1
|
Ruiz Pérez M, Vandenabeele P, Tougaard P. The thymus road to a T cell: migration, selection, and atrophy. Front Immunol 2024; 15:1443910. [PMID: 39257583 PMCID: PMC11384998 DOI: 10.3389/fimmu.2024.1443910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024] Open
Abstract
The thymus plays a pivotal role in generating a highly-diverse repertoire of T lymphocytes while preventing autoimmunity. Thymus seeding progenitors (TSPs) are a heterogeneous group of multipotent progenitors that migrate to the thymus via CCR7 and CCR9 receptors. While NOTCH guides thymus progenitors toward T cell fate, the absence or disruption of NOTCH signaling renders the thymus microenvironment permissive to other cell fates. Following T cell commitment, developing T cells undergo multiple selection checkpoints by engaging with the extracellular matrix, and interacting with thymic epithelial cells (TECs) and other immune subsets across the different compartments of the thymus. The different selection checkpoints assess the T cell receptor (TCR) performance, with failure resulting in either repurposing (agonist selection), or cell death. Additionally, environmental cues such as inflammation and endocrine signaling induce acute thymus atrophy, contributing to the demise of most developing T cells during thymic selection. We discuss the occurrence of acute thymus atrophy in response to systemic inflammation. The thymus demonstrates high plasticity, shaping inflammation by abrogating T cell development and undergoing profound structural changes, and facilitating regeneration and restoration of T cell development once inflammation is resolved. Despite the challenges, thymic selection ensures a highly diverse T cell repertoire capable of discerning between self and non-self antigens, ultimately egressing to secondary lymphoid organs where they complete their maturation and exert their functions.
Collapse
Affiliation(s)
- Mario Ruiz Pérez
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Tougaard
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
2
|
Seyedhassantehrani N, Burns CS, Verrinder R, Okafor V, Abbasizadeh N, Spencer JA. Intravital two-photon microscopy of the native mouse thymus. PLoS One 2024; 19:e0307962. [PMID: 39088574 PMCID: PMC11293686 DOI: 10.1371/journal.pone.0307962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/15/2024] [Indexed: 08/03/2024] Open
Abstract
The thymus, a key organ in the adaptive immune system, is sensitive to a variety of insults including cytotoxic preconditioning, which leads to atrophy, compression of the blood vascular system, and alterations in hemodynamics. Although the thymus has innate regenerative capabilities, the production of T cells relies on the trafficking of lymphoid progenitors from the bone marrow through the altered thymic blood vascular system. Our understanding of thymic blood vascular hemodynamics is limited due to technical challenges associated with accessing the native thymus in live mice. To overcome this challenge, we developed an intravital two-photon imaging method to visualize the native thymus in vivo and investigated functional changes to the vascular system following sublethal irradiation. We quantified blood flow velocity and shear rate in cortical blood vessels and identified a subtle but significant increase in vessel leakage and diameter ~24 hrs post-sublethal irradiation. Ex vivo whole organ imaging of optically cleared thymus lobes confirmed a disruption of the thymus vascular structure, resulting in an increase in blood vessel diameter and vessel area, and concurrent thymic atrophy. This novel two-photon intravital imaging method enables a new paradigm for directly investigating the thymic microenvironment in vivo.
Collapse
Affiliation(s)
- Negar Seyedhassantehrani
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
| | - Christian S. Burns
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
| | - Ruth Verrinder
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
| | - Victoria Okafor
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
| | - Nastaran Abbasizadeh
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
| | - Joel A. Spencer
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
- Health Science Research Institute, University of California Merced, Merced, California, United States of America
| |
Collapse
|
3
|
Lee BC. Challenges and innovations in hematopoietic stem cell transplantation: exploring bone marrow niches and new model systems. BMB Rep 2024; 57:352-362. [PMID: 38919014 PMCID: PMC11362137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/27/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) remains an indispensable therapeutic strategy for various hematological diseases. This review discusses the pivotal role of bone marrow (BM) niches in influencing the efficacy of HSCT and evaluates the current animal models, emphasizing their limitations and the need for alternative models. Traditional animal models, mainly murine xenograft, have provided significant insights, but due to species-specific differences, are often constrained from accurately mimicking human physiological responses. These limitations highlight the importance of developing alternative models that can more realistically replicate human hematopoiesis. Emerging models that include BM organoids and BM-on-a-chip microfluidic systems promise enhanced understanding of HSCT dynamics. These models aim to provide more accurate simulations of the human BM microenvironment, potentially leading to improved preclinical assessments and therapeutic outcomes. This review highlights the complexities of the BM niche, discusses the limitations of current models, and suggests directions for future research using advanced model systems. [BMB Reports 2024; 57(8): 352-362].
Collapse
Affiliation(s)
- Byung-Chul Lee
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
- Research Institute of Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| |
Collapse
|
4
|
Tong Q, Yao L, Su M, Yang YG, Sun L. Thymocyte migration and emigration. Immunol Lett 2024; 267:106861. [PMID: 38697225 DOI: 10.1016/j.imlet.2024.106861] [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: 09/08/2023] [Revised: 12/15/2023] [Accepted: 04/26/2024] [Indexed: 05/04/2024]
Abstract
Hematopoietic precursors (HPCs) entering into the thymus undergo a sequential process leading to the generation of a variety of T cell subsets. This developmental odyssey unfolds in distinct stages within the thymic cortex and medulla, shaping the landscape of T cell receptor (TCR) expression and guiding thymocytes through positive and negative selection. Initially, early thymic progenitors (ETPs) take residence in the thymic cortex, where thymocytes begin to express their TCR and undergo positive selection. Subsequently, thymocytes transition to the thymic medulla, where they undergo negative selection. Both murine and human thymocyte development can be broadly classified into distinct stages based on the expression of CD4 and CD8 coreceptors, resulting in categorizations as double negative (DN), double positive (DP) or single positive (SP) cells. Thymocyte migration to the appropriate thymic microenvironment at the right differentiation stage is pivotal for the development and the proper functioning of T cells, which is critical for adaptive immune responses. The journey of lymphoid progenitor cells into the T cell developmental pathway hinges on an ongoing dialogue between the differentiating cell and the signals emanating from the thymus niche. Herein, we review the contribution of the key factors mentioned above for the localization, migration and emigration of thymocytes.
Collapse
Affiliation(s)
- Qingyue Tong
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Liyu Yao
- Department of Pediatric Surgery, The First Hospital of Jilin University, Changchun, China
| | - Mengting Su
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China.
| | - Liguang Sun
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China.
| |
Collapse
|
5
|
Watabe T, Kaneda-Nakashima K, Kadonaga Y, Ooe K, Sampunta T, Hirose N, Yin X, Haba H, Kon Y, Toyoshima A, Cardinale J, Giesel FL, Fukase K, Tomiyama N, Shirakami Y. Preclinical Evaluation of Biodistribution and Toxicity of [ 211At]PSMA-5 in Mice and Primates for the Targeted Alpha Therapy against Prostate Cancer. Int J Mol Sci 2024; 25:5667. [PMID: 38891856 PMCID: PMC11172375 DOI: 10.3390/ijms25115667] [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: 04/17/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Astatine (211At) is a cyclotron-produced alpha emitter with a physical half-life of 7.2 h. In our previous study, the 211At-labeled prostate-specific membrane antigen (PSMA) compound ([211At]PSMA-5) exhibited excellent tumor growth suppression in a xenograft model. We conducted preclinical biodistribution and toxicity studies for the first-in-human clinical trial. [211At]PSMA-5 was administered to both normal male ICR mice (n = 85) and cynomolgus monkeys (n = 2). The mice were divided into four groups for the toxicity study: 5 MBq/kg, 12 MBq/kg, 35 MBq/kg, and vehicle control, with follow-ups at 1 day (n = 10 per group) and 14 days (n = 5 per group). Monkeys were observed 24 h post-administration of [211At]PSMA-5 (9 MBq/kg). Blood tests and histopathological examinations were performed at the end of the observation period. Blood tests in mice indicated no significant myelosuppression or renal dysfunction. However, the monkeys displayed mild leukopenia 24 h post-administration. Despite the high accumulation in the kidneys and thyroid, histological analysis revealed no abnormalities. On day 1, dose-dependent single-cell necrosis/apoptosis was observed in the salivary glands of mice and intestinal tracts of both mice and monkeys. Additionally, tingible body macrophages in the spleen and lymph nodes indicated phagocytosis of apoptotic B lymphocytes. Cortical lymphopenia (2/10) in the thymus and a decrease in the bone marrow cells (9/10) were observed in the 35 MBq/kg group in mice. These changes were transient, with no irreversible toxicity observed in mice 14 days post-administration. This study identified no severe toxicities associated with [211At]PSMA-5, highlighting its potential as a next-generation targeted alpha therapy for prostate cancer. The sustainable production of 211At using a cyclotron supports its applicability for clinical use.
Collapse
Affiliation(s)
- Tadashi Watabe
- Department of Radiology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Institute for Radiation Sciences, Osaka University, Osaka 565-0871, Japan
| | - Kazuko Kaneda-Nakashima
- Institute for Radiation Sciences, Osaka University, Osaka 565-0871, Japan
- Core for Medicine and Science Collaborative Research and Education, Project Research Center for Fundamental Sciences, Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| | - Yuichiro Kadonaga
- Institute for Radiation Sciences, Osaka University, Osaka 565-0871, Japan
| | - Kazuhiro Ooe
- Institute for Radiation Sciences, Osaka University, Osaka 565-0871, Japan
| | - Thosapol Sampunta
- Department of Radiology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Naoki Hirose
- Institute of Experimental Animal Sciences, Faculty of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Xiaojie Yin
- Nishina Center for Accelerator-Based Science, RIKEN, Wako 351-0198, Japan
| | - Hiromitsu Haba
- Nishina Center for Accelerator-Based Science, RIKEN, Wako 351-0198, Japan
| | - Yukiyoshi Kon
- Institute for Radiation Sciences, Osaka University, Osaka 565-0871, Japan
| | - Atsushi Toyoshima
- Institute for Radiation Sciences, Osaka University, Osaka 565-0871, Japan
| | - Jens Cardinale
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Frederik L. Giesel
- Institute for Radiation Sciences, Osaka University, Osaka 565-0871, Japan
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Duesseldorf, 40225 Duesseldorf, Germany
| | - Koichi Fukase
- Institute for Radiation Sciences, Osaka University, Osaka 565-0871, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| | - Noriyuki Tomiyama
- Department of Radiology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Institute for Radiation Sciences, Osaka University, Osaka 565-0871, Japan
| | | |
Collapse
|
6
|
Learmonth M, Corker A, Dasgupta S, DeLeon-Pennell KY. Regulation of cardiac fibroblasts by lymphocytes after a myocardial infarction: playing in the major league. Am J Physiol Heart Circ Physiol 2023; 325:H553-H561. [PMID: 37450290 PMCID: PMC10538980 DOI: 10.1152/ajpheart.00250.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Cardiac fibrosis is a pathological condition characterized by excessive accumulation of extracellular matrix components within the myocardium, which can lead to impaired cardiac function and heart failure. Studies have shown that lymphocytes including B and T cells play important roles in the development and progression of cardiac fibrosis after a myocardial infarction. In this review, we focus on the regulation of cardiac fibrosis by lymphocyte subsets, with a particular emphasis on CD4+ and CD8+ T cells and their effects on fibroblasts and cardiac remodeling. We also highlight areas for further exploration of the interactions between T cells and fibroblasts necessary for understanding and treating cardiac fibrosis and heart failure.
Collapse
Affiliation(s)
- Maya Learmonth
- College of Graduate Studies, Medical University of South Carolina, Charleston, South Carolina, United States
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Alexa Corker
- College of Graduate Studies, Medical University of South Carolina, Charleston, South Carolina, United States
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Shaoni Dasgupta
- College of Graduate Studies, Medical University of South Carolina, Charleston, South Carolina, United States
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Kristine Y DeLeon-Pennell
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States
| |
Collapse
|
7
|
Chen M, Venturi V, Munier CML. Dissecting the Protective Effect of CD8 + T Cells in Response to SARS-CoV-2 mRNA Vaccination and the Potential Link with Lymph Node CD8 + T Cells. BIOLOGY 2023; 12:1035. [PMID: 37508464 PMCID: PMC10376827 DOI: 10.3390/biology12071035] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/04/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
SARS-CoV-2 vaccines have played a crucial role in effectively reducing COVID-19 disease severity, with a new generation of vaccines that use messenger RNA (mRNA) technology being administered globally. Neutralizing antibodies have featured as the heroes of vaccine-induced immunity. However, vaccine-elicited CD8+ T cells may have a significant impact on the early protective effects of the mRNA vaccine, which are evident 12 days after initial vaccination. Vaccine-induced CD8+ T cells have been shown to respond to multiple epitopes of SARS-CoV-2 and exhibit polyfunctionality in the periphery at the early stage, even when neutralizing antibodies are scarce. Furthermore, SARS-CoV-2 mRNA vaccines induce diverse subsets of memory CD8+ T cells that persist for more than six months following vaccination. However, the protective role of CD8+ T cells in response to the SARS-CoV-2 mRNA vaccines remains a topic of debate. In addition, our understanding of CD8+ T cells in response to vaccination in the lymph nodes, where they first encounter antigen, is still limited. This review delves into the current knowledge regarding the protective role of polyfunctional CD8+ T cells in controlling the virus, the response to SARS-CoV-2 mRNA vaccines, and the contribution to supporting B cell activity and promoting immune protection in the lymph nodes.
Collapse
Affiliation(s)
- Mengfei Chen
- The Kirby Institute, UNSW, Sydney, NSW 2052, Australia
| | | | | |
Collapse
|
8
|
Klein F, Veiga-Villauriz C, Börsch A, Maio S, Palmer S, Dhalla F, Handel AE, Zuklys S, Calvo-Asensio I, Musette L, Deadman ME, White AJ, Lucas B, Anderson G, Holländer GA. Combined multidimensional single-cell protein and RNA profiling dissects the cellular and functional heterogeneity of thymic epithelial cells. Nat Commun 2023; 14:4071. [PMID: 37429879 DOI: 10.1038/s41467-023-39722-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 06/21/2023] [Indexed: 07/12/2023] Open
Abstract
The network of thymic stromal cells provides essential niches with unique molecular cues controlling T cell development and selection. Recent single-cell RNA sequencing studies have uncovered previously unappreciated transcriptional heterogeneity among thymic epithelial cells (TEC). However, there are only very few cell markers that allow a comparable phenotypic identification of TEC. Here, using massively parallel flow cytometry and machine learning, we deconvoluted known TEC phenotypes into novel subpopulations. Using CITEseq, these phenotypes were related to corresponding TEC subtypes defined by the cells' RNA profiles. This approach allowed the phenotypic identification of perinatal cTEC and their physical localisation within the cortical stromal scaffold. In addition, we demonstrate the dynamic change in the frequency of perinatal cTEC in response to developing thymocytes and reveal their exceptional efficiency in positive selection. Collectively, our study identifies markers that allow for an unprecedented dissection of the thymus stromal complexity, as well as physical isolation of TEC populations and assignment of specific functions to individual TEC subtypes.
Collapse
Affiliation(s)
- Fabian Klein
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Clara Veiga-Villauriz
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | | | - Stefano Maio
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Sam Palmer
- Mathematical Institute, University of Oxford, Oxford, UK
| | - Fatima Dhalla
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Adam E Handel
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Saulius Zuklys
- Paediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Irene Calvo-Asensio
- Paediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Lucas Musette
- Paediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Mary E Deadman
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Andrea J White
- Institute for Immunology and Immunotherapy, Medical School, University of Birmingham, Birmingham, UK
| | - Beth Lucas
- Institute for Immunology and Immunotherapy, Medical School, University of Birmingham, Birmingham, UK
| | - Graham Anderson
- Institute for Immunology and Immunotherapy, Medical School, University of Birmingham, Birmingham, UK
| | - Georg A Holländer
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK.
- Paediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland.
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
| |
Collapse
|
9
|
Alkazmi L, Al-Kuraishy HM, Al-Gareeb AI, Alexiou A, Papadakis M, Saad HM, Batiha GES. The potential role of scavenger receptor B type I (SR-BI) in SARS-CoV-2 infection. Immun Inflamm Dis 2023; 11:e786. [PMID: 37102664 PMCID: PMC10103078 DOI: 10.1002/iid3.786] [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: 11/12/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 04/28/2023] Open
Abstract
Scavenger receptor type B I (SR-BI), the major receptor for high-density lipoprotein (HDL) mediates the delivery of cholesterol ester and cholesterol from HDL to the cell membrane. SR-BI is implicated as a receptor for entry of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2). SR-BI is colocalized with the angiotensin-converting enzyme 2 (ACE2) increasing the binding and affinity of SARS-CoV-2 to ACE2 with subsequent viral internalization. SR-BI regulates lymphocyte proliferation and the release of pro-inflammatory cytokines from activated macrophages and lymphocytes. SR-BI is reduced during COVID-19 due to consumption by SARS-CoV-2 infection. COVID-19-associated inflammatory changes and high angiotensin II (AngII) might be possible causes of repression of SR-BI in SARS-CoV-2 infection. In conclusion, the downregulation of SR-BI in COVID-19 could be due to direct invasion by SARS-CoV-2 or through upregulation of pro-inflammatory cytokines, inflammatory signaling pathways, and high circulating AngII. Reduction of SR-BI in COVID-19 look like ACE2 may provoke COVID-19 severity through exaggeration of the immune response. Further studies are invoked to clarify the potential role of SR-BI in the pathogenesis of COVID-19 that could be protective rather than detrimental.
Collapse
Affiliation(s)
- Luay Alkazmi
- Biology Department, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia
- AFNP Med, Wien, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Wuppertal, Germany
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
10
|
Kang M, Park S, Chung Y, Lim JO, Kang JS, Park JH. Hematopoietic Effects of Angelica gigas Nakai Extract on Cyclophosphamide-Induced Myelosuppression. PLANTS (BASEL, SWITZERLAND) 2022; 11:3476. [PMID: 36559587 PMCID: PMC9781469 DOI: 10.3390/plants11243476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
Myelosuppression is a major adverse effect of chemotherapy. With the increasing number of cancer patients worldwide, there is a growing interest in therapeutic approaches that reduce the adverse effects of chemotherapy. Angelica gigas Nakai (AGN) roots have been widely used in oriental medicine to treat blood-related diseases, including cancer. However, the effects of AGN on myelosuppression have not been studied. Here, we investigated the effects of AGN ethanol extract (AGNEX) on cyclophosphamide-induced myelosuppression. AGNEX treatment significantly decreased white blood cell levels while increasing red blood cell and platelet levels in the peripheral blood. It inhibited thymus and spleen atrophy. It also enhanced serum levels of interleukin (IL)-6 and tumor necrosis factor (TNF)-α. qRT-PCR results showed that AGNEX decreased the expression of IL-1b and stem cell factor (SCF) in the bone marrow (BM) while increasing the mRNA expression of IL-3 and IL-6 in the spleen. Although AGNEX did not significantly decrease apoptosis and cell cycle arrest in the BM and splenocytes, AGNEX plays a positive role in cyclophosphamide-induced myelosuppression. AGNEX administration increased BM cells in the femur while decreasing apoptotic BM cells. These findings suggest that AGNEX could be used to treat myelosuppression and as a combination therapy in cancer patients.
Collapse
Affiliation(s)
- Mincheol Kang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si 58245, Republic of Korea
| | - Seojin Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si 58245, Republic of Korea
| | - Yuseong Chung
- Bio and Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Je-Oh Lim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si 58245, Republic of Korea
| | - Jae Seon Kang
- Department of Pharmacy, Kyungsung University, Busan 48434, Republic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si 58245, Republic of Korea
| |
Collapse
|
11
|
The CXCR4-CXCL12 axis promotes T cell reconstitution via efficient hematopoietic immigration. J Genet Genomics 2022; 49:1138-1150. [PMID: 35483564 DOI: 10.1016/j.jgg.2022.04.005] [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: 12/18/2021] [Revised: 04/07/2022] [Accepted: 04/13/2022] [Indexed: 01/20/2023]
Abstract
T cells play a critical role in immunity to protect against pathogens and malignant cells. T cell immunodeficiency is detrimental, especially when T cell perturbation occurs during severe infection, irradiation, chemotherapy, and age-related thymic atrophy. Therefore, strategies that enhance T cell reconstitution provide considerable benefit and warrant intensive investigation. Here, we report the construction of a T cell ablation model in Tg(coro1a:DenNTR) zebrafish via metronidazole administration. The nascent T cells are mainly derived from the hematopoietic cells migrated from the kidney, the functional homolog of bone marrow and the complete recovery time is 6.5 days post-treatment. The cxcr4b gene is upregulated in the responsive hematopoietic cells. Functional interference of CXCR4 via both genetic and chemical manipulations does not greatly affect T lymphopoiesis, but delays T cell regeneration by disrupting hematopoietic migration. In contrast, cxcr4b accelerates the replenishment of hematopoietic cells in the thymus. Consistently, Cxcl12b, a ligand of Cxcr4, is increased in the thymic epithelial cells of the injured animals. Decreased or increased expression of Cxcl12b results in compromised or accelerated T cell recovery, respectively, similar to those observed with Cxcr4b. Taken together, our study reveals a role of CXCR4-CXCL12 signaling in promoting T cell recovery and provides a promising target for the treatment of immunodeficiency due to T cell injury.
Collapse
|
12
|
Schutt SD, Wu Y, Kharel A, Bastian D, Choi HJ, Hanief Sofi M, Mealer C, McDaniel Mims B, Nguyen H, Liu C, Helke K, Cui W, Zhang X, Ben-David Y, Yu XZ. The druggable transcription factor Fli-1 regulates T cell immunity and tolerance in graft-versus-host disease. J Clin Invest 2022; 132:143950. [PMID: 36074578 PMCID: PMC9621143 DOI: 10.1172/jci143950] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Graft-versus-host disease (GVHD), manifesting as either acute (aGVHD) or chronic (cGVHD), presents significant life-threatening complications following allogeneic hematopoietic cell transplantation. Here, we investigated Friend virus leukemia integration 1 (Fli-1) in GVHD pathogenesis and validated Fli-1 as a therapeutic target. Using genetic approaches, we found that Fli-1 dynamically regulated different T cell subsets in allogeneic responses and pathogenicity in the development of aGVHD and cGVHD. Compared with homozygous Fli1-deficient or WT T cells, heterozygous Fli1-deficient T cells induced the mildest GVHD, as evidenced by the lowest Th1 and Th17 cell differentiation. Single-cell RNA-Seq analysis revealed that Fli-1 differentially regulated CD4+ and CD8+ T cell responses. Fli-1 promoted the transcription of Th1/Th17 pathways and T cell receptor-inducible (TCR-inducible) transcription factors in CD4+ T cells, while suppressing activation- and function-related gene pathways in CD8+ T cells. Importantly, a low dose of camptothecin, topotecan, or etoposide acted as a potent Fli-1 inhibitor and significantly attenuated GVHD severity, while preserving the graft-versus-leukemia (GVL) effect. This observation was extended to a xenograft model, in which GVHD was induced by human T cells. In conclusion, we provide evidence that Fli-1 plays a crucial role in alloreactive CD4+ T cell activation and differentiation and that targeting Fli-1 may be an attractive strategy for treating GVHD without compromising the GVL effect.
Collapse
Affiliation(s)
- Steven D. Schutt
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, South Carolina, USA
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, South Carolina, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin (MCW), Milwaukee, Wisconsin, USA
| | - Arjun Kharel
- Department of Microbiology and Immunology, Medical College of Wisconsin (MCW), Milwaukee, Wisconsin, USA
| | - David Bastian
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, South Carolina, USA
| | - Hee-Jin Choi
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, South Carolina, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin (MCW), Milwaukee, Wisconsin, USA
| | - Mohammed Hanief Sofi
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, South Carolina, USA
| | - Corey Mealer
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, South Carolina, USA
| | - Brianyell McDaniel Mims
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, South Carolina, USA
| | - Hung Nguyen
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, South Carolina, USA
| | - Chen Liu
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Weiguo Cui
- Department of Microbiology and Immunology, Medical College of Wisconsin (MCW), Milwaukee, Wisconsin, USA
| | - Xian Zhang
- Department of Medicine at MUSC, Charleston, South Carolina, USA
| | - Yaacov Ben-David
- Guizhou Medical University and the Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang, China
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina (MUSC), Charleston, South Carolina, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin (MCW), Milwaukee, Wisconsin, USA.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA.,The Cancer Center in MCW, Milwaukee, Wisconsin, USA
| |
Collapse
|
13
|
Ren B, Xia H, Liao Y, Zhou H, Wang Z, Shi Y, Zhu M. Endothelial SIRPα signaling controls VE-cadherin endocytosis for thymic homing of progenitor cells. eLife 2022; 11:69219. [PMID: 35511221 PMCID: PMC9071265 DOI: 10.7554/elife.69219] [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: 04/08/2021] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Thymic homing of hematopoietic progenitor cells (HPCs) is tightly regulated for proper T cell development. Previously we have identified a subset of specialized thymic portal endothelial cells (TPECs), which is important for thymic HPC homing. However, the underlying molecular mechanism still remains unknown. Here, we found that signal regulatory protein alpha (SIRPα) is preferentially expressed on TPECs. Disruption of CD47-SIRPα signaling in mice resulted in reduced number of thymic early T cell progenitors (ETPs), impaired thymic HPC homing, and altered early development of thymocytes. Mechanistically, Sirpa-deficient ECs and Cd47-deficient bone marrow progenitor cells or T lymphocytes demonstrated impaired transendothelial migration (TEM). Specifically, SIRPα intracellular ITIM motif-initiated downstream signaling in ECs was found to be required for TEM in an SHP2- and Src-dependent manner. Furthermore, CD47 signaling from migrating cells and SIRPα intracellular signaling were found to be required for VE-cadherin endocytosis in ECs. Thus, our study reveals a novel role of endothelial SIRPα signaling for thymic HPC homing for T cell development.
Collapse
Affiliation(s)
- Boyang Ren
- The Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Huan Xia
- The Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Yijun Liao
- The Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Hang Zhou
- The Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Zhongnan Wang
- The Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yaoyao Shi
- The Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Mingzhao Zhu
- The Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Garcia P, Wang Y, Viallet J, Macek Jilkova Z. The Chicken Embryo Model: A Novel and Relevant Model for Immune-Based Studies. Front Immunol 2021; 12:791081. [PMID: 34868080 PMCID: PMC8640176 DOI: 10.3389/fimmu.2021.791081] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of the immune system is associated with many pathologies, including cardiovascular diseases, diabetes, and cancer. To date, the most commonly used models in biomedical research are rodents, and despite the various advantages they offer, their use also raises numerous drawbacks. Recently, another in vivo model, the chicken embryo and its chorioallantoic membrane, has re-emerged for various applications. This model has many benefits compared to other classical models, as it is cost-effective, time-efficient, and easier to use. In this review, we explain how the chicken embryo can be used as a model for immune-based studies, as it gradually develops an embryonic immune system, yet which is functionally similar to humans'. We mainly aim to describe the avian immune system, highlighting the differences and similarities with the human immune system, including the repertoire of lymphoid tissues, immune cells, and other key features. We also describe the general in ovo immune ontogeny. In conclusion, we expect that this review will help future studies better tailor their use of the chicken embryo model for testing specific experimental hypotheses or performing preclinical testing.
Collapse
Affiliation(s)
- Paul Garcia
- Université Grenoble Alpes, Grenoble, France
- R&D Department, Inovotion, La Tronche, France
- Institute for Advanced Biosciences, Research Center Université Grenoble Alpes (UGA)/Inserm U 1209/CNRS 5309, La Tronche, France
| | - Yan Wang
- R&D Department, Inovotion, La Tronche, France
| | | | - Zuzana Macek Jilkova
- Université Grenoble Alpes, Grenoble, France
- Institute for Advanced Biosciences, Research Center Université Grenoble Alpes (UGA)/Inserm U 1209/CNRS 5309, La Tronche, France
- Service d’Hépato-Gastroentérologie, Pôle Digidune, Centre Hospitalo-Universitaire (USA) Grenoble Alpes, La Tronche, France
| |
Collapse
|
15
|
The sialyltransferase ST3Gal-IV guides murine T-cell progenitors to the thymus. Blood Adv 2021; 4:1930-1941. [PMID: 32380539 DOI: 10.1182/bloodadvances.2019001046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 04/12/2020] [Indexed: 12/15/2022] Open
Abstract
T lymphocytes are important players in beneficial and detrimental immune responses. In contrast to other lymphocyte populations that develop in the bone marrow, T-cell precursors need to migrate to the thymus for further development. The interaction of P-selectin and P-selectin glycoprotein ligand-1 (PSGL-1) is crucial for thymic entry of T-cell precursors during settings of T-cell lineage reconstitution. PSGL-1 has to be sialylated to function as a ligand for P-selectin, and the sialyltransferase ST3Gal-IV might play a critical role in this process. We therefore investigated the role of ST3Gal-IV for T-cell development using competitive mixed bone marrow chimeric mice. We found that ST3Gal-IV is dispensable for homing and engraftment of hematopoietic precursors in the bone marrow. However, ST3Gal-IV deficiency affects seeding of the thymus by early T-cell progenitors, leading to impaired restoration of the peripheral T-cell compartment. This defect could be restored by ectopic retroviral expression of ST3Gal-IV in hematopoietic stem cells derived from ST3Gal-IV-deficient donor mice. Our findings show that ST3Gal-IV plays a critical and nonredundant role for efficient T-cell lineage reconstitution after bone marrow transplantation.
Collapse
|
16
|
Kumar S, Singh SK, Rana B, Rana A. The regulatory function of mixed lineage kinase 3 in tumor and host immunity. Pharmacol Ther 2021; 219:107704. [PMID: 33045253 PMCID: PMC7887016 DOI: 10.1016/j.pharmthera.2020.107704] [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: 08/03/2020] [Accepted: 10/02/2020] [Indexed: 12/26/2022]
Abstract
Protein kinases are the second most sought-after G-protein coupled receptors as drug targets because of their overexpression, mutations, and dysregulated catalytic activities in various pathological conditions. Till 2019, 48 protein kinase inhibitors have received FDA approval for the treatment of multiple illnesses, of which the majority of them are indicated for different malignancies. One of the attractive sub-group of protein kinases that has attracted attention for drug development is the family members of MAPKs that are recognized to play significant roles in different cancers. Several inhibitors have been developed against various MAPK members; however, none of them as monotherapy has shown sustainable efficacy. One of the MAPK members, called Mixed Lineage Kinase 3 (MLK3), has attracted considerable attention due to its role in inflammation and neurodegenerative diseases; however, its role in cancer is an emerging area that needs more investigation. Recent advances have shown that MLK3 plays a role in cancer cell survival, migration, drug resistance, cell death, and tumor immunity. This review describes how MLK3 regulates different MAPK pathways, cancer cell growth and survival, apoptosis, and host's immunity. We also discuss how MLK3 inhibitors can potentially be used along with immunotherapy for different malignancies.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA.
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA
| | - Basabi Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, IL 60612, USA; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA; Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
17
|
Tolstykh EI, Vozilova AV, Degteva MO, Akleyev AV. Concept of T-Cell Genus as a Basis for Analysis of the Results of Cytogenetic Studies after Local Bone Marrow Exposure. BIOL BULL+ 2021. [DOI: 10.1134/s1062359020110151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Deng Y, Chen H, Zeng Y, Wang K, Zhang H, Hu H. Leaving no one behind: tracing every human thymocyte by single-cell RNA-sequencing. Semin Immunopathol 2021; 43:29-43. [PMID: 33449155 DOI: 10.1007/s00281-020-00834-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 12/22/2020] [Indexed: 02/05/2023]
Abstract
The thymus is the primary organ for T-cell development, providing an essential microenvironment consisting of the appropriate cytokine milieu and specialized stromal cells. Thymus-seeding progenitors from circulation immigrate into the thymus and undergo the stepwise T-cell specification, commitment, and selection processes. The transcriptional factors, epigenetic regulators, and signaling pathways involved in the T-cell development have been intensively studied using mouse models. Despite our growing knowledge of T-cell development, major questions remain unanswered regarding the ontogeny and early events of T-cell development at the fetal stage, especially in humans. The recently developed single-cell RNA-sequencing technique provides an ideal tool to investigate the heterogeneity of T-cell precursors and the molecular mechanisms underlying the divergent fates of certain T-cell precursors at the single-cell level. In this review, we aim to summarize the current progress of the study on human thymus organogenesis and thymocyte and thymic epithelial cell development, which is to shed new lights on developing novel strategies for in vitro T-cell regeneration and thymus rejuvenation.
Collapse
Affiliation(s)
- Yujun Deng
- Department of Rheumatology and Immunology and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Hong Chen
- Department of Rheumatology and Immunology and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yang Zeng
- State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.,State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, 100071, China
| | - Keyue Wang
- Department of Rheumatology and Immunology and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huiyuan Zhang
- Department of Rheumatology and Immunology and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| | - Hongbo Hu
- Department of Rheumatology and Immunology and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| |
Collapse
|
19
|
Liu C, Lan Y, Liu B, Zhang H, Hu H. T Cell Development: Old Tales Retold By Single-Cell RNA Sequencing. Trends Immunol 2021; 42:165-175. [PMID: 33446417 DOI: 10.1016/j.it.2020.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Mammalian T cell development initiates from the migration of hematopoietic progenitors to the thymus, which undergo cell proliferation, T-lineage specification and commitment, as well as positive and negative selection. These processes are precisely controlled at multiple levels and have been intensively studied using gene-modified animal models and in vitro coculture systems. However, several long-standing questions, including the characterization of the rare but crucial progenitors/precursors and the molecular mechanisms underlying their fate decision, have been dampened because of cell scarcity and lack of appropriate techniques. Single-cell RNA sequencing (scRNA-seq) makes it possible to investigate and resolve some of these questions, leading to new remarkable progress in identifying and characterizing early thymic progenitors and delineating the refined developmental trajectories of conventional and unconventional T cells.
Collapse
Affiliation(s)
- Chen Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100071, China
| | - Yu Lan
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100071, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China; State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Huiyuan Zhang
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Hongbo Hu
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
20
|
Olariu V, Yui MA, Krupinski P, Zhou W, Deichmann J, Andersson E, Rothenberg EV, Peterson C. Multi-scale Dynamical Modeling of T Cell Development from an Early Thymic Progenitor State to Lineage Commitment. Cell Rep 2021; 34:108622. [PMID: 33440162 DOI: 10.1016/j.celrep.2020.108622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 04/24/2020] [Accepted: 12/18/2020] [Indexed: 01/13/2023] Open
Abstract
Intrathymic development of committed progenitor (pro)-T cells from multipotent hematopoietic precursors offers an opportunity to dissect the molecular circuitry establishing cell identity in response to environmental signals. This transition encompasses programmed shutoff of stem/progenitor genes, upregulation of T cell specification genes, proliferation, and ultimately commitment. To explain these features in light of reported cis-acting chromatin effects and experimental kinetic data, we develop a three-level dynamic model of commitment based upon regulation of the commitment-linked gene Bcl11b. The levels are (1) a core gene regulatory network (GRN) architecture from transcription factor (TF) perturbation data, (2) a stochastically controlled chromatin-state gate, and (3) a single-cell proliferation model validated by experimental clonal growth and commitment kinetic assays. Using RNA fluorescence in situ hybridization (FISH) measurements of genes encoding key TFs and measured bulk population dynamics, this single-cell model predicts state-switching kinetics validated by measured clonal proliferation and commitment times. The resulting multi-scale model provides a mechanistic framework for dissecting commitment dynamics.
Collapse
Affiliation(s)
- Victor Olariu
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden
| | - Mary A Yui
- Division of Biology and Biological Engineering, 156-29, California Institute of Technology, Pasadena, CA 91125, USA
| | - Pawel Krupinski
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden
| | - Wen Zhou
- Division of Biology and Biological Engineering, 156-29, California Institute of Technology, Pasadena, CA 91125, USA
| | - Julia Deichmann
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden
| | - Emil Andersson
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering, 156-29, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Carsten Peterson
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden.
| |
Collapse
|
21
|
Koeniger T, Bell L, Mifka A, Enders M, Hautmann V, Mekala SR, Kirchner P, Ekici AB, Schulz C, Wörsdörfer P, Mencl S, Kleinschnitz C, Ergün S, Kuerten S. Bone marrow-derived myeloid progenitors in the leptomeninges of adult mice. STEM CELLS (DAYTON, OHIO) 2020; 39:227-239. [PMID: 33270951 DOI: 10.1002/stem.3311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/29/2020] [Accepted: 11/17/2020] [Indexed: 11/11/2022]
Abstract
Although the bone marrow contains most hematopoietic activity during adulthood, hematopoietic stem and progenitor cells can be recovered from various extramedullary sites. Cells with hematopoietic progenitor properties have even been reported in the adult brain under steady-state conditions, but their nature and localization remain insufficiently defined. Here, we describe a heterogeneous population of myeloid progenitors in the leptomeninges of adult C57BL/6 mice. This cell pool included common myeloid, granulocyte/macrophage, and megakaryocyte/erythrocyte progenitors. Accordingly, it gave rise to all major myelo-erythroid lineages in clonogenic culture assays. Brain-associated progenitors persisted after tissue perfusion and were partially inaccessible to intravenous antibodies, suggesting their localization behind continuous blood vessel endothelium such as the blood-arachnoid barrier. Flt3Cre lineage tracing and bone marrow transplantation showed that the precursors were derived from adult hematopoietic stem cells and were most likely continuously replaced via cell trafficking. Importantly, their occurrence was tied to the immunologic state of the central nervous system (CNS) and was diminished in the context of neuroinflammation and ischemic stroke. Our findings confirm the presence of myeloid progenitors at the meningeal border of the brain and lay the foundation to unravel their possible functions in CNS surveillance and local immune cell production.
Collapse
Affiliation(s)
- Tobias Koeniger
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Luisa Bell
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Anika Mifka
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Enders
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Valentin Hautmann
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Subba Rao Mekala
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Philipp Kirchner
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Ludwig Maximilian University of Munich, Munich, Germany
| | - Philipp Wörsdörfer
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Stine Mencl
- University Hospital Essen, Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Christoph Kleinschnitz
- University Hospital Essen, Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany.,Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Anatomisches Institut, Neuroanatomie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
22
|
Fadini GP, Mehta A, Dhindsa DS, Bonora BM, Sreejit G, Nagareddy P, Quyyumi AA. Circulating stem cells and cardiovascular outcomes: from basic science to the clinic. Eur Heart J 2020; 41:4271-4282. [PMID: 31891403 PMCID: PMC7825095 DOI: 10.1093/eurheartj/ehz923] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/19/2019] [Accepted: 12/05/2019] [Indexed: 02/06/2023] Open
Abstract
The cardiovascular and haematopoietic systems have fundamental inter-relationships during development, as well as in health and disease of the adult organism. Although haematopoietic stem cells (HSCs) emerge from a specialized haemogenic endothelium in the embryo, persistence of haemangioblasts in adulthood is debated. Rather, the vast majority of circulating stem cells (CSCs) is composed of bone marrow-derived HSCs and the downstream haematopoietic stem/progenitors (HSPCs). A fraction of these cells, known as endothelial progenitor cells (EPCs), has endothelial specification and vascular tropism. In general, the levels of HSCs, HSPCs, and EPCs are considered indicative of the endogenous regenerative capacity of the organism as a whole and, particularly, of the cardiovascular system. In the last two decades, the research on CSCs has focused on their physiologic role in tissue/organ homoeostasis, their potential application in cell therapies, and their use as clinical biomarkers. In this review, we provide background information on the biology of CSCs and discuss in detail the clinical implications of changing CSC levels in patients with cardiovascular risk factors or established cardiovascular disease. Of particular interest is the mounting evidence available in the literature on the close relationships between reduced levels of CSCs and adverse cardiovascular outcomes in different cohorts of patients. We also discuss potential mechanisms that explain this association. Beyond CSCs' ability to participate in cardiovascular repair, levels of CSCs need to be interpreted in the context of the broader connections between haematopoiesis and cardiovascular function, including the role of clonal haematopoiesis and inflammatory myelopoiesis.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Anurag Mehta
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| | - Devinder Singh Dhindsa
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| | | | - Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Prabhakara Nagareddy
- Division of Cardiac Surgery, Department of Surgery, Ohio State University, Columbus, OH 43210, USA
| | - Arshed Ali Quyyumi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, 201 Dowman Drive, Atlanta, GA 30322, USA
| |
Collapse
|
23
|
Wu T, Plett PA, Chua HL, Jacobsen M, Sandusky GE, MacVittie TJ, Orschell CM. Immune Reconstitution and Thymic Involution in the Acute and Delayed Hematopoietic Radiation Syndromes. HEALTH PHYSICS 2020; 119:647-658. [PMID: 32947490 PMCID: PMC7541734 DOI: 10.1097/hp.0000000000001352] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Lymphoid lineage recovery and involution after exposure to potentially lethal doses of ionizing radiation have not been well defined, especially the long-term effects in aged survivors and with regard to male/female differences. To examine these questions, male and female C57BL/6 mice were exposed to lethal radiation at 12 wk of age in a model of the Hematopoietic-Acute Radiation Syndrome, and bone marrow, thymus, spleen, and peripheral blood examined up to 24 mo of age for the lymphopoietic delayed effects of acute radiation exposure. Aged mice showed myeloid skewing and incomplete lymphocyte recovery in all lymphoid tissues. Spleen and peripheral blood both exhibited a monophasic recovery pattern, while thymus demonstrated a biphasic pattern. Naïve T cells in blood and spleen and all subsets of thymocytes were decreased in aged irradiated mice compared to age-matched non-irradiated controls. Of interest, irradiated males experienced significantly improved reconstitution of thymocyte subsets and peripheral blood elements compared to females. Bone marrow from aged irradiated survivors was significantly deficient in the primitive lymphoid-primed multipotent progenitors and common lymphoid progenitors, which were only 8-10% of levels in aged-matched non-irradiated controls. Taken together, these analyses define significant age- and sex-related deficiencies at all levels of lymphopoiesis throughout the lifespan of survivors of the Hematopoietic-Acute Radiation Syndrome and may provide a murine model suitable for assessing the efficacy of potential medical countermeasures and therapeutic strategies to alleviate the severe immune suppression that occurs after radiation exposure.
Collapse
Affiliation(s)
- Tong Wu
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - P. Artur Plett
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Hui Lin Chua
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Max Jacobsen
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - George E. Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Thomas J. MacVittie
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
| | | |
Collapse
|
24
|
Bradshaw AD, DeLeon-Pennell KY. T-cell regulation of fibroblasts and cardiac fibrosis. Matrix Biol 2020; 91-92:167-175. [PMID: 32438054 PMCID: PMC7434661 DOI: 10.1016/j.matbio.2020.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/30/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
Inflammation contributes to the development of heart failure (HF) through multiple mechanisms including regulating extracellular matrix (ECM) degradation and deposition. Interactions between cells in the myocardium orchestrates the magnitude and duration of inflammatory cell recruitment and ECM remodeling events associated with HF. More recently, studies have shown T-cells have signficant roles in post-MI wound healing. T-cell biology in HF illustrates the complexity of cross-talk between inflammatory cell types and resident fibroblasts. This review will focus on T-cell recruitment to the myocardium and T-cell specific factors that might influence cardiac wound healing and fibrosis in the heart with consideration of age and sex as important factors in T-cell activity.
Collapse
Affiliation(s)
- Amy D Bradshaw
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee Street Charleston, SC 29401, United States
| | - Kristine Y DeLeon-Pennell
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center, 109 Bee Street Charleston, SC 29401, United States.
| |
Collapse
|
25
|
|
26
|
Enciso J, Pelayo R, Villarreal C. From Discrete to Continuous Modeling of Lymphocyte Development and Plasticity in Chronic Diseases. Front Immunol 2019; 10:1927. [PMID: 31481957 PMCID: PMC6710364 DOI: 10.3389/fimmu.2019.01927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 07/30/2019] [Indexed: 12/12/2022] Open
Abstract
The molecular events leading to differentiation, development, and plasticity of lymphoid cells have been subject of intense research due to their key roles in multiple pathologies, such as lymphoproliferative disorders, tumor growth maintenance and chronic diseases. The emergent roles of lymphoid cells and the use of high-throughput technologies have led to an extensive accumulation of experimental data allowing the reconstruction of gene regulatory networks (GRN) by integrating biochemical signals provided by the microenvironment with transcriptional modules of lineage-specific genes. Computational modeling of GRN has been useful for the identification of molecular switches involved in lymphoid specification, prediction of microenvironment-dependent cell plasticity, and analyses of signaling events occurring downstream the activation of antigen recognition receptors. Among most common modeling strategies to analyze the dynamical behavior of GRN, discrete dynamic models are widely used for their capacity to capture molecular interactions when a limited knowledge of kinetic parameters is present. However, they are less powerful when modeling complex systems sensitive to biochemical gradients. To compensate it, discrete models may be transformed into regulatory networks that includes state variables and parameters varying within a continuous range. This approach is based on a system of differential equations dynamics with regulatory interactions described by fuzzy logic propositions. Here, we discuss the applicability of this method on modeling of development and plasticity processes of adaptive lymphocytes, and its potential implications in the study of pathological landscapes associated to chronic diseases.
Collapse
Affiliation(s)
- Jennifer Enciso
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rosana Pelayo
- Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Carlos Villarreal
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departamento de Física Cuántica y Fotónica, Instituto de Física, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
27
|
Zheng Z, Ai J, Guo L, Ye X, Bondada S, Howatt D, Daugherty A, Li XA. SR-BI (Scavenger Receptor Class B Type 1) Is Critical in Maintaining Normal T-Cell Development and Enhancing Thymic Regeneration. Arterioscler Thromb Vasc Biol 2019; 38:2706-2717. [PMID: 30354229 DOI: 10.1161/atvbaha.118.311728] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- Continuous T-cell production from thymus is essential in replenishing naïve T-cell pool and maintaining optimal T-cell functions. However, the underlying mechanisms regulating the T-cell development in thymus remains largely unknown. Approach and Results- We identified SR-BI (scavenger receptor class B type 1), an HDL (high-density lipoprotein) receptor, as a novel modulator in T-cell development. We found that SR-BI deficiency in mice led to reduced thymus size and decreased T-cell production, which was accompanied by narrowed peripheral naïve T-cell pool. Further investigation revealed that SR-BI deficiency impaired progenitor thymic homing, causing a dramatic reduction in the percentage of earliest thymic progenitors, but did not affect other downstream T-cell developmental steps inside the thymus. As a result of the impaired progenitor thymic homing, SR-BI-deficient mice displayed delayed thymic regeneration postirradiation. Using a variety of experimental approaches, we revealed that the impaired T-cell development in SR-BI-deficient mice was not caused by hematopoietic SR-BI deficiency or SR-BI deficiency-induced hypercholesterolemia, but mainly attributed to the SR-BI deficiency in adrenal glands, as adrenal-specific SR-BI-deficient mice exhibited similar defects in T-cell development and thymic regeneration with SR-BI-deficient mice. Conclusions- This study demonstrates that SR-BI deficiency impaired T-cell development and delayed thymic regeneration by affecting progenitor thymic homing in mice, elucidating a previously unrecognized link between SR-BI and adaptive immunity.
Collapse
Affiliation(s)
- Zhong Zheng
- From the Department of Pharmacology and Nutritional Sciences (Z.Z., J.A., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Junting Ai
- From the Department of Pharmacology and Nutritional Sciences (Z.Z., J.A., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Ling Guo
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Xiang Ye
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Subbarao Bondada
- Department of Microbiology (S.B.), University of Kentucky College of Medicine, Lexington
| | - Deborah Howatt
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Alan Daugherty
- Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Department of Physiology (A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| | - Xiang-An Li
- From the Department of Pharmacology and Nutritional Sciences (Z.Z., J.A., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Saha Cardiovascular Research Center (Z.Z., J.A., L.G., X.Y., D.H., A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington.,Department of Physiology (A.D., X.-A.L.), University of Kentucky College of Medicine, Lexington
| |
Collapse
|
28
|
Effect of early pregnancy on the expression of prostaglandin synthases in the ovine thymus. Theriogenology 2019; 136:166-171. [PMID: 31265945 DOI: 10.1016/j.theriogenology.2019.06.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/09/2019] [Accepted: 06/24/2019] [Indexed: 11/21/2022]
Abstract
Thymus is a primary lymphoid organ, must adapt to the presence of fetal alloantigens. Prostaglandins (PGs) have diverse effects to activate or inhibit the immune response, but effects of early pregnancy on the expression of PG synthases in ovine maternal thymus are unclear. In this study, ovine thymic samples were obtained at day 16 of the estrous cycle, and days 13, 16 and 25 of pregnancy. The expression of PG synthases, including cyclooxygenase 1 (COX-1), COX-2, PGE2 synthase (PTGES), and a prostaglandin F2α synthase (Aldo-keto reductase family 1, member B1, AKR1B1), was evaluated using quantitative real-time PCR, Western blot and immunohistochemistry analysis. In addition, the thymus/body ratio was also calculated. Our results showed that the expression of COX-2 mRNA and protein, AKR1B1 mRNA and dimer were up-regulated on day 25 of pregnancy (P < 0.05), and expression of COX-1, PTGES mRNA and protein, AKR1B1 monomer and thymus/body ratio were similar at different stages of pregnancy and the estrous cycle. The immunohistochemistry results showed that the COX-2 and AKR1B1 proteins were located in the stromal cells, capillaries and thymic corpuscles. This is the first study to report that expression of COX-2 and AKR1B1 dimer is up-regulated in the maternal thymus during early pregnancy, suggesting that early pregnancy exerts its effects on maternal thymus, which is involved in immunomodulation during early pregnancy in sheep.
Collapse
|
29
|
Milic J, Tian Y, Bernhagen J. Role of the COP9 Signalosome (CSN) in Cardiovascular Diseases. Biomolecules 2019; 9:biom9060217. [PMID: 31195722 PMCID: PMC6628250 DOI: 10.3390/biom9060217] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/02/2019] [Accepted: 06/03/2019] [Indexed: 12/19/2022] Open
Abstract
The constitutive photomorphogenesis 9 (COP9) signalosome (CSN) is an evolutionarily conserved multi-protein complex, consisting of eight subunits termed CSN1-CSN8. The main biochemical function of the CSN is the control of protein degradation via the ubiquitin-proteasome-system through regulation of cullin-RING E3-ligase (CRL) activity by deNEDDylation of cullins, but the CSN also serves as a docking platform for signaling proteins. The catalytic deNEDDylase (isopeptidase) activity of the complex is executed by CSN5, but only efficiently occurs in the three-dimensional architectural context of the complex. Due to its positioning in a central cellular pathway connected to cell responses such as cell-cycle, proliferation, and signaling, the CSN has been implicated in several human diseases, with most evidence available for a role in cancer. However, emerging evidence also suggests that the CSN is involved in inflammation and cardiovascular diseases. This is both due to its role in controlling CRLs, regulating components of key inflammatory pathways such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and complex-independent interactions of subunits such as CSN5 with inflammatory proteins. In this case, we summarize and discuss studies suggesting that the CSN may have a key role in cardiovascular diseases such as atherosclerosis and heart failure. We discuss the implicated molecular mechanisms ranging from inflammatory NF-κB signaling to proteotoxicity and necrosis, covering disease-relevant cell types such as myeloid and endothelial cells or cardiomyocytes. While the CSN is considered to be disease-exacerbating in most cancer entities, the cardiovascular studies suggest potent protective activities in the vasculature and heart. The underlying mechanisms and potential therapeutic avenues will be critically discussed.
Collapse
Affiliation(s)
- Jelena Milic
- Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München (KUM), Ludwig-Maximilians-University (LMU), 81377 Munich, Germany.
| | - Yuan Tian
- Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München (KUM), Ludwig-Maximilians-University (LMU), 81377 Munich, Germany.
| | - Jürgen Bernhagen
- Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München (KUM), Ludwig-Maximilians-University (LMU), 81377 Munich, Germany.
- Munich Heart Alliance, 80802 Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany.
| |
Collapse
|
30
|
White AJ, Lucas B, Jenkinson WE, Anderson G. Invariant NKT Cells and Control of the Thymus Medulla. THE JOURNAL OF IMMUNOLOGY 2019; 200:3333-3339. [PMID: 29735644 DOI: 10.4049/jimmunol.1800120] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/20/2018] [Indexed: 12/29/2022]
Abstract
Most αβ T cells that form in the thymus are generated during mainstream conventional thymocyte development and involve the generation and selection of a diverse αβ TCR repertoire that recognizes self-peptide/MHC complexes. Additionally, the thymus also supports the production of T cell subsets that express αβ TCRs but display unique developmental and functional features distinct from conventional αβ T cells. These include multiple lineages of CD1d-restricted invariant NKT (iNKT) cells that express an invariant αβ TCR, branch off from mainstream thymocytes at the CD4+CD8+ stage, and are potent producers of polarizing cytokines. Importantly, and despite their differences, iNKT cells and conventional αβ T cells share common requirements for thymic epithelial microenvironments during their development. Moreover, emerging evidence suggests that constitutive cytokine production by iNKT cells influences both conventional thymocyte development and the intrathymic formation of additional innate CD8+ αβ T cells with memory-like properties. In this article, we review evidence for an intrathymic innate lymphocyte network in which iNKT cells play key roles in multiple aspects of thymus function.
Collapse
Affiliation(s)
- Andrea J White
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Beth Lucas
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - William E Jenkinson
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Graham Anderson
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
31
|
Bouderlique T, Peña-Pérez L, Kharazi S, Hils M, Li X, Krstic A, De Paepe A, Schachtrup C, Gustafsson C, Holmberg D, Schachtrup K, Månsson R. The Concerted Action of E2-2 and HEB Is Critical for Early Lymphoid Specification. Front Immunol 2019; 10:455. [PMID: 30936870 PMCID: PMC6433000 DOI: 10.3389/fimmu.2019.00455] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/20/2019] [Indexed: 12/21/2022] Open
Abstract
The apparition of adaptive immunity in Gnathostomata correlates with the expansion of the E-protein family to encompass E2-2, HEB, and E2A. Within the family, E2-2 and HEB are more closely evolutionarily related but their concerted action in hematopoiesis remains to be explored. Here we show that the combined disruption of E2-2 and HEB results in failure to express the early lymphoid program in Common lymphoid precursors (CLPs) and a near complete block in B-cell development. In the thymus, Early T-cell progenitors (ETPs) were reduced and T-cell development perturbed, resulting in reduced CD4 T- and increased γδ T-cell numbers. In contrast, hematopoietic stem cells (HSCs), erythro-myeloid progenitors, and innate immune cells were unaffected showing that E2-2 and HEB are dispensable for the ancestral hematopoietic lineages. Taken together, this E-protein dependence suggests that the appearance of the full Gnathostomata E-protein repertoire was critical to reinforce the gene regulatory circuits that drove the emergence and expansion of the lineages constituting humoral immunity.
Collapse
Affiliation(s)
- Thibault Bouderlique
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lucia Peña-Pérez
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shabnam Kharazi
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Miriam Hils
- Faculty of Medicine & Faculty of Biology, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Freiburg, Germany
| | - Xiaoze Li
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Aleksandra Krstic
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ayla De Paepe
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Christian Schachtrup
- Faculty of Medicine, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Charlotte Gustafsson
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Dan Holmberg
- Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - Kristina Schachtrup
- Faculty of Medicine & Faculty of Biology, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Freiburg, Germany
| | - Robert Månsson
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden.,Hematology Center, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
32
|
Klein F, Mitrovic M, Roux J, Engdahl C, von Muenchow L, Alberti-Servera L, Fehling HJ, Pelczar P, Rolink A, Tsapogas P. The transcription factor Duxbl mediates elimination of pre-T cells that fail β-selection. J Exp Med 2019; 216:638-655. [PMID: 30765463 PMCID: PMC6400535 DOI: 10.1084/jem.20181444] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/13/2018] [Accepted: 01/22/2019] [Indexed: 12/13/2022] Open
Abstract
During β-selection, T cells without productive TCRβ rearrangements are eliminated. Klein et al. show that the transcription factor Duxbl regulates this process by inducing apoptosis through activation of the Oas/RNaseL pathway. Successful TCRβ rearrangement rescues cells by pre-TCR–mediated Duxbl suppression. T cell development is critically dependent on successful rearrangement of antigen-receptor chains. At the β-selection checkpoint, only cells with a functional rearrangement continue in development. However, how nonselected T cells proceed in their dead-end fate is not clear. We identified low CD27 expression to mark pre-T cells that have failed to rearrange their β-chain. Expression profiling and single-cell transcriptome clustering identified a developmental trajectory through β-selection and revealed specific expression of the transcription factor Duxbl at a stage of high recombination activity before β-selection. Conditional transgenic expression of Duxbl resulted in a developmental block at the DN3-to-DN4 transition due to reduced proliferation and enhanced apoptosis, whereas RNA silencing of Duxbl led to a decrease in apoptosis. Transcriptome analysis linked Duxbl to elevated expression of the apoptosis-inducing Oas/RNaseL pathway. RNaseL deficiency or sustained Bcl2 expression led to a partial rescue of cells in Duxbl transgenic mice. These findings identify Duxbl as a regulator of β-selection by inducing apoptosis in cells with a nonfunctional rearrangement.
Collapse
Affiliation(s)
- Fabian Klein
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Mladen Mitrovic
- Immune Regulation, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Julien Roux
- Bioinformatics Core Facility, Department of Biomedicine, University of Basel, Basel, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Corinne Engdahl
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Lilly von Muenchow
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Llucia Alberti-Servera
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel, Switzerland
| | - Antonius Rolink
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Panagiotis Tsapogas
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
33
|
Pabst R. The thymus is relevant in the migration of mature lymphocytes. Cell Tissue Res 2019; 376:19-24. [DOI: 10.1007/s00441-019-02994-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/15/2019] [Indexed: 12/11/2022]
|
34
|
Fu J, Zuber J, Martinez M, Shonts B, Obradovic A, Wang H, Lau SP, Xia A, Waffarn EE, Frangaj K, Savage TM, Simpson MT, Yang S, Guo XV, Miron M, Senda T, Rogers K, Rahman A, Ho SH, Shen Y, Griesemer A, Farber DL, Kato T, Sykes M. Human Intestinal Allografts Contain Functional Hematopoietic Stem and Progenitor Cells that Are Maintained by a Circulating Pool. Cell Stem Cell 2019; 24:227-239.e8. [PMID: 30503142 PMCID: PMC6398344 DOI: 10.1016/j.stem.2018.11.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/20/2018] [Accepted: 11/02/2018] [Indexed: 01/09/2023]
Abstract
Human intestinal transplantation often results in long-term mixed chimerism of donor and recipient blood in transplant patients. We followed the phenotypes of chimeric peripheral blood cells in 21 patients receiving intestinal allografts over 5 years. Donor lymphocyte phenotypes suggested a contribution of hematopoietic stem and progenitor cells (HSPCs) from the graft. Surprisingly, we detected donor-derived HSPCs in intestinal mucosa, Peyer's patches, mesenteric lymph nodes, and liver. Human gut HSPCs are phenotypically similar to bone marrow HSPCs and have multilineage differentiation potential in vitro and in vivo. Analysis of circulating post-transplant donor T cells suggests that they undergo selection in recipient lymphoid organs to acquire immune tolerance. Our longitudinal study of human HSPCs carried in intestinal allografts demonstrates their turnover kinetics and gradual replacement of donor-derived HSPCs from a circulating pool. Thus, we have demonstrated the existence of functioning HSPCs in human intestines with implications for promoting tolerance in transplant recipients.
Collapse
Affiliation(s)
- Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Julien Zuber
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Mercedes Martinez
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Brittany Shonts
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Aleksandar Obradovic
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Hui Wang
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Sai-Ping Lau
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Amy Xia
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Elizabeth E Waffarn
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Kristjana Frangaj
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Thomas M Savage
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Michael T Simpson
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Suxiao Yang
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Xinzheng V Guo
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michelle Miron
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Microbiology & Immunology, Columbia University, New York, NY 10032, USA
| | - Takashi Senda
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Surgery, Columbia University, New York, NY 10032, USA
| | - Kortney Rogers
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Adeeb Rahman
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Siu-Hong Ho
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Yufeng Shen
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Adam Griesemer
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Surgery, Columbia University, New York, NY 10032, USA
| | - Donna L Farber
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Microbiology & Immunology, Columbia University, New York, NY 10032, USA; Department of Surgery, Columbia University, New York, NY 10032, USA
| | - Tomoaki Kato
- Department of Surgery, Columbia University, New York, NY 10032, USA
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY 10032, USA; Department of Microbiology & Immunology, Columbia University, New York, NY 10032, USA; Department of Surgery, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
35
|
Profiling calcium signals of in vitro polarized human effector CD4 + T cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:932-943. [PMID: 29626493 DOI: 10.1016/j.bbamcr.2018.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/29/2018] [Accepted: 04/03/2018] [Indexed: 12/21/2022]
Abstract
Differentiation of naïve CD4+ T cells into effector subtypes with distinct cytokine profiles and physiological roles is a tightly regulated process, the imbalance of which can lead to an inadequate immune response or autoimmune disease. The crucial role of Ca2+ signals, mainly mediated by the store operated Ca2+ entry (SOCE) in shaping the immune response is well described. However, it is unclear if human effector CD4+ T cell subsets show differential Ca2+ signatures in response to different stimulation methods. Herein, we provide optimized in vitro culture conditions for polarization of human CD4+ effector T cells and characterize their SOCE following both pharmacological store depletion and direct T-cell receptor (TCR) activation. Moreover, we measured whole cell Ca2+ release activated Ca2+ currents (ICRAC) and investigated whether the observed differences correlate to the expression of CRAC genes. Our results show that Ca2+ profiles of helper CD4+ Th1, Th2 and Th17 are distinct and in part shaped by the intensity of stimulation. Regulatory T cells (Treg) are unique being the subtype with the most prominent SOCE response. Analysis of in vivo differentiated Treg unraveled the role of differential expression of ORAI2 in fine-tuning signals in Treg vs. conventional CD4+ T cells.
Collapse
|
36
|
Tolstykh EI, Degteva MO, Vozilova AV, Anspaugh LR. Local bone-marrow exposure: how to interpret the data on stable chromosome aberrations in circulating lymphocytes? (some comments on the use of FISH method for dose reconstruction for Techa riverside Residents). RADIATION AND ENVIRONMENTAL BIOPHYSICS 2017; 56:389-403. [PMID: 28889186 DOI: 10.1007/s00411-017-0712-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 08/30/2017] [Indexed: 06/07/2023]
Abstract
The method of fluorescence in situ hybridization (FISH) applied to peripheral blood T lymphocytes is used for retrospective dose estimation, and the results obtained from the analysis of stable chromosomal aberrations are usually interpreted as a dose accumulated in the red bone marrow (RBM). However, after local internal exposure of the RBM, doses derived from FISH were found to be lower than those derived from direct measurements of radionuclides accumulated in the bodies of exposed persons. These results were obtained for people residing near the Techa River contaminated by 89,90Sr (beta-emitters) in 1949-1956 (Chelyabinsk Oblast, Russia). A new analysis has been performed of the combined results of FISH studies (n = 178) undertaken during 1994-2012 for persons living on the Techa Riverside. Analysis confirms the lower slope of the translocation yield per Gy (8.0 ± 0.7 × 10-3) for Techa residents in comparison with FISH data for donors with external exposures (11.6 ± 1.6 × 10-3, Tawn et al., Radiat Res 184(3):296-303, 2015). It was suggested that some portion of T cells remained unexposed, because they represented the descendants of T cell progenitors, which had migrated to the thymus before the start of 89,90Sr intakes. To clarify this problem, the dynamics of T-cell Genera (TG), combining all descendants of specific T-cell progenitor reaching the thymus, was considered. Rates of TGs produced by RBM over different age periods of human life were estimated with the use of the mathematic model of T-cell homeostasis (Bains, Mathematical modeling of T-cell homeostasis. A thesis submitted for the degree of Doctor of Philosophy of the University College London. http://discovery.ucl.ac.uk/20159/1/20159.pdf , 2010). The rate of TG loss during the lifetime was assumed to be very small in comparison with production rate. The recirculation of mature T lymphocytes in contaminated RBM was taken into account. According to our model estimates, at the time of blood sampling, the fraction of exposed T lymphocytes (whose progenitors were irradiated) ranged from 20 to 80% depending on the donors' age at the start of exposure to 89,90Sr. Dose to T lymphocytes, estimated from FISH studies, should be about 0.6-0.9 of RBM dose for residents of the upper Techa region and about 0.4-0.8 in the middle Techa region. Our results could explain the lower value of translocation yield per Gy obtained for Techa residents. The approaches for further model improvement and validation are discussed in this paper.
Collapse
Affiliation(s)
- Evgenia I Tolstykh
- Urals Research Center for Radiation Medicine, 68-A, Vorovsky Street, Chelyabinsk, 454076, Russia.
| | - Marina O Degteva
- Urals Research Center for Radiation Medicine, 68-A, Vorovsky Street, Chelyabinsk, 454076, Russia
| | - Alexandra V Vozilova
- Urals Research Center for Radiation Medicine, 68-A, Vorovsky Street, Chelyabinsk, 454076, Russia
| | - Lynn R Anspaugh
- Emeritus, Radiobiology Division, Department of Radiology, University of Utah, Salt Lake City, USA
| |
Collapse
|
37
|
Tormo A, Khodayarian F, Cui Y, Al-Chami E, Kanjarawi R, Noé B, Wang H, Rafei M. Interleukin-21 promotes thymopoiesis recovery following hematopoietic stem cell transplantation. J Hematol Oncol 2017; 10:120. [PMID: 28615039 PMCID: PMC5471903 DOI: 10.1186/s13045-017-0490-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 06/06/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Impaired T cell reconstitution remains a major deterrent in the field of bone marrow (BM) transplantation (BMT) due to pre-conditioning-induced damages inflicted to the thymi of recipient hosts. Given the previously reported thymo-stimulatory property of interleukin (IL)-21, we reasoned that its use post-BMT could have a profound effect on de novo T cell development. METHODS To evaluate the effect of IL-21 on de novo T cell development in vivo, BM derived from RAG2p-GFP mice was transplanted into LP/J mice. Lymphocyte reconstitution was first assessed using a hematological analyzer and a flow cytometer on collected blood samples. Detailed flow cytometry analysis was then performed on the BM, thymus, and spleen of transplanted animals. Finally, the effect of human IL-21 on thymopoiesis was validated in humanized mice. RESULTS Using a major histocompatibility complex (MHC)-matched allogeneic BMT model, we found that IL-21 administration improves immune reconstitution by triggering the proliferation of BM Lin-Sca1+c-kit+ (LSK) subsets. The pharmacological effect of IL-21 also culminates in the recovery of both hematopoietic (thymocytes) and non-hematopoietic (stromal) cells within the thymi of IL-21-treated recipient animals. Although T cells derived from all transplanted groups proliferate, secrete various cytokines, and express granzyme B similarly in response to T cell receptor (TCR) stimulation, full regeneration of peripheral naïve CD4+ and CD8+ T cells and normal TCRvβ distribution could only be detected in IL-21-treated recipient mice. Astonishingly, none of the recipient mice who underwent IL-21 treatment developed graft-versus-host disease (GVHD) in the MHC-matched allogeneic setting while the graft-versus-tumor (GVT) effect was strongly retained. Inhibition of GVHD onset could also be attributed to the enhanced generation of regulatory B cells (B10) observed in the IL-21, but not PBS, recipient mice. We also tested the thymopoiesis-stimulating property of human IL-21 in NSG mice transplanted with cord blood (CB) and found significant improvement in de novo human CD3+ T cell development. CONCLUSIONS In sum, our study indicates that IL-21 represents a new class of unforeseen thymopoietin capable of restoring thymic function following BMT.
Collapse
Affiliation(s)
- Aurélie Tormo
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Fatemeh Khodayarian
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Yun Cui
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Edouard Al-Chami
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Reem Kanjarawi
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Beatriz Noé
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Huijie Wang
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada
| | - Moutih Rafei
- The Department of Pharmacology and Physiology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada. .,The Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, 2900 Edouard-Montpetit BLVD, Montréal, Québec, H3T 1J4, Canada. .,The Department of Microbiology and Immunology, McGill University, 3775 University Street, Montréal, Québec, H3A 2B4, Canada.
| |
Collapse
|
38
|
Punwani D, Zhang Y, Yu J, Cowan MJ, Rana S, Kwan A, Adhikari AN, Lizama CO, Mendelsohn BA, Fahl SP, Chellappan A, Srinivasan R, Brenner SE, Wiest DL, Puck JM. Multisystem Anomalies in Severe Combined Immunodeficiency with Mutant BCL11B. N Engl J Med 2016; 375:2165-2176. [PMID: 27959755 PMCID: PMC5215776 DOI: 10.1056/nejmoa1509164] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Severe combined immunodeficiency (SCID) is characterized by arrested T-lymphocyte production and by B-lymphocyte dysfunction, which result in life-threatening infections. Early diagnosis of SCID through population-based screening of newborns can aid clinical management and help improve outcomes; it also permits the identification of previously unknown factors that are essential for lymphocyte development in humans. METHODS SCID was detected in a newborn before the onset of infections by means of screening of T-cell-receptor excision circles, a biomarker for thymic output. On confirmation of the condition, the affected infant was treated with allogeneic hematopoietic stem-cell transplantation. Exome sequencing in the patient and parents was followed by functional analysis of a prioritized candidate gene with the use of human hematopoietic stem cells and zebrafish embryos. RESULTS The infant had "leaky" SCID (i.e., a form of SCID in which a minimal degree of immune function is preserved), as well as craniofacial and dermal abnormalities and the absence of a corpus callosum; his immune deficit was fully corrected by hematopoietic stem-cell transplantation. Exome sequencing revealed a heterozygous de novo missense mutation, p.N441K, in BCL11B. The resulting BCL11B protein had dominant negative activity, which abrogated the ability of wild-type BCL11B to bind DNA, thereby arresting development of the T-cell lineage and disrupting hematopoietic stem-cell migration; this revealed a previously unknown function of BCL11B. The patient's abnormalities, when recapitulated in bcl11ba-deficient zebrafish, were reversed by ectopic expression of functionally intact human BCL11B but not mutant human BCL11B. CONCLUSIONS Newborn screening facilitated the identification and treatment of a previously unknown cause of human SCID. Coupling exome sequencing with an evaluation of candidate genes in human hematopoietic stem cells and in zebrafish revealed that a constitutional BCL11B mutation caused human multisystem anomalies with SCID and also revealed a prethymic role for BCL11B in hematopoietic progenitors. (Funded by the National Institutes of Health and others.).
Collapse
Affiliation(s)
- Divya Punwani
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Yong Zhang
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Jason Yu
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Morton J Cowan
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Sadhna Rana
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Antonia Kwan
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Aashish N Adhikari
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Carlos O Lizama
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Bryce A Mendelsohn
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Shawn P Fahl
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Ajithavalli Chellappan
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Rajgopal Srinivasan
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Steven E Brenner
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - David L Wiest
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| | - Jennifer M Puck
- From the Department of Pediatrics, University of California, San Francisco (UCSF), School of Medicine and UCSF Benioff Children's Hospital (D.P., J.Y., M.J.C., A.K., B.A.M., J.M.P.), and the Cardiovascular Research Institute, UCSF (C.O.L.), San Francisco, and the Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley (A.N.A., S.E.B.) - all in California; the Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia (Y.Z., S.P.F., D.L.W.); and Innovation Labs, Tata Consultancy Services, Telangana, India (S.R., A.C., R.S.)
| |
Collapse
|
39
|
The non-canonical Wnt receptor Ryk regulates hematopoietic stem cell repopulation in part by controlling proliferation and apoptosis. Cell Death Dis 2016; 7:e2479. [PMID: 27882948 PMCID: PMC5260899 DOI: 10.1038/cddis.2016.380] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/17/2016] [Accepted: 10/18/2016] [Indexed: 12/27/2022]
Abstract
The development of blood and immune cells requires strict control by various signaling pathways in order to regulate self-renewal, differentiation and apoptosis in stem and progenitor cells. Recent evidence indicates critical roles for the canonical and non-canonical Wnt pathways in hematopoiesis. The non-canonical Wnt pathway is important for establishment of cell polarity and cell migration and regulates apoptosis in the thymus. We here investigate the role of the non-canonical Wnt receptor Ryk in hematopoiesis and lymphoid development. We show that there are dynamic changes in Ryk expression during development and in different hematopoietic tissues. Functionally, Ryk regulates NK cell development in a temporal fashion. Moreover, Ryk-deficient mice show diminished, but not absent self-renewal of hematopoietic stem cells (HSC), via effects on mildly increased proliferation and apoptosis. Thus, Ryk deficiency in HSCs from fetal liver reduces their quiescence, leading to proliferation-induced apoptosis and decreased self-renewal.
Collapse
|
40
|
Golec DP, Henao Caviedes LM, Baldwin TA. RasGRP1 and RasGRP3 Are Required for Efficient Generation of Early Thymic Progenitors. THE JOURNAL OF IMMUNOLOGY 2016; 197:1743-53. [PMID: 27465532 DOI: 10.4049/jimmunol.1502107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 06/28/2016] [Indexed: 11/19/2022]
Abstract
T cell development is dependent on the migration of progenitor cells from the bone marrow to the thymus. Upon reaching the thymus, progenitors undergo a complex developmental program that requires inputs from various highly conserved signaling pathways including the Notch and Wnt pathways. To date, Ras signaling has not been implicated in the very earliest stages of T cell differentiation, but members of a family of Ras activators called RasGRPs have been shown to be involved at multiple stages of T cell development. We examined early T cell development in mice lacking RasGRP1, RasGRP3, and RasGRPs 1 and 3. We report that RasGRP1- and RasGRP3-deficient thymi show significantly reduced numbers of early thymic progenitors (ETPs) relative to wild type thymi. Furthermore, RasGRP1/3 double-deficient thymi show significant reductions in ETP numbers compared with either RasGRP1 or RasGRP3 single-deficient thymi, suggesting that both RasGRP1 and RasGRP3 regulate the generation of ETPs. In addition, competitive bone marrow chimera experiments reveal that RasGRP1/3 double-deficient progenitors intrinsically generate ETPs less efficiently than wild type progenitors. Finally, RasGRP1/3-deficient progenitors show impaired migration toward the CCR9 ligand, CCL25, suggesting that RasGRP1 and RasGRP3 may regulate progenitor entry into the thymus through a CCR9-dependent mechanism. These data demonstrate that, in addition to Notch and Wnt, the highly conserved Ras pathway is critical for the earliest stages of T cell development and further highlight the importance of Ras signaling during thymocyte maturation.
Collapse
Affiliation(s)
- Dominic P Golec
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Laura M Henao Caviedes
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Troy A Baldwin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| |
Collapse
|
41
|
Montero-Herradón S, García-Ceca J, Sánchez Del Collado B, Alfaro D, Zapata AG. Eph/ephrin-B-mediated cell-to-cell interactions govern MTS20(+) thymic epithelial cell development. Histochem Cell Biol 2016; 146:167-82. [PMID: 27060907 DOI: 10.1007/s00418-016-1431-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2016] [Indexed: 10/22/2022]
Abstract
Thymus development is a complex process in which cell-to-cell interactions between thymocytes and thymic epithelial cells (TECs) are essential to allow a proper maturation of both thymic cell components. Although signals that control thymocyte development are well known, mechanisms governing TEC maturation are poorly understood, especially those that regulate the maturation of immature TEC populations during early fetal thymus development. In this study, we show that EphB2-deficient, EphB2LacZ and EphB3-deficient fetal thymuses present a lower number of cells and delayed maturation of DN cell subsets compared to WT values. Moreover, deficits in the production of chemokines, known to be involved in the lymphoid seeding into the thymus, contribute in decreased proportions of intrathymic T cell progenitors (PIRA/B(+)) in the mutant thymuses from early stages of development. These features correlate with increased proportions of MTS20(+) cells but fewer MTS20(-) cells from E13.5 onward in the deficient thymuses, suggesting a delayed development of the first epithelial cells. In addition, in vitro the lack of thymocytes or the blockade of Eph/ephrin-B-mediated cell-to-cell interactions between either thymocytes-TECs or TECs-TECs in E13.5 fetal thymic lobes coursed with increased proportions of MTS20(+) TECs. This confirms, for the first time, that the presence of CD45(+) cells, corresponding at these stages to DN1 and DN2 cells, and Eph/ephrin-B-mediated heterotypic or homotypic cell interactions between thymocytes and TECs, or between TECs and themselves, contribute to the early maturation of MTS20(+) TECs.
Collapse
Affiliation(s)
- Sara Montero-Herradón
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, C/José Antonio Novais 2, 28040, Madrid, Spain
| | - Javier García-Ceca
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, C/José Antonio Novais 2, 28040, Madrid, Spain
| | - Beatriz Sánchez Del Collado
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, C/José Antonio Novais 2, 28040, Madrid, Spain
| | - David Alfaro
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, C/José Antonio Novais 2, 28040, Madrid, Spain
| | - Agustín G Zapata
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, C/José Antonio Novais 2, 28040, Madrid, Spain.
| |
Collapse
|
42
|
Bredenkamp N, Jin X, Liu D, O'Neill KE, Manley NR, Blackburn CC. Construction of a functional thymic microenvironment from pluripotent stem cells for the induction of central tolerance. Regen Med 2016; 10:317-29. [PMID: 25933240 DOI: 10.2217/rme.15.8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The thymus is required for generation of a self-tolerant, self-restricted T-cell repertoire. The capacity to manipulate or replace thymus function therapeutically would be beneficial in a variety of clinical settings, including for improving recovery following bone marrow transplantation, restoring immune system function in the elderly and promoting tolerance to transplanted organs or cells. An attractive strategy would be transplantation of thymus organoids generated from cells produced in vitro, for instance from pluripotent stem cells. Here, we review recent progress toward this goal, focusing on advances in directing differentiation of pluripotent stem cells to thymic epithelial cells, a key cell type of the thymic stroma, and related direct reprogramming strategies.
Collapse
Affiliation(s)
- Nicholas Bredenkamp
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, SCRM Building, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | | | | | | | | | | |
Collapse
|
43
|
Famili F, Naber BAE, Vloemans S, de Haas EFE, Tiemessen MM, Staal FJT. Discrete roles of canonical and non-canonical Wnt signaling in hematopoiesis and lymphopoiesis. Cell Death Dis 2015; 6:e1981. [PMID: 26583322 PMCID: PMC4670932 DOI: 10.1038/cddis.2015.326] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/30/2015] [Accepted: 10/09/2015] [Indexed: 12/17/2022]
Abstract
The mechanisms that regulate proliferation, fate decisions and differentiation of hematopoietic stem cells (HSC) and thymic stem cells are highly complex. Several signaling pathways including Wnt signaling have important roles during these processes. Both canonical and non-canonical Wnt signaling are important in normal and malignant hematopoiesis and lymphoid development, yet their precise roles are controversial. In a side-by-side comparison, we investigated the roles of the canonical and non-canonical Wnt pathway in hematopoiesis and thymopoiesis. As complete loss-of-function models for non-canonical Wnt signaling are not yet available and highly complex for canonical Wnt signaling, we decided to use a gain-of-function approach. To this end, Wnt3a and Wn5a, two well-known prototypical canonical and non-canonical Wnt ligands were produced in hematopoiesis supporting stromal assays. High levels of Wnt3a signaling blocked T-cell development at early stages, whereas intermediate levels accelerated T-cell development. In contrast, Wnt5a signaling prompted apoptosis in developing thymocytes, without affecting differentiation at a particular stage. To explore the role of Wnt3a and Wnt5a in vivo, we transduced HSCs isolated from fetal liver, transduced with Wnt3a and Wnt5a vectors, and performed reconstitution assays in irradiated C57Bl/6 mice. Wnt3a overexpression led to increased lymphopoiesis, whereas Wnt5a augments myelopoiesis in the bone marrow (BM) and spleen. Thus, the canonical and non-canonical Wnt signaling have discrete roles in hematopoiesis and thymopoiesis, and understanding their right dose of action is crucial for prospective translational applications.
Collapse
Affiliation(s)
- F Famili
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center, Leiden, The Netherlands
| | - B A E Naber
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center, Leiden, The Netherlands
| | - S Vloemans
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center, Leiden, The Netherlands
| | - E F E de Haas
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center, Leiden, The Netherlands
| | - M M Tiemessen
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center, Leiden, The Netherlands
| | - F J T Staal
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
44
|
Farese AM, Hankey KG, Cohen MV, MacVittie TJ. Lymphoid and Myeloid Recovery in Rhesus Macaques Following Total Body X-Irradiation. HEALTH PHYSICS 2015; 109:414-26. [PMID: 26425902 PMCID: PMC4593069 DOI: 10.1097/hp.0000000000000348] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Recovery from severe immunosuppression requires hematopoietic stem cell reconstitution and effective thymopoiesis to restore a functional immune cell repertoire. Herein, a model of immune cell reconstitution consequent to potentially lethal doses of irradiation is described, which may be valuable in evaluating potential medical countermeasures. Male rhesus macaques were total body irradiated by exposure to 6.00 Gy 250 kVp x-radiation (midline tissue dose, 0.13 Gy min), resulting in an approximate LD10/60 (n = 5/59). Animals received medical management, and hematopoietic and immune cell recovery was assessed (n ≤ 14) through 370 d post exposure. A subset of animals (n ≤ 8) was examined through 700 d. Myeloid recovery was assessed by neutrophil and platelet-related parameters. Lymphoid recovery was assessed by the absolute lymphocyte count and FACS-based phenotyping of B- and T-cell subsets. Recent thymic emigrants were identified by T cell receptor excision circle quantification. Severe neutropenia, lymphopenia, and thrombocytopenia resolved within 30 d. Total CD3+ cells μL required 60 d to reach values 60% of normal, followed by subsequent slow recovery to approximately normal by 180 d post irradiation. Recovery of CD3+4+ and CD3+8+ cell memory and naïve subsets were markedly different. Memory populations were ≥ 100% of normal by day 60, whereas naïve populations were only 57% normal at 180 d and never fully recovered to baseline post irradiation. Total (CD20+) B cells μL were within normal levels by 77 d post exposure. This animal model elucidates the variable T- and B-cell subset recovery kinetics after a potentially lethal dose of total-body irradiation that are dependent on marrow-derived stem and progenitor cell recovery, peripheral homeostatic expansion, and thymopoiesis.
Collapse
Affiliation(s)
- Ann M. Farese
- University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD
| | - Kim G. Hankey
- University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD
| | | | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD
| |
Collapse
|
45
|
Alexandropoulos K, Bonito AJ, Weinstein EG, Herbin O. Medullary thymic epithelial cells and central tolerance in autoimmune hepatitis development: novel perspective from a new mouse model. Int J Mol Sci 2015; 16:1980-2000. [PMID: 25603179 PMCID: PMC4307344 DOI: 10.3390/ijms16011980] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/07/2015] [Indexed: 02/07/2023] Open
Abstract
Autoimmune hepatitis (AIH) is an immune-mediated disorder that affects the liver parenchyma. Diagnosis usually occurs at the later stages of the disease, complicating efforts towards understanding the causes of disease development. While animal models are useful for studying the etiology of autoimmune disorders, most of the existing animal models of AIH do not recapitulate the chronic course of the human condition. In addition, approaches to mimic AIH-associated liver inflammation have instead led to liver tolerance, consistent with the high tolerogenic capacity of the liver. Recently, we described a new mouse model that exhibited spontaneous and chronic liver inflammation that recapitulated the known histopathological and immunological parameters of AIH. The approach involved liver-extrinsic genetic engineering that interfered with the induction of T-cell tolerance in the thymus, the very process thought to inhibit AIH induction by liver-specific expression of exogenous antigens. The mutation led to depletion of specialized thymic epithelial cells that present self-antigens and eliminate autoreactive T-cells before they exit the thymus. Based on our findings, which are summarized below, we believe that this mouse model represents a relevant experimental tool towards elucidating the cellular and molecular aspects of AIH development and developing novel therapeutic strategies for treating this disease.
Collapse
Affiliation(s)
- Konstantina Alexandropoulos
- Department of Medicine and Clinical Immunology, the Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1089, New York, NY 10029, USA.
| | - Anthony J Bonito
- Department of Medicine and Clinical Immunology, the Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1089, New York, NY 10029, USA.
| | - Erica G Weinstein
- Department of Medicine and Clinical Immunology, the Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1089, New York, NY 10029, USA
| | - Olivier Herbin
- Department of Medicine and Clinical Immunology, the Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1089, New York, NY 10029, USA.
| |
Collapse
|
46
|
El-Sayed YS, Shimizu R, Onoda A, Takeda K, Umezawa M. Carbon black nanoparticle exposure during middle and late fetal development induces immune activation in male offspring mice. Toxicology 2015; 327:53-61. [DOI: 10.1016/j.tox.2014.11.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 11/20/2014] [Accepted: 11/20/2014] [Indexed: 12/26/2022]
|
47
|
Huijskens MJAJ, Walczak M, Koller N, Briedé JJ, Senden-Gijsbers BLMG, Schnijderberg MC, Bos GMJ, Germeraad WTV. Technical advance: ascorbic acid induces development of double-positive T cells from human hematopoietic stem cells in the absence of stromal cells. J Leukoc Biol 2014; 96:1165-75. [PMID: 25157026 DOI: 10.1189/jlb.1ta0214-121rr] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The efficacy of donor HSCT is partly reduced as a result of slow post-transplantation immune recovery. In particular, T cell regeneration is generally delayed, resulting in high infection-related mortality in the first years post-transplantation. Adoptive transfer of in vitro-generated human T cell progenitors seems a promising approach to accelerate T cell recovery in immunocompromised patients. AA may enhance T cell proliferation and differentiation in a controlled, feeder-free environment containing Notch ligands and defined growth factors. Our experiments show a pivotal role for AA during human in vitro T cell development. The blocking of NOS diminished this effect, indicating a role for the citrulline/NO cycle. AA promotes the transition of proT1 to proT2 cells and of preT to DP T cells. Furthermore, the addition of AA to feeder cocultures resulted in development of DP and SP T cells, whereas without AA, a preT cell-stage arrest occurred. We conclude that neither DLL4-expressing feeder cells nor feeder cell conditioned media are required for generating DP T cells from CB and G-CSF-mobilized HSCs and that generation and proliferation of proT and DP T cells are greatly improved by AA. This technology could potentially be used to generate T cell progenitors for adoptive therapy.
Collapse
Affiliation(s)
- Mirelle J A J Huijskens
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center, and
| | - Mateusz Walczak
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center, and
| | - Nicole Koller
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center, and
| | - Jacob J Briedé
- Department of Toxicogenomics, School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | | | - Melanie C Schnijderberg
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center, and
| | - Gerard M J Bos
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center, and
| | - Wilfred T V Germeraad
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center, and
| |
Collapse
|
48
|
Bose TO, Colpitts SL, Pham QM, Puddington L, Lefrançois L. CD11a is essential for normal development of hematopoietic intermediates. THE JOURNAL OF IMMUNOLOGY 2014; 193:2863-72. [PMID: 25108025 DOI: 10.4049/jimmunol.1301820] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The process of lymphopoiesis begins in the bone marrow (BM) and requires multiple cellular intermediates. For T cell production, lymphoid progenitors exit the BM and home to the thymus where maturation and selection ensue. These processes are dependent on a number of factors, including chemokines and adhesion molecules. Although the β2 integrin CD11a plays an important role in the migration of lymphocytes to lymph nodes, the role of CD11a in T cell development is largely undefined. Our studies now show that, in CD11a(-/-) mice, thymic cellularity was decreased and early T cell development was partially impaired. Remarkably, CD11a was critical for generation of common lymphoid progenitors (CLPs) and lymphoid-primed multipotent progenitors. However, in intact CD11a(-/-) mice, peripheral B and T cell subsets were only modestly altered, suggesting that compensatory mechanisms were operating. In contrast, competitive BM-reconstitution assays revealed an essential role for CD11a in the generation of thymocytes and mature T and B cells. This defect was linked to the requirement for CD11a in the development of CLPs. Furthermore, our results identified CLPs, and not lymphoid-primed multipotent progenitors, as the requisite CD11a-dependent precursor for lymphocyte development. Thus, these findings established a key role for CD11a in lymphopoiesis.
Collapse
Affiliation(s)
- Tina O Bose
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030
| | - Sara L Colpitts
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030
| | - Quynh-Mai Pham
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030
| | - Lynn Puddington
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030
| | - Leo Lefrançois
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030
| |
Collapse
|
49
|
Kyoizumi S, Kubo Y, Kajimura J, Yoshida K, Hayashi T, Nakachi K, Young LF, Moore MA, van den Brink MRM, Kusunoki Y. Linkage between dendritic and T cell commitments in human circulating hematopoietic progenitors. THE JOURNAL OF IMMUNOLOGY 2014; 192:5749-60. [PMID: 24835400 DOI: 10.4049/jimmunol.1303260] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The relationships between commitments of dendritic cells (DCs) and T cells in human hematopoietic stem cells are not well understood. In this study, we enumerate and characterize conventional DC and plasmacytoid DC precursors in association with T cell and thymus-derived types of NK cell precursors among CD34(+) hematopoietic progenitor cells (HPCs) circulating in human peripheral blood. By limiting-dilution analyses using coculture with stroma cells expressing Notch1 ligand, the precursor frequencies (PFs) of DCs in HPCs were found to significantly correlate with T cell PFs, but not with NK cell PFs, among healthy donors. Clonal analyses showed that the majority of T/NK dual- and T single-lineage precursors-but only a minority of NK single-lineage precursors-were associated with the generation of DC progenies. All clones producing both DC and T cell progenies were found with monocyte and/or granulocyte progenies, suggesting DC differentiation via myeloid DC pathways. Analyses of peripheral blood HPC subpopulations revealed that the lineage split between DC and T/NK cell progenitor occurs at the stage prior to bifurcation into T and NK cell lineages. The findings suggest a strong linkage between DC and T cell commitments, which may be imprinted in circulating lymphoid-primed multipotent progenitors or in more upstream HPCs.
Collapse
Affiliation(s)
- Seishi Kyoizumi
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan;
| | - Yoshiko Kubo
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| | - Junko Kajimura
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| | - Kengo Yoshida
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| | - Tomonori Hayashi
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| | - Kei Nakachi
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| | - Lauren F Young
- Department of Medicine and Immunology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065; and
| | - Malcolm A Moore
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Marcel R M van den Brink
- Department of Medicine and Immunology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065; and
| | - Yoichiro Kusunoki
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| |
Collapse
|
50
|
Glauzy S, André-Schmutz I, Larghero J, Ezine S, de Latour RP, Moins-Teisserenc H, Servais S, Robin M, Socié G, Clave E, Toubert A. CXCR4-related increase of circulating human lymphoid progenitors after allogeneic hematopoietic stem cell transplantation. PLoS One 2014; 9:e91492. [PMID: 24621606 PMCID: PMC3951398 DOI: 10.1371/journal.pone.0091492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/10/2014] [Indexed: 01/05/2023] Open
Abstract
Immune recovery after profound lymphopenia is a major challenge in many clinical situations, such as allogeneic hematopoietic stem cell transplantation (allo-HSCT). Recovery depends, in a first step, on hematopoietic lymphoid progenitors production in the bone marrow (BM). In this study, we characterized CD34+Lin-CD10+ lymphoid progenitors in the peripheral blood of allo-HSCT patients. Our data demonstrate a strong recovery of this population 3 months after transplantation. This rebound was abolished in patients who developed acute graft-versus-host disease (aGVHD). A similar recovery profile was found for both CD24+ and CD24- progenitor subpopulations. CD34+lin-CD10+CD24- lymphoid progenitors sorted from allo-HSCT patients preserved their T cell potentiel according to in vitro T-cell differentiation assay and the expression profile of 22 genes involved in T-cell differentiation and homing. CD34+lin-CD10+CD24- cells from patients without aGVHD had reduced CXCR4 gene expression, consistent with an enhanced egress from the BM. CCR7 gene expression was reduced in patients after allo-HSCT, as were its ligands CCL21 and CCL19. This reduction was particularly marked in patients with aGVHD, suggesting a possible impact on thymic homing. Thus, the data presented here identify this population as an important early step in T cell reconstitution in humans and so, an important target when seeking to enhance immune reconstitution.
Collapse
Affiliation(s)
- Salomé Glauzy
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Isabelle André-Schmutz
- INSERM U768, Université Paris Descartes, Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Jérôme Larghero
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Unité de Thérapie Cellulaire et CIC de Biothérapies, Hôpital Saint-Louis, AP-HP, Paris, France
| | | | - Régis Peffault de Latour
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- Service d'Hématologie-Greffe de Moelle, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Hélène Moins-Teisserenc
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Sophie Servais
- Service d'Hématologie-Greffe de Moelle, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Marie Robin
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- Service d'Hématologie-Greffe de Moelle, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Gérard Socié
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Service d'Hématologie-Greffe de Moelle, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Emmanuel Clave
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Antoine Toubert
- Univ Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
- INSERM UMR1160, Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Hôpital Saint-Louis, AP-HP, Paris, France
| |
Collapse
|