [Accepted: 06/13/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Programmed death 1 (PD-1) checkpoint inhibition has shown promising results in patients with hepatocellular carcinoma, inducing objective responses in approximately 20% of treated patients. The roles of other coinhibitory molecules and their individual contributions to T-cell dysfunction in liver cancer, however, remain largely elusive. APPROACH AND RESULTS We performed a comprehensive mRNA profiling of cluster of differentiation 8 (CD8) T cells in a murine model of autochthonous liver cancer by comparing the transcriptome of naive, functional effector, and exhausted, tumor-specific CD8 T cells. Subsequently, we functionally validated the role of identified genes in T-cell exhaustion. Our results reveal a unique transcriptome signature of exhausted T cells and demonstrate that up-regulation of the inhibitory immune receptor T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitor motif domains (TIGIT) represents a hallmark in the process of T-cell exhaustion in liver cancer. Compared to PD-1, expression of TIGIT more reliably identified exhausted CD8 T cells at different stages of their differentiation. In combination with PD-1 inhibition, targeting of TIGIT with antagonistic antibodies resulted in synergistic inhibition of liver cancer growth in immunocompetent mice. Finally, we demonstrate expression of TIGIT on tumor-infiltrating CD8 T cells in tissue samples of patients with hepatocellular carcinoma and intrahepatic cholangiocarcinoma and identify two subsets of patients based on differential expression of TIGIT on tumor-specific T cells. CONCLUSIONS Our transcriptome analysis provides a valuable resource for the identification of key pathways involved in T-cell exhaustion in patients with liver cancer and identifies TIGIT as a potential target in checkpoint combination therapies.
Collapse
Affiliation(s)
- Dmitrij Ostroumov
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Steven Duong
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jessica Wingerath
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Norman Woller
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Michael P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Kai Timrott
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Moritz Kleine
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Wolf Ramackers
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sven Nahnsen
- Quantitative Biology Center (QBIC), University of Tübingen, Tübingen, Germany
| | - Stefan Czemmel
- Quantitative Biology Center (QBIC), University of Tübingen, Tübingen, Germany
| | | | - Tobias Eggert
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Florian Kühnel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Thomas C Wirth
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|