201
|
Takahashi H, Ikeda K, Ogawa K, Saito S, Ngoma AM, Mashimo Y, Ueda K, Furukawa M, Shichishima-Nakamura A, Ohkawara H, Nollet KE, Ohto H, Takeishi Y. CD4+ T cells in aged or thymectomized recipients of allogeneic stem cell transplantations. Biol Res 2015. [PMID: 26210500 PMCID: PMC4514962 DOI: 10.1186/s40659-015-0033-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background CD4+CD25highFOXP3+ regulatory T (Treg) cells, which include thymus-derived and peripherally induced cells, play a central role in immune regulation, and are therefore crucial to prevent graft-versus-host disease (GVHD). The increasing use of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for elderly patients with thymus regression, and our case of allo-HSCT shortly after total thymectomy, raised questions about the activity of thymus-derived Treg cells and peripherally induced Treg cells, which are otherwise indistinguishable. Results We found that despite pre-transplant thymectomy or older age, both naïve and effector Treg cells, as well as naïve and effector conventional T cells, proliferated in allo-HSCT recipients. Higher proportions of total Treg cells 1 month post allo-HSCT, and naïve Treg cells 1 year post allo-HSCT, appeared in patients achieving complete chimera without developing significant chronic GVHD, including our thymectomized patient, compared with patients who developed chronic GVHD. Conclusions Treg cells that modulate human allogeneic immunity may arise peripherally as well as in the thymus of allo-HSCT recipients.
Collapse
Affiliation(s)
- Hiroshi Takahashi
- Department of Cardiology and Hematology, School of Medicine, Fukushima Medical University, Fukushima, Japan.
| | - Kazuhiko Ikeda
- Department of Cardiology and Hematology, School of Medicine, Fukushima Medical University, Fukushima, Japan. .,Department of Blood Transfusion and Transplantation Immunology, School of Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima, Fukushima, 960-1295, Japan.
| | - Kazuei Ogawa
- Department of Cardiology and Hematology, School of Medicine, Fukushima Medical University, Fukushima, Japan.
| | - Syunnichi Saito
- Department of Blood Transfusion and Transplantation Immunology, School of Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima, Fukushima, 960-1295, Japan.
| | - Alain M Ngoma
- Department of Blood Transfusion and Transplantation Immunology, School of Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima, Fukushima, 960-1295, Japan. .,Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada.
| | - Yumiko Mashimo
- Department of Cardiology and Hematology, School of Medicine, Fukushima Medical University, Fukushima, Japan.
| | - Koki Ueda
- Department of Cardiology and Hematology, School of Medicine, Fukushima Medical University, Fukushima, Japan.
| | - Miki Furukawa
- Department of Cardiology and Hematology, School of Medicine, Fukushima Medical University, Fukushima, Japan.
| | - Akiko Shichishima-Nakamura
- Department of Cardiology and Hematology, School of Medicine, Fukushima Medical University, Fukushima, Japan.
| | - Hiroshi Ohkawara
- Department of Cardiology and Hematology, School of Medicine, Fukushima Medical University, Fukushima, Japan.
| | - Kenneth E Nollet
- Department of Blood Transfusion and Transplantation Immunology, School of Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima, Fukushima, 960-1295, Japan.
| | - Hitoshi Ohto
- Department of Blood Transfusion and Transplantation Immunology, School of Medicine, Fukushima Medical University, 1 Hikariga-oka, Fukushima, Fukushima, 960-1295, Japan.
| | - Yasuchika Takeishi
- Department of Cardiology and Hematology, School of Medicine, Fukushima Medical University, Fukushima, Japan.
| |
Collapse
|
202
|
St-Pierre C, Trofimov A, Brochu S, Lemieux S, Perreault C. Differential Features of AIRE-Induced and AIRE-Independent Promiscuous Gene Expression in Thymic Epithelial Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:498-506. [PMID: 26034170 DOI: 10.4049/jimmunol.1500558] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/04/2015] [Indexed: 12/16/2023]
Abstract
Establishment of self-tolerance in the thymus depends on promiscuous expression of tissue-restricted Ags (TRA) by thymic epithelial cells (TEC). This promiscuous gene expression (pGE) is regulated in part by the autoimmune regulator (AIRE). To evaluate the commonalities and discrepancies between AIRE-dependent and -independent pGE, we analyzed the transcriptome of the three main TEC subsets in wild-type and Aire knockout mice. We found that the impact of AIRE-dependent pGE is not limited to generation of TRA. AIRE decreases, via non-cell autonomous mechanisms, the expression of genes coding for positive regulators of cell proliferation, and it thereby reduces the number of cortical TEC. In mature medullary TEC, AIRE-driven pGE upregulates non-TRA coding genes that enhance cell-cell interactions (e.g., claudins, integrins, and selectins) and are probably of prime relevance to tolerance induction. We also found that AIRE-dependent and -independent TRA present several distinctive features. In particular, relative to AIRE-induced TRA, AIRE-independent TRA are more numerous and show greater splicing complexity. Furthermore, we report that AIRE-dependent versus -independent TRA project nonredundant representations of peripheral tissues in the thymus.
Collapse
Affiliation(s)
- Charles St-Pierre
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada; and
| | - Assya Trofimov
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada; and Department of Computer Science and Operations Research, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Sylvie Brochu
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada; and
| | - Sébastien Lemieux
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Computer Science and Operations Research, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Claude Perreault
- Institute for Research in Immunology and Cancer, University of Montreal, Montreal, Quebec H3C 3J7, Canada; Department of Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada; and
| |
Collapse
|
203
|
Nikoopour E, Bellemore SM, Singh B. IL-22, cell regeneration and autoimmunity. Cytokine 2015; 74:35-42. [PMID: 25467639 DOI: 10.1016/j.cyto.2014.09.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/18/2014] [Accepted: 09/20/2014] [Indexed: 12/16/2022]
Abstract
IL-22 as a cytokine is described with opposing pro-inflammatory and anti-inflammatory functions. Cell regeneration, tissue remodelling and balance between commensal bacteria in the gut and host immune system are considered as anti-inflammatory features of IL-22, whereas production of IL-22 from Th17 cells links this cytokine to pro-inflammatory pathways. Th17 cells and group 3 innate lymphoid cells (ILC3) are two major producers of IL-22 and both cell types express ROR-γt and Aryl hydrocarbon receptor (AhR) transcription factors. Typically, the immune system cells are the main producers of IL-22. However, targets of this cytokine are mostly non-hematopoietic cells such as hepatocytes, keratinocytes, and epithelial cells of lung and intestine. Association of IL-22 with other cytokines or transcription factors in different cell types might explain its contrasting role in health and disease. In this review we discuss the regulation of IL-22 production by AhR- and IL-23-driven pathways. A clear understanding of the biology of IL-22 will provide new opportunities for its application to improve human health involving many debilitating conditions.
Collapse
Affiliation(s)
- Enayat Nikoopour
- Centre for Human Immunology, Department of Microbiology and Immunology, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Stacey M Bellemore
- Centre for Human Immunology, Department of Microbiology and Immunology, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Bhagirath Singh
- Centre for Human Immunology, Department of Microbiology and Immunology, Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
204
|
Hernández PP, Mahlakoiv T, Yang I, Schwierzeck V, Nguyen N, Guendel F, Gronke K, Ryffel B, Hoelscher C, Dumoutier L, Renauld JC, Suerbaum S, Staeheli P, Diefenbach A. Interferon-λ and interleukin 22 act synergistically for the induction of interferon-stimulated genes and control of rotavirus infection. Nat Immunol 2015; 16:698-707. [PMID: 26006013 PMCID: PMC4589158 DOI: 10.1038/ni.3180] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/27/2015] [Indexed: 12/13/2022]
Abstract
The epithelium is the main entry point for many viruses, but the processes that protect barrier surfaces against viral infections are incompletely understood. Here we identified interleukin 22 (IL-22) produced by innate lymphoid cell group 3 (ILC3) as an amplifier of signaling via interferon-λ (IFN-λ), a synergism needed to curtail the replication of rotavirus, the leading cause of childhood gastroenteritis. Cooperation between the receptor for IL-22 and the receptor for IFN-λ, both of which were 'preferentially' expressed by intestinal epithelial cells (IECs), was required for optimal activation of the transcription factor STAT1 and expression of interferon-stimulated genes (ISGs). These data suggested that epithelial cells are protected against viral replication by co-option of two evolutionarily related cytokine networks. These data may inform the design of novel immunotherapy for viral infections that are sensitive to interferons.
Collapse
Affiliation(s)
- Pedro P. Hernández
- Research Centre Immunology and Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
- Max-Planck-Institute for Immunobiology and Epigenetics, Stübeweg 51, D-79108 Freiburg, Germany
| | - Tanel Mahlakoiv
- Department of Medical Microbiology and Hygiene, Institute for Virology, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstrasse 19A, D-79104 Freiburg, Germany
| | - Ines Yang
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany and DZIF – German Center for Infection Research, Hannover-Braunschweig Site, D-30625 Hannover, Germany
| | - Vera Schwierzeck
- Research Centre Immunology and Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
| | - Nam Nguyen
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
| | - Fabian Guendel
- Research Centre Immunology and Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
- Research Training Group (GRK1104) of Organogenesis, Hauptstrasse 1, D-79104 Freiburg, Germany
| | - Konrad Gronke
- Research Centre Immunology and Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
- Max-Planck-Institute for Immunobiology and Epigenetics, Stübeweg 51, D-79108 Freiburg, Germany
| | - Bernhard Ryffel
- INEM - UMR7355, Molecular Immunology, University and CNRS, F-45071 Orleans, France and Institute of Infectious Disease, University of Cape Town, RSA
| | - Christoph Hoelscher
- Infection Immunology Research, Research Center Borstel, D-23845 Borstel, Germany
- Cluster of Excellence Inflammation at Interfaces (Borstel-Kiel-Lübeck-Plön)
| | - Laure Dumoutier
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, 74 Avenue Hippocrate, B-1200 Brussels, Belgium
| | - Jean-Christophe Renauld
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, 74 Avenue Hippocrate, B-1200 Brussels, Belgium
| | - Sebastian Suerbaum
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany and DZIF – German Center for Infection Research, Hannover-Braunschweig Site, D-30625 Hannover, Germany
| | - Peter Staeheli
- Department of Medical Microbiology and Hygiene, Institute for Virology, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
| | - Andreas Diefenbach
- Research Centre Immunology and Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
- Research Training Group (GRK1104) of Organogenesis, Hauptstrasse 1, D-79104 Freiburg, Germany
| |
Collapse
|
205
|
Innate lymphoid cells in the initiation, regulation and resolution of inflammation. Nat Med 2015; 21:698-708. [PMID: 26121198 DOI: 10.1038/nm.3892] [Citation(s) in RCA: 380] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/27/2015] [Indexed: 12/12/2022]
Abstract
A previously unappreciated cell type of the innate immune system, termed innate lymphoid cells (ILCs), has been characterized in mice and humans and found to influence the induction, regulation and resolution of inflammation. ILCs have an important role in these processes in mouse models of infection, inflammation and tissue repair. Further, disease-association studies in defined patient populations have identified significant alterations in ILC responses, suggesting a potential role for these cell populations in human health and disease. In this review we discuss the emerging family of ILCs, the role of ILCs in inflammation, and how current or novel therapeutic strategies could be used to selectively modulate ILC responses and limit chronic inflammatory diseases.
Collapse
|
206
|
Kim KY, Lee G, Yoon M, Cho EH, Park CS, Kim MG. Expression Analyses Revealed Thymic Stromal Co-Transporter/Slc46A2 Is in Stem Cell Populations and Is a Putative Tumor Suppressor. Mol Cells 2015; 38:548-61. [PMID: 26013383 PMCID: PMC4469913 DOI: 10.14348/molcells.2015.0044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/10/2015] [Accepted: 03/10/2015] [Indexed: 01/08/2023] Open
Abstract
By combining conventional single cell analysis with flow cytometry and public database searches with bioinformatics tools, we extended the expression profiling of thymic stromal cotransporter (TSCOT), Slc46A2/Ly110, that was shown to be expressed in bipotent precursor and cortical thymic epithelial cells. Genome scale analysis verified TSCOT expression in thymic tissue- and cell type- specific fashion and is also expressed in some other epithelial tissues including skin and lung. Coexpression profiling with genes, Foxn1 and Hoxa3, revealed the role of TSCOT during the organogenesis. TSCOT expression was detected in all thymic epithelial cells (TECs), but not in the CD31(+) endothelial cell lineage in fetal thymus. In addition, ABC transporter-dependent side population and Sca-1(+) fetal TEC populations both contain TSCOT-expressing cells, indicating TEC stem cells express TSCOT. TSCOT expression was identified as early as in differentiating embryonic stem cells. TSCOT expression is not under the control of Foxn1 since TSCOT is present in the thymic rudiment of nude mice. By searching variations in the expression levels, TSCOT is positively associated with Grhl3 and Irf6. Cytokines such as IL1b, IL22 and IL24 are the potential regulators of the TSCOT expression. Surprisingly, we found TSCOT expression in the lung is diminished in lung cancers, suggesting TSCOT may be involved in the suppression of lung tumor development. Based on these results, a model for TEC differentiation from the stem cells was proposed in context of multiple epithelial organ formation.
Collapse
Affiliation(s)
- Ki Yeon Kim
- Department of Biological Sciences, Inha University, Incheon 402-720,
Korea
| | - Gwanghee Lee
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110,
USA
| | - Minsang Yoon
- Department of Biological Sciences, Inha University, Incheon 402-720,
Korea
| | - Eun Hye Cho
- Department of Biological Sciences, Inha University, Incheon 402-720,
Korea
| | - Chan-Sik Park
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 138-736,
Korea
| | - Moon Gyo Kim
- Department of Biological Sciences, Inha University, Incheon 402-720,
Korea
| |
Collapse
|
207
|
Eberl G, Colonna M, Di Santo JP, McKenzie ANJ. Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology. Science 2015; 348:aaa6566. [PMID: 25999512 DOI: 10.1126/science.aaa6566] [Citation(s) in RCA: 609] [Impact Index Per Article: 67.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Innate lymphoid cells (ILCs) are a growing family of immune cells that mirror the phenotypes and functions of T cells. However, in contrast to T cells, ILCs do not express acquired antigen receptors or undergo clonal selection and expansion when stimulated. Instead, ILCs react promptly to signals from infected or injured tissues and produce an array of secreted proteins termed cytokines that direct the developing immune response into one that is adapted to the original insult. The complex cross-talk between microenvironment, ILCs, and adaptive immunity remains to be fully deciphered. Only by understanding these complex regulatory networks can the power of ILCs be controlled or unleashed in order to regulate or enhance immune responses in disease prevention and therapy.
Collapse
Affiliation(s)
- Gérard Eberl
- Institut Pasteur, Microenvironment and Immunity Unit, 75724 Paris, France.
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James P Di Santo
- Institut Pasteur, Innate Immunity Unit, INSERM U668, 75724 Paris, France
| | - Andrew N J McKenzie
- Medical Research Council (MRC) Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| |
Collapse
|
208
|
Weidenbusch M, Rodler S, Anders HJ. Interleukin-22 in kidney injury and regeneration. Am J Physiol Renal Physiol 2015; 308:F1041-6. [PMID: 25740595 DOI: 10.1152/ajprenal.00005.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/02/2015] [Indexed: 12/20/2022] Open
Abstract
Interleukins have become well-known regulators of innate and adaptive immunity-related tissue inflammation. Recently, IL-22 has gained a lot of interest for its unique functions in maintaining and regaining epithelial integrity. IL-22 is exclusively secreted by different immune cell subsets, while IL-22 receptors are mainly expressed by epithelial cells. As the kidney is largely an epithelial organ, the functional role of IL-22 in the kidney deserves to be explored in detail. Here, we briefly summarize the key features of IL-22 biology and review the available data on its expression and functional roles in kidney injury and kidney regeneration. Furthermore, we provide suggestions on how to explore this evolving field in the future.
Collapse
Affiliation(s)
- Marc Weidenbusch
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, München, Germany
| | - Severin Rodler
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, München, Germany
| | - Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, München, Germany
| |
Collapse
|
209
|
Leyva-Rangel JP, de Los Angeles Hernández-Cueto M, Galan-Enriquez CS, López-Medina M, Ortiz-Navarrete V. Bacterial clearance reverses a skewed T-cell repertoire induced by Salmonella infection. IMMUNITY INFLAMMATION AND DISEASE 2015; 3:209-23. [PMID: 26417438 PMCID: PMC4578521 DOI: 10.1002/iid3.60] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 11/06/2022]
Abstract
Salmonella typhimurium invades the spleen, liver, and peripheral lymph nodes and has recently been detected in the bone marrow and thymus, resulting in a reduced thymic size and a decline in the total number of thymic cells. A specific deletion of the double-positive cell subset has been characterized, yet the export of mature T cells to the periphery remains normal. We analyzed Salmonella pathogenesis regarding thymic structure and the T-cell maturation process. We demonstrate that, despite alterations in the thymic structure, T-cell development is maintained during Salmonella infection, allowing the selection of single-positive T-cell clones expressing particular T-cell receptor beta chains (TCR-Vβ). Moreover, the treatment of infected mice with an antibiotic restored the normal thymic architecture and thymocyte subset distribution. Additionally, the frequency of TCR-Vβ usage after treatment was comparable to that in non-infected mice. However, bacteria were still recovered from the thymus after 1 month of treatment. Our data reveal that a skewed T-cell developmental process is present in the Salmonella-infected thymus that alters the TCR-Vβ usage frequency. Likewise, the post-treatment persistence of Salmonella reveals a novel function of the thymus as a potential reservoir for this infectious agent.
Collapse
Affiliation(s)
- Jessica P Leyva-Rangel
- Doctorado en Ciencias Biomédicas Facultad de Medicina, UNAM Mexico City, CP 045510, Mexico ; Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politecnico Nacional (CINVESTAV) Mexico City
| | | | - Carlos-Samuel Galan-Enriquez
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politecnico Nacional (CINVESTAV) Mexico City
| | - Marcela López-Medina
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politecnico Nacional (CINVESTAV) Mexico City
| | - Vianney Ortiz-Navarrete
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politecnico Nacional (CINVESTAV) Mexico City
| |
Collapse
|
210
|
Ohtani S, Ushiyama A, Maeda M, Ogasawara Y, Wang J, Kunugita N, Ishii K. The effects of radio-frequency electromagnetic fields on T cell function during development. JOURNAL OF RADIATION RESEARCH 2015; 56:467-74. [PMID: 25835473 PMCID: PMC4426920 DOI: 10.1093/jrr/rru126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/13/2014] [Accepted: 12/20/2014] [Indexed: 05/24/2023]
Abstract
With the widespread use of radio-frequency devices, it is increasingly important to understand the biological effects of the associated electromagnetic fields. Thus, we investigated the effects of radio-frequency electromagnetic fields (RF-EMF) on T cell responses during development due to the lack of science-based evidence for RF-EMF effects on developmental immune systems. Sprague Dawley (SD) rats were exposed to 2.14-GHz wideband code division multiple-access (W-CDMA) RF signals at a whole-body specific absorption rate (SAR) of 0.2 W/kg. Exposures were performed for a total of 9 weeks spanning in utero development, lactation and the juvenile period. Rats were continuously exposed to RF-EMF for 20 h/day, 7 days/week. Comparisons of control and exposed rats using flow cytometry revealed no changes in the numbers of CD4/CD8 T cells, activated T cells or regulatory T cells among peripheral blood cells, splenocytes and thymocytes. Expression levels of 16 genes that regulate the immunological Th1/Th2 paradigm were analyzed using real-time PCR in the spleen and thymus tissues of control and RF-EMF-exposed rats. Although only the Il5 gene was significantly regulated in spleen tissues, Il4, Il5 and Il23a genes were significantly upregulated in thymus tissues following exposure to RF-EMF. However, ELISAs showed no changes in serum IL-4 protein concentrations. These data indicate no adverse effects of long-term RF-EMF exposure on immune-like T cell populations, T cell activation, or Th1/Th2 balance in developing rats, although significant transcriptional effects were observed.
Collapse
Affiliation(s)
- Shin Ohtani
- Department of Hygienic Chemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Akira Ushiyama
- Department of Environmental Health, National Institute of Public Health, 2-3-6 Minami, Wako, Saitama, 351-0197, Japan
| | - Machiko Maeda
- Department of Hygienic Chemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Yuki Ogasawara
- Department of Hygienic Chemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Jianqing Wang
- Department of Computer Science and Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya, Aichi, 466-8555, Japan
| | - Naoki Kunugita
- Department of Environmental Health, National Institute of Public Health, 2-3-6 Minami, Wako, Saitama, 351-0197, Japan
| | - Kazuyuki Ishii
- Department of Hygienic Chemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| |
Collapse
|
211
|
Fan Y, Tajima A, Goh SK, Geng X, Gualtierotti G, Grupillo M, Coppola A, Bertera S, Rudert WA, Banerjee I, Bottino R, Trucco M. Bioengineering Thymus Organoids to Restore Thymic Function and Induce Donor-Specific Immune Tolerance to Allografts. Mol Ther 2015; 23:1262-1277. [PMID: 25903472 DOI: 10.1038/mt.2015.77] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 04/05/2015] [Indexed: 02/07/2023] Open
Abstract
One of the major obstacles in organ transplantation is to establish immune tolerance of allografts. Although immunosuppressive drugs can prevent graft rejection to a certain degree, their efficacies are limited, transient, and associated with severe side effects. Induction of thymic central tolerance to allografts remains challenging, largely because of the difficulty of maintaining donor thymic epithelial cells in vitro to allow successful bioengineering. Here, the authors show that three-dimensional scaffolds generated from decellularized mouse thymus can support thymic epithelial cell survival in culture and maintain their unique molecular properties. When transplanted into athymic nude mice, the bioengineered thymus organoids effectively promoted homing of lymphocyte progenitors and supported thymopoiesis. Nude mice transplanted with thymus organoids promptly rejected skin allografts and were able to mount antigen-specific humoral responses against ovalbumin on immunization. Notably, tolerance to skin allografts was achieved by transplanting thymus organoids constructed with either thymic epithelial cells coexpressing both syngeneic and allogenic major histocompatibility complexes, or mixtures of donor and recipient thymic epithelial cells. Our results demonstrate the technical feasibility of restoring thymic function with bioengineered thymus organoids and highlight the clinical implications of this thymus reconstruction technique in organ transplantation and regenerative medicine.
Collapse
Affiliation(s)
- Yong Fan
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Asako Tajima
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Saik Kia Goh
- Department of Chemical and Petroleum Engineering, University of Pittsburgh School of Engineering, Pittsburgh, Pennsylvania, USA
| | - Xuehui Geng
- Division of Immunogenetics, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Giulio Gualtierotti
- Division of Immunogenetics, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Maria Grupillo
- Division of Immunogenetics, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Antonina Coppola
- Division of Immunogenetics, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Current address: Section of Endocrinology, Dipartimento Biomedico di Medicina Interna e Specialistica (DIBIMIS), University of Palermo, Palermo, Italy
| | - Suzanne Bertera
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - William A Rudert
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Ipsita Banerjee
- Department of Chemical and Petroleum Engineering, University of Pittsburgh School of Engineering, Pittsburgh, Pennsylvania, USA
| | - Rita Bottino
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Massimo Trucco
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
212
|
Kim MJ, Miller CM, Shadrach JL, Wagers AJ, Serwold T. Young, proliferative thymic epithelial cells engraft and function in aging thymuses. THE JOURNAL OF IMMUNOLOGY 2015; 194:4784-95. [PMID: 25870244 DOI: 10.4049/jimmunol.1403158] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/09/2015] [Indexed: 01/17/2023]
Abstract
The thymus reaches its maximum size early in life and then begins to shrink, producing fewer T cells with increasing age. This thymic decline is thought to contribute to age-related T cell lymphopenias and hinder T cell recovery after bone marrow transplantation. Although several cellular and molecular processes have been implicated in age-related thymic involution, their relative contributions are not known. Using heterochronic parabiosis, we observe that young circulating factors are not sufficient to drive regeneration of the aged thymus. In contrast, we find that resupplying young, engraftable thymic epithelial cells (TECs) to a middle-aged or defective thymus leads to thymic growth and increased T cell production. Intrathymic transplantation and in vitro colony-forming assays reveal that the engraftment and proliferative capacities of TECs diminish early in life, whereas the receptivity of the thymus to TEC engraftment remains relatively constant with age. These results support a model in which thymic growth and subsequent involution are driven by cell-intrinsic changes in the proliferative capacity of TECs, and further show that young TECs can engraft and directly drive the growth of involuted thymuses.
Collapse
Affiliation(s)
- Mi-Jeong Kim
- Joslin Diabetes Center, Boston, MA 02215; Harvard Stem Cell Institute, Cambridge, MA 02138
| | - Christine M Miller
- Joslin Diabetes Center, Boston, MA 02215; Harvard Stem Cell Institute, Cambridge, MA 02138; Howard Hughes Medical Institute, Cambridge, MA 02138; and Department of Stem Cell and Regenerative Biology, Paul F. Glenn Laboratories for the Biological Mechanisms of Aging, Harvard Medical School, Harvard University, Cambridge, MA 02138
| | - Jennifer L Shadrach
- Joslin Diabetes Center, Boston, MA 02215; Harvard Stem Cell Institute, Cambridge, MA 02138; Howard Hughes Medical Institute, Cambridge, MA 02138; and Department of Stem Cell and Regenerative Biology, Paul F. Glenn Laboratories for the Biological Mechanisms of Aging, Harvard Medical School, Harvard University, Cambridge, MA 02138
| | - Amy J Wagers
- Joslin Diabetes Center, Boston, MA 02215; Harvard Stem Cell Institute, Cambridge, MA 02138; Howard Hughes Medical Institute, Cambridge, MA 02138; and Department of Stem Cell and Regenerative Biology, Paul F. Glenn Laboratories for the Biological Mechanisms of Aging, Harvard Medical School, Harvard University, Cambridge, MA 02138
| | - Thomas Serwold
- Joslin Diabetes Center, Boston, MA 02215; Harvard Stem Cell Institute, Cambridge, MA 02138;
| |
Collapse
|
213
|
Klose CSN, Diefenbach A. Transcription factors controlling innate lymphoid cell fate decisions. Curr Top Microbiol Immunol 2015; 381:215-55. [PMID: 25038936 DOI: 10.1007/82_2014_381] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mucosal epithelium is in direct contact with symbiotic and pathogenic microorganisms. Therefore, the mucosal surface is the principal portal of entry for invading pathogens and immune cells accumulated in the intestine to prevent infections. In addition to these conventional immune system functions, it has become clear that immune cells during steady-state continuously integrate microbial and nutrient-derived signals from the environment to support organ homeostasis. A major role in both processes is played by a recently discovered group of lymphocytes referred to as innate lymphoid cells (ILCs) Innate lymphoid cells (ILCs) that are specifically enriched at mucosal surfaces but are rather rare in secondary lymphoid organs. In analogy to the dichotomy between CD8 and CD4 T cells, we propose to classify ILCs into interleukin-7 receptor α-negative cytotoxic ILCs and IL-7Rα(+) helper-like ILCs. Dysregulated immune responses triggered by the various ILC subsets have been linked to inflammatory diseases such as inflammatory bowel disease, atopic dermatitis and airway hyperresponsiveness. Here, we will review recent progress in determining the transcriptional and developmental programs that control ILC fate decisions.
Collapse
Affiliation(s)
- Christoph S N Klose
- Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, 55131, Mainz, Germany
| | | |
Collapse
|
214
|
Allan DSJ, Kirkham CL, Aguilar OA, Qu LC, Chen P, Fine JH, Serra P, Awong G, Gommerman JL, Zúñiga-Pflücker JC, Carlyle JR. An in vitro model of innate lymphoid cell function and differentiation. Mucosal Immunol 2015; 8:340-51. [PMID: 25138665 DOI: 10.1038/mi.2014.71] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/28/2014] [Indexed: 02/04/2023]
Abstract
Innate lymphoid cells (ILC) are RAG-independent lymphocytes with important roles in innate immunity, and include group-1 (natural killer (NK) cell, ILC1), group-2 (ILC2), and group-3 (lymphoid tissue inducer (LTi), NCR(+) ILC3) subsets. Group-3 ILC express Rorγt, produce interleukin (IL)-22, and are critically important in the normal function of mucosal tissues. Here, we describe a novel model cell line for the study of ILC function and differentiation. The parental MNK cell line, derived from NKR-P1B(+) fetal thymocytes, shows a capacity to differentiate in γc cytokines. One IL-7-responsive subline, designated MNK-3, expresses Rorγt and produces high levels of IL-22 in response to IL-23 and IL-1β stimulation. MNK-3 cells display surface markers and transcript expression characteristic of group-3 ILC, including IL-7Rα (CD127), c-kit (CD117), CCR6, Thy1 (CD90), RANK, RANKL, and lymphotoxin (LTα1β2). Using an in vitro assay of LTi cell activity, MNK-3 cells induce ICAM-1 and VCAM-1 expression on stromal cells in a manner dependent upon LTα1β2 expression. A second IL-2-responsive subline, MNK-1, expresses several NK cell receptors, perforin and granzymes, and shows some cytotoxic activity. Thus, MNK-1 cells serve as a model of ILC1/NK development and differentiation, whereas MNK-3 cells provide an attractive in vitro system to study the function of ILC3/LTi cells.
Collapse
Affiliation(s)
- D S J Allan
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - C L Kirkham
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - O A Aguilar
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - L C Qu
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - P Chen
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - J H Fine
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - P Serra
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - G Awong
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - J L Gommerman
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - J C Zúñiga-Pflücker
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - J R Carlyle
- 1] Department of Immunology, University of Toronto, Toronto, Ontario, Canada [2] Sunnybrook Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
215
|
Abstract
Interleukin-22 (IL-22) is a recently described IL-10 family cytokine that is produced by T helper (Th) 17 cells, γδ T cells, NKT cells, and newly described innate lymphoid cells (ILCs). Knowledge of IL-22 biology has evolved rapidly since its discovery in 2000, and a role for IL-22 has been identified in numerous tissues, including the intestines, lung, liver, kidney, thymus, pancreas, and skin. IL-22 primarily targets nonhematopoietic epithelial and stromal cells, where it can promote proliferation and play a role in tissue regeneration. In addition, IL-22 regulates host defense at barrier surfaces. However, IL-22 has also been linked to several conditions involving inflammatory tissue pathology. In this review, we assess the current understanding of this cytokine, including its physiologic and pathologic effects on epithelial cell function.
Collapse
|
216
|
Abstract
The lymphocyte family has expanded significantly in recent years to include not only the adaptive lymphocytes (T cells, B cells) and NK cells, but also several additional innate lymphoid cell (ILC) types. ILCs lack clonally distributed antigen receptors characteristic of adaptive lymphocytes and instead respond exclusively to signaling via germline-encoded receptors. ILCs resemble T cells more closely than any other leukocyte lineage at the transcriptome level and express many elements of the core T cell transcriptional program, including Notch, Gata3, Tcf7, and Bcl11b. We present our current understanding of the shared and distinct transcriptional regulatory mechanisms involved in the development of adaptive T lymphocytes and closely related ILCs. We discuss the possibility that a core set of transcriptional regulators common to ILCs and T cells establish enhancers that enable implementation of closely aligned effector pathways. Studies of the transcriptional regulation of lymphopoiesis will support the development of novel therapeutic approaches to correct early lymphoid developmental defects and aberrant lymphocyte function.
Collapse
Affiliation(s)
- Maria Elena De Obaldia
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | |
Collapse
|
217
|
Abstract
The innate immune system is composed of a diverse array of evolutionarily ancient haematopoietic cell types, including dendritic cells, monocytes, macrophages and granulocytes. These cell populations collaborate with each other, with the adaptive immune system and with non-haematopoietic cells to promote immunity, inflammation and tissue repair. Innate lymphoid cells are the most recently identified constituents of the innate immune system and have been the focus of intense investigation over the past five years. We summarize the studies that formally identified innate lymphoid cells and highlight their emerging roles in controlling tissue homeostasis in the context of infection, chronic inflammation, metabolic disease and cancer.
Collapse
Affiliation(s)
- David Artis
- Weill Cornell Medical College, Cornell University, New York, New York 10021, USA
| | - Hergen Spits
- Academic Medical Center at the University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| |
Collapse
|
218
|
van den Brink MRM, Velardi E, Perales MA. Immune reconstitution following stem cell transplantation. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2015; 2015:215-219. [PMID: 26637724 DOI: 10.1182/asheducation-2015.1.215] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Affiliation(s)
- Marcel R M van den Brink
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medical College, New York, NY; Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Enrico Velardi
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY; and Division of Pharmacology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medical College, New York, NY
| |
Collapse
|
219
|
Li LJ, Gong C, Zhao MH, Feng BS. Role of interleukin-22 in inflammatory bowel disease. World J Gastroenterol 2014; 20:18177-88. [PMID: 25561785 PMCID: PMC4277955 DOI: 10.3748/wjg.v20.i48.18177] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 06/21/2014] [Accepted: 09/05/2014] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease thought to be mediated by the microbiota of the intestinal lumen and inappropriate immune responses. Aberrant immune responses can cause secretion of harmful cytokines that destroy the epithelium of the gastrointestinal tract, leading to further inflammation. Interleukin (IL)-22 is a member of the IL-10 family of cytokines that was recently discovered to be mainly produced by both adaptive and innate immune cells. Several cytokines and many of the transcriptional factors and T regulatory cells are known to regulate IL-22 expression through activation of signal transducer and activator of transcription 3 signaling cascades. This cytokine induces antimicrobial molecules and proliferative and antiapoptotic pathways, which help prevent tissue damage and aid in its repair. All of these processes play a beneficial role in IBD by enhancing intestinal barrier integrity and epithelial innate immunity. In this review, we discuss recent progress in the involvement of IL-22 in the pathogenesis of IBD, as well as its therapeutic potential.
Collapse
|
220
|
Hülsdünker J, Zeiser R. Insights into the pathogenesis of GvHD: what mice can teach us about man. ACTA ACUST UNITED AC 2014; 85:2-9. [PMID: 25532439 DOI: 10.1111/tan.12497] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Acute graft-vs-host disease (GvHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation (allo-HCT). Most of the knowledge about the biology of GvHD is derived from mouse models of this disease and therefore a critical analysis of potential advantages and disadvantages of the murine GvHD models is important to classify and understand the findings made in these models. The central events leading up to GvHD were characterized in three phases which includes the tissue damage-phase, the T cell priming-phase and the effector-phase, when the disease becomes clinically overt. The role of individual cytokines, chemokines, transcription factor or receptors was studied in these models by using gene deficient or transgenic mice in the donor or recipient compartments. Besides, numerous studies have been performed in these models to prevent or treat GvHD. Several recent clinical trials were all based on previously reported findings from the mouse model of GvHD such as the trials on CCR5-blockade, donor statin treatment, vorinostat treatment or adoptive transfer of regulatory T cells for GvHD prevention. The different mouse models for GvHD and graft-vs-leukemia effects are critically reviewed and their impact on current clinical practice is discussed.
Collapse
Affiliation(s)
- J Hülsdünker
- Department of Hematology, Oncology and Stem Cell Transplantation, Freiburg University Medical Center, Albert Ludwigs University Freiburg, Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
221
|
Rutz S, Wang X, Ouyang W. The IL-20 subfamily of cytokines--from host defence to tissue homeostasis. Nat Rev Immunol 2014; 14:783-95. [PMID: 25421700 DOI: 10.1038/nri3766] [Citation(s) in RCA: 260] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The interleukin-20 (IL-20) subfamily of cytokines comprises IL-19, IL-20, IL-22, IL-24 and IL-26. These cytokines are all members of the larger IL-10 family, but have been grouped together to form the IL-20 subfamily based on their usage of common receptor subunits and similarities in their target-cell profiles and biological functions. Members of the IL-20 subfamily facilitate the communication between leukocytes and epithelial cells, thereby enhancing innate defence mechanisms and tissue repair processes at epithelial surfaces. In this Review, we describe the cellular sources and targets of the IL-20 subfamily cytokines, and we detail how their expression is regulated. Much of our understanding of the unique biology of this group of cytokines is still based on IL-22, which is the most studied member of the IL-20 subfamily. Nevertheless, we attempt a broader discussion of the emerging functions of IL-20 subfamily cytokines in host defence, inflammatory diseases, cancer and metabolism.
Collapse
Affiliation(s)
- Sascha Rutz
- Department of Immunology, Genentech, South San Francisco, California 94080, USA
| | - Xiaoting Wang
- Department of Immunology, Genentech, South San Francisco, California 94080, USA
| | - Wenjun Ouyang
- Department of Immunology, Genentech, South San Francisco, California 94080, USA
| |
Collapse
|
222
|
The role of the thymus in T-cell immune reconstitution after umbilical cord blood transplantation. Blood 2014; 124:3201-11. [PMID: 25287708 DOI: 10.1182/blood-2014-07-589176] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Umbilical cord blood (UCB) is an alternative source of hematopoietic stem cells for patients without HLA-matched adult donors. UCB contains a low number of nucleated cells and mostly naive T cells, resulting in prolonged time to engraftment and lack of transferred T-cell memory. Although the first phase of T-cell reconstitution after UCB transplantation (UCBT) depends on peripheral expansion of transferred T cells, permanent T-cell reconstitution is mediated via a central mechanism, which depends on de novo production of naive T lymphocytes by the recipient's thymus from donor-derived lymphoid-myeloid progenitors (LMPs). Thymopoiesis can be assessed by quantification of recent thymic emigrants, T-cell receptor excision circle levels, and T-cell receptor repertoire diversity. These assays are valuable tools for monitoring posttransplantation thymic recovery, but more importantly they have shown the significant prognostic value of thymic reconstitution for clinical outcomes after UCBT, including opportunistic infections, disease relapse, and overall survival. Strategies to improve thymic entry and differentiation of LMPs and to accelerate recovery of the thymic stromal microenvironment may improve thymic lymphopoiesis. Here, we discuss the mechanisms and clinical implications of thymic recovery and new approaches to improve reconstitution of the T-cell repertoire after UCBT.
Collapse
|
223
|
Zhao K, Zhao D, Huang D, Yin L, Chen C, Pan B, Wu Q, Li Z, Yao Y, Shen E, Zeng L, Xu K. Interleukin-22 aggravates murine acute graft-versus-host disease by expanding effector T cell and reducing regulatory T cell. J Interferon Cytokine Res 2014; 34:707-15. [PMID: 24720737 DOI: 10.1089/jir.2013.0099] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Graft-versus-host disease (GVHD) as a major complication after allogeneic hematopoietic stem cell transplantation is not well prevented now. We have observed that interleukin-22 (IL-22) produced by Th22, Th1, and Th17 cells participated in GVHD development in our previous study. However, the role of IL-22 in GVHD is still ambiguous. The aim of this study was to illuminate the pathological or protective function and the potential mechanism of IL-22 in the GVHD process. In the present study, we found that compared with mice cotransferred with bone marrow and spleen cells (BS mice) without IL-22 administration, more serious tissue damage and higher GVHD clinical score were observed in BS+IL-22 mice. IL-22 administration was a benefit to early recovery of thymus after irradiation-induced injury. Administration of IL-22 could promote Th1 and Tc1 cell expansion in mesenteric lymph nodes but reduce CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cell number. Levels of systemic inflammatory cytokines (IFN-γ and TNF-α) were upregulated, while the level of immunosuppressive cytokine IL-10 was downregulated in recipients with IL-22 injection. In conclusion, IL-22, which exacerbates both local immune responses and systemic inflammation of recipients, plays a pathogenic role in the GVHD process. The potential mechanism of IL-22 in GVHD may attribute to increased alloreactive effector Th1 and Tc1 cells and decreased inhibitory Treg cell.
Collapse
Affiliation(s)
- Kai Zhao
- 1 Laboratory of Transplantation and Immunology, Xuzhou Medical College , Xuzhou, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Fernandes SM, Pires AR, Ferreira C, Foxall RB, Rino J, Santos C, Correia L, Poças J, Veiga-Fernandes H, Sousa AE. Enteric mucosa integrity in the presence of a preserved innate interleukin 22 compartment in HIV type 1-treated individuals. J Infect Dis 2014; 210:630-40. [PMID: 24604817 DOI: 10.1093/infdis/jiu126] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
BACKGROUND Interleukin 22 (IL-22) is emerging as a key cytokine for gut epithelial homeostasis and mucosal repair. Gut disruption is a hallmark of human immunodeficiency virus (HIV) infection. Here, we investigated IL-22 production and gut mucosal integrity in HIV type 1 (HIV-1)-infected individuals receiving long-term antiretroviral therapy (ART). METHODS Biopsy specimens from 37 individuals who underwent colonoscopy primarily for cancer screening and from 17 HIV-1-infected and 20 healthy age-matched controls were assessed. RESULTS We found significant depletion of sigmoid IL-22-producing CD4(+) T cells (T-helper type 22 [Th22] cells) even after prolonged ART, contrasting with the apparently normal compartments of regulatory and interleukin 17 (IL-17)-producing CD4(+) T cells, as well as total mucosal CD4(+) T cells. Despite the preferential Th22 cell depletion, IL-22 production by innate lymphoid cells (ILCs) was similar to that observed in HIV-1-seronegative subjects, and transcription of genes encoding molecules relevant for IL-22 production (ie, AHR, IL23, IL23R, IL1B, IL6, and TGFB1) was preserved. Remarkably, levels of transcripts of IL-22-target genes (ie, REG3G, DEFB4A, S100A9, MUC1, and MUC13) were unaltered, suggesting an adequate production of antimicrobial peptides and mucins. In agreement, enteric epithelial architecture was fully preserved. CONCLUSIONS Despite the reduced Th22 cell subset, innate IL-22-mediated mechanisms, essential for sigmoid mucosa integrity, were fully operational in long-term-treated HIV-1-infected individuals. Our data highlight IL-22 production by ILCs as an important target for therapies aimed at facilitating human mucosal reconstitution.
Collapse
Affiliation(s)
- Susana M Fernandes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa Clínica Universitária de Medicina II
| | - Ana R Pires
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa
| | | | - Russell B Foxall
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa
| | - José Rino
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa
| | | | - Luís Correia
- Clínica Universitária de Gastroenterologia, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte-EPE, Lisboa
| | - José Poças
- Serviço de Infecciologia, Hospital de S. Bernardo, Setúbal, Portugal
| | | | - Ana E Sousa
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa
| |
Collapse
|
225
|
Li H, Hsu HC, Wu Q, Yang P, Li J, Luo B, Oukka M, Steele CH, Cua DJ, Grizzle WE, Mountz JD. IL-23 promotes TCR-mediated negative selection of thymocytes through the upregulation of IL-23 receptor and RORγt. Nat Commun 2014; 5:4259. [PMID: 25001511 PMCID: PMC4136447 DOI: 10.1038/ncomms5259] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 05/30/2014] [Indexed: 01/06/2023] Open
Abstract
Transient thymic involution is frequently found during inflammation, yet the mode of action of inflammatory cytokines is not well defined. Here we report that interleukin-23 (IL-23) production by the thymic dendritic cells (DCs) promotes apoptosis of the CD4hiCD8hi double positive (DP) thymocytes. A deficiency in IL-23 signaling interferes with negative selection in the male Db/H-Y T-cell receptor (TCR) transgenic mice. IL-23 plus TCR signaling results in significant up-regulation of IL-23 receptor (IL-23R) expressed predominantly on CD4hiCD8hiCD3+αβTCR+ DP thymocytes, and leads to RORγt dependent apoptosis. These results extend the action of IL-23 beyond its peripheral effects to a unique role in TCR mediated negative selection including elimination of natural T regulatory cells in the thymus.
Collapse
Affiliation(s)
- Hao Li
- 1] Division of Clinical Immunology & Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA [2] Department Microbiology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Hui-Chen Hsu
- Division of Clinical Immunology & Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Qi Wu
- Division of Clinical Immunology & Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - PingAr Yang
- Division of Clinical Immunology & Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Jun Li
- Division of Clinical Immunology & Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Bao Luo
- Division of Clinical Immunology & Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Mohamed Oukka
- Department of Pediatrics, University of Washington, Seattle, Washington 98195, USA
| | - Claude H Steele
- Division of Pulmonary, Allergy & Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Daniel J Cua
- Merck Research Laboratories, Palo Alto, California 94304, USA
| | - William E Grizzle
- Clinical Pathology & Anatomic Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - John D Mountz
- 1] Division of Clinical Immunology & Rheumatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA [2] Birmingham VA Medical Center, Birmingham, Alabama 35233, USA
| |
Collapse
|
226
|
Complexity of cytokine network regulation of innate lymphoid cells in protective immunity. Cytokine 2014; 70:1-10. [PMID: 24972988 DOI: 10.1016/j.cyto.2014.06.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 06/02/2014] [Indexed: 02/07/2023]
Abstract
The body's surface provides a critical barrier shielding us from various mechanical and pathogenic insults by virtue of the physical protection it provides and the presence of specialized populations of innate lymphoid cells (ILCs) that sense inflammatory signals induced by pathogens. This response plays a central role in the development and activation of early immune responses. While ILCs depend on common γ-chain cytokine signaling for their development, an essential component of the armory of these cells is their capacity to produce defensive cytokines when activated by viruses, microbes and other parasites. In this review, we describe the multiple intrinsic and extrinsic pathways that comprise the cytokine circuitry regulating the development and function of ILC necessary for protective immunity.
Collapse
|
227
|
Abstract
The last 6 decades have seen major advances in the understanding of immunologic diseases, driven by preclinical animal models. Indeed, bone marrow transplantation (BMT) has its genesis in rodent models dating back to the 1950s. Allogeneic BMT and its major complication, graft-versus-host disease (GVHD), represent a paradigm for the translation of preclinical concepts into clinical practice. The appreciation that GVHD can be thought of as a stepwise escalation in immune activation characterized by eventual massive target tissue apoptosis has allowed the design of rational approaches to better manage patients. Here, we describe the pathophysiology of GVHD as defined in preclinical models, focusing on the successes and failures of this research to instruct and translate clinical practice. We also provide a commentary on the limitations of these models so that they may be better appreciated and addressed in future studies. Notable preclinical successes include the definition of modern immune suppression, reductions in conditioning intensity, posttransplant cyclophosphamide, and the promotion of regulatory T-cell reconstitution. New strategies including naïve T-cell depletion, focused cytokine and chemokine inhibition, and the blockade of costimulation now also appear highly promising and very likely to translate into patients in the near future.
Collapse
|
228
|
Lim C, Savan R. The role of the IL-22/IL-22R1 axis in cancer. Cytokine Growth Factor Rev 2014; 25:257-71. [PMID: 24856143 DOI: 10.1016/j.cytogfr.2014.04.005] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 04/29/2014] [Indexed: 12/18/2022]
Abstract
Interleukin-22 (IL-22) is an IL-10 family cytokine produced by T cells and innate lymphoid cells. The IL-22 signaling pathway orchestrates mucosal immune defense and tissue regeneration through pleiotropic effects including pro-survival signaling, cell migration, dysplasia and angiogenesis. While these functions can prevent initial establishment of tumors, they can also be hijacked by aggressive cancers to enhance tumor growth and metastasis. Thus, the role of the IL-22/IL-22R1 axis in cancer is complex and context-specific. Evidence of IL-22 involvement manifests as dysregulation of IL-22 expression and signaling in patients with many common cancers including those of the gut, skin, lung and liver. Unlike other cancer-associated cytokines, IL-22 has restricted tissue specificity as its unique receptor IL-22R1 is exclusively expressed on epithelial and tissue cells, but not immune cells. This makes it an attractive target for therapy as there is potential achieve anti-tumor immunity with fewer side effects. This review summarizes current findings on functions of IL-22 in association with general mechanisms for tumorigenesis as well as specific contributions to particular cancers, and ponders how best to approach further research in the field.
Collapse
Affiliation(s)
- Chrissie Lim
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Ram Savan
- Department of Immunology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
229
|
Bredenkamp N, Nowell CS, Blackburn CC. Regeneration of the aged thymus by a single transcription factor. Development 2014; 141:1627-37. [PMID: 24715454 PMCID: PMC3978836 DOI: 10.1242/dev.103614] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Thymic involution is central to the decline in immune system function that occurs with age. By regenerating the thymus, it may therefore be possible to improve the ability of the aged immune system to respond to novel antigens. Recently, diminished expression of the thymic epithelial cell (TEC)-specific transcription factor Forkhead box N1 (FOXN1) has been implicated as a component of the mechanism regulating age-related involution. The effects of upregulating FOXN1 function in the aged thymus are, however, unknown. Here, we show that forced, TEC-specific upregulation of FOXN1 in the fully involuted thymus of aged mice results in robust thymus regeneration characterized by increased thymopoiesis and increased naive T cell output. We demonstrate that the regenerated organ closely resembles the juvenile thymus in terms of architecture and gene expression profile, and further show that this FOXN1-mediated regeneration stems from an enlarged TEC compartment, rebuilt from progenitor TECs. Collectively, our data establish that upregulation of a single transcription factor can substantially reverse age-related thymic involution, identifying FOXN1 as a specific target for improving thymus function and, thus, immune competence in patients. More widely, they demonstrate that organ regeneration in an aged mammal can be directed by manipulation of a single transcription factor, providing a provocative paradigm that may be of broad impact for regenerative biology.
Collapse
Affiliation(s)
- Nicholas Bredenkamp
- Medical Research Council Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, SCRM Building, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | | | | |
Collapse
|
230
|
Escobar TM, Kanellopoulou C, Kugler DG, Kilaru G, Nguyen CK, Nagarajan V, Bhairavabhotla RK, Northrup D, Zahr R, Burr P, Liu X, Zhao K, Sher A, Jankovic D, Zhu J, Muljo SA. miR-155 activates cytokine gene expression in Th17 cells by regulating the DNA-binding protein Jarid2 to relieve polycomb-mediated repression. Immunity 2014; 40:865-79. [PMID: 24856900 DOI: 10.1016/j.immuni.2014.03.014] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 03/27/2014] [Indexed: 01/02/2023]
Abstract
Specification of the T helper 17 (Th17) cell lineage requires a well-defined set of transcription factors, but how these integrate with posttranscriptional and epigenetic programs to regulate gene expression is poorly understood. Here we found defective Th17 cell cytokine expression in miR-155-deficient CD4+ T cells in vitro and in vivo. Mir155 was bound by Th17 cell transcription factors and was highly expressed during Th17 cell differentiation. miR-155-deficient Th17 and T regulatory (Treg) cells expressed increased amounts of Jarid2, a DNA-binding protein that recruits the Polycomb Repressive Complex 2 (PRC2) to chromatin. PRC2 binding to chromatin and H3K27 histone methylation was increased in miR-155-deficient cells, coinciding with failure to express Il22, Il10, Il9, and Atf3. Defects in Th17 cell cytokine expression and Treg cell homeostasis in the absence of Mir155 could be partially suppressed by Jarid2 deletion. Thus, miR-155 contributes to Th17 cell function by suppressing the inhibitory effects of Jarid2.
Collapse
Affiliation(s)
- Thelma M Escobar
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Chrysi Kanellopoulou
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - David G Kugler
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Gokhul Kilaru
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Cuong K Nguyen
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Vijayaraj Nagarajan
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Ravikiran K Bhairavabhotla
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Daniel Northrup
- Systems Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Rami Zahr
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Patrick Burr
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Xiuhuai Liu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Keji Zhao
- Systems Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Alan Sher
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Dragana Jankovic
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Jinfang Zhu
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Stefan A Muljo
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|
231
|
Kryczek I, Lin Y, Nagarsheth N, Peng D, Zhao L, Zhao E, Vatan L, Szeliga W, Dou Y, Owens S, Zgodzinski W, Majewski M, Wallner G, Fang J, Huang E, Zou W. IL-22(+)CD4(+) T cells promote colorectal cancer stemness via STAT3 transcription factor activation and induction of the methyltransferase DOT1L. Immunity 2014; 40:772-784. [PMID: 24816405 PMCID: PMC4032366 DOI: 10.1016/j.immuni.2014.03.010] [Citation(s) in RCA: 296] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 03/07/2014] [Indexed: 12/12/2022]
Abstract
Little is known about how the immune system impacts human colorectal cancer invasiveness and stemness. Here we detected interleukin-22 (IL-22) in patient colorectal cancer tissues that was produced predominantly by CD4(+) T cells. In a mouse model, migration of these cells into the colon cancer microenvironment required the chemokine receptor CCR6 and its ligand CCL20. IL-22 acted on cancer cells to promote activation of the transcription factor STAT3 and expression of the histone 3 lysine 79 (H3K79) methytransferase DOT1L. The DOT1L complex induced the core stem cell genes NANOG, SOX2, and Pou5F1, resulting in increased cancer stemness and tumorigenic potential. Furthermore, high DOT1L expression and H3K79me2 in colorectal cancer tissues was a predictor of poor patient survival. Thus, IL-22(+) cells promote colon cancer stemness via regulation of stemness genes that negatively affects patient outcome. Efforts to target this network might be a strategy in treating colorectal cancer patients.
Collapse
Affiliation(s)
- Ilona Kryczek
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Yanwei Lin
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai 200001, China
| | - Nisha Nagarsheth
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Graduate Programs in Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Dongjun Peng
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Ende Zhao
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Wojciech Szeliga
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Yali Dou
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Scott Owens
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Witold Zgodzinski
- The Second Department of General Surgery, Medical University in Lublin, Lublin 20-081, Poland
| | - Marek Majewski
- The Second Department of General Surgery, Medical University in Lublin, Lublin 20-081, Poland
| | - Grzegorz Wallner
- The Second Department of General Surgery, Medical University in Lublin, Lublin 20-081, Poland
| | - Jingyuan Fang
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai 200001, China
| | - Emina Huang
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Graduate Programs in Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Tumor Biology, Univxexrsity of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- The University of Michigan Comprehensive Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| |
Collapse
|
232
|
De Barros SC, Zimmermann VS, Taylor N. Concise review: hematopoietic stem cell transplantation: targeting the thymus. Stem Cells 2014; 31:1245-51. [PMID: 23554173 DOI: 10.1002/stem.1378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/15/2013] [Indexed: 12/28/2022]
Abstract
Allogeneic hematopoietic stem cell (HSC) transplantation can cure patients suffering from diverse genetic and acquired diseases as well as cancers. Nevertheless, under conditions where T-cell reconstitution is critical, the entry of donor progenitors into the thymus remains a major bottleneck. It is assumed that following the intravenous injection of HSC, they first home to the BM. More committed progenitors can then be exported to the thymus in response to a myriad of signals regulating thymus seeding. Notably although, the thymus is not continually receptive to the import of hematopoietic progenitors. Furthermore, as stem cells with self-renewing capacity do not take up residence in the thymus under physiological conditions, the periodic colonization of the thymus is essential for the sustained differentiation of T lymphocytes. As such, we and others have invested significant efforts into exploring avenues that might foster a long-term thymus-autonomous differentiation. Here, we review strategic approaches that have resulted in long-term T-cell differentiation in immunodeficient (SCID) mice, even across histocompatibility barriers. These include the forced thymic entry of BM precursors by their direct intrathymic injection as well as the transplantation of neonatal thymi. The capacity of the thymus to support hematopoietic progenitors with renewal potential will hopefully promote the development of new therapeutic strategies aimed at enhancing T-cell differentiation in patients undergoing HSC transplantation.
Collapse
Affiliation(s)
- Stéphanie C De Barros
- Institut de Génétique Moléculaire de Montpellier, Université Montpellier , Montpellier, France
| | | | | |
Collapse
|
233
|
Yeste A, Mascanfroni ID, Nadeau M, Burns EJ, Tukpah AM, Santiago A, Wu C, Patel B, Kumar D, Quintana FJ. IL-21 induces IL-22 production in CD4+ T cells. Nat Commun 2014; 5:3753. [PMID: 24796415 PMCID: PMC4157605 DOI: 10.1038/ncomms4753] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 03/28/2014] [Indexed: 02/06/2023] Open
Abstract
Interleukin (IL)-22 produced by innate lymphoid cells (ILCs) and CD4+ T cells plays an important role in host defence and mucosal homeostasis, thus it is important to investigate the mechanisms that regulate IL-22 production. We investigated the regulation IL-22 production by CD4+ T cells. Here we show that IL-21 triggers IL-22, but not IL-17 production by CD4+ T cells. STAT3, activated by IL-21, controls the epigenetic status of the il22 promoter and its interaction with the aryl hydrocarbon receptor (AhR). Moreover, IL-21 and AhR signalling in T cells control IL-22 production and the development of dextran sodium sulphate-induced colitis in ILC-deficient mice. Thus, we have identified IL-21 as an inducer of IL-22 production in CD4+ T cells in vitro and in vivo.
Collapse
Affiliation(s)
- Ada Yeste
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Ivan D. Mascanfroni
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Meghan Nadeau
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Evan J. Burns
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Ann-Marcia Tukpah
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Andrezza Santiago
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Chuan Wu
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Bonny Patel
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Deepak Kumar
- Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston 02115, MA, USA
| | - Francisco J. Quintana
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
234
|
Johnston HF, Xu Y, Racine JJ, Cassady K, Ni X, Wu T, Chan A, Forman S, Zeng D. Administration of anti-CD20 mAb is highly effective in preventing but ineffective in treating chronic graft-versus-host disease while preserving strong graft-versus-leukemia effects. Biol Blood Marrow Transplant 2014; 20:1089-103. [PMID: 24796279 DOI: 10.1016/j.bbmt.2014.04.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 04/25/2014] [Indexed: 12/15/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) is an autoimmune-like syndrome, and donor B cells play important roles in augmenting its pathogenesis. B cell-depleting anti-CD20 mAb has been administered before or after cGVHD onset for preventing or treating cGVHD in the clinic. Although administration before onset appeared to be more effective, the effect is variable and sometimes minimal. Here, we used 2 mouse cGVHD models to evaluate the preventive and therapeutic effect of anti-CD20 mAb. With the model of DBA/2 donor to MHC-matched BALB/c recipient, 1 intravenous injection of anti-CD20 mAb (40 mg/kg) the following day or on day 7 after hematopoietic cell transplantation when serum autoantibodies were undetectable effectively prevented induction of cGVHD and preserved a strong graft-versus-leukemia (GVL) effect. The separation of GVL effect from GVHD was associated with a significant reduction of donor CD4(+) T cell proliferation and expansion and protection of host thymic medullary epithelial cells. Anti-CD20 mAb administration also prevented expansion of donor T cells and induction of cGVHD in another mouse model of C57BL/6 donor to MHC-mismatched BALB/c recipients. In contrast, administration of anti-CD20 mAb after GVHD onset was not able to effectively deplete donor B cells or ameliorate cGVHD in either model. These results indicate that administration of anti-CD20 mAb before signs of cGVHD can prevent induction of autoimmune-like cGVHD while preserving a GVL effect; there is little effect if administered after cGVHD onset. This provides new insights into clinical prevention and therapy of cGVHD with B cell-depleting reagents.
Collapse
Affiliation(s)
- Heather F Johnston
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Yajing Xu
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Jeremy J Racine
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Kaniel Cassady
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Xiong Ni
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Department of Hematology, Changhai Hospital, The Second Military Medical School, Shanghai, China
| | - Tao Wu
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Department of Hematology, Changhai Hospital, The Second Military Medical School, Shanghai, China
| | - Andrew Chan
- Department of Research Biology, Genentech, San Francisco, California
| | - Stephen Forman
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute at City of Hope National Medical Center, Duarte, California
| | - Defu Zeng
- Departments of Diabetes Research and Hematology/Hematopoietic Cell Transplantation, Beckman Research Institute at City of Hope National Medical Center, Duarte, California; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute at City of Hope National Medical Center, Duarte, California.
| |
Collapse
|
235
|
Kulkarni OP, Hartter I, Mulay SR, Hagemann J, Darisipudi MN, Kumar Vr S, Romoli S, Thomasova D, Ryu M, Kobold S, Anders HJ. Toll-like receptor 4-induced IL-22 accelerates kidney regeneration. J Am Soc Nephrol 2014; 25:978-89. [PMID: 24459235 PMCID: PMC4005301 DOI: 10.1681/asn.2013050528] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 12/11/2013] [Indexed: 12/14/2022] Open
Abstract
AKI involves early Toll-like receptor (TLR)-driven immunopathology, and resolution of inflammation is needed for rapid regeneration of injured tubule cells. Notably, activation of TLRs also has been implicated in epithelial repair. We hypothesized that TLR signaling drives tubule regeneration after acute injury through the induction of certain ILs. Systematic screening in vitro identified IL-22 as a candidate proregeneratory factor in primary tubular cell recovery, and IL-22 deficiency or IL-22 blockade impaired post-ischemic tubular recovery after AKI in mice. Interstitial mononuclear cells, such as dendritic cells and macrophages, were the predominant source of IL-22 secretion, whereas IL-22 receptor was expressed by tubular epithelial cells exclusively. Depleting IL-22-producing cells during the healing phase impaired epithelial recovery, which could be rescued entirely by reconstituting mice with IL-22. In vitro, necrotic tubular cells and oxidative stress induced IL-22 secretion selectively through TLR4. Although TLR4 blockade during the early injury phase prevented tubular necrosis and AKI, TLR4 blockade during the healing phase suppressed IL-22 production and impaired kidney regeneration. Taken together, these results suggest that necrotic cell-derived TLR4 agonists activate intrarenal mononuclear cells to secrete IL-22, which accelerates tubular regeneration and recovery in AKI.
Collapse
Affiliation(s)
- Onkar P Kulkarni
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany; and
| | - Ingo Hartter
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany; and
| | - Shrikant R Mulay
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany; and
| | - Jan Hagemann
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany; and
| | - Murthy N Darisipudi
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany; and
| | - Santhosh Kumar Vr
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany; and
| | - Simone Romoli
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany; and
| | - Dana Thomasova
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany; and
| | - Mi Ryu
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany; and
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich and Division of Clinical Pharmacology, Department of Internal Medicine IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Munich, Germany; and
| |
Collapse
|
236
|
Anders HJ. Immune system modulation of kidney regeneration--mechanisms and implications. Nat Rev Nephrol 2014; 10:347-58. [PMID: 24776845 DOI: 10.1038/nrneph.2014.68] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The immune system is an important guardian of tissue homeostasis. In response to injury, resident and infiltrating immune cells orchestrate all phases of danger control, resolution of inflammation and tissue regeneration or scar formation. As mammalian postnatal kidneys are not capable of de novo nephrogenesis, recovery is limited to the regeneration or repair of existing nephrons. The regenerative capacity of the nephron varies between compartments; the epithelial cells of the tubule regenerate more efficiently than the structurally highly organized podocytes. Cells of the surrounding environment modulate nephron regeneration by secreting paracrine mediators. This Review discusses immune mediators and pathways that regulate the intrinsic regenerative capacity of the nephron. Eliminating injurious triggers, modulating renal inflammation and specifically enhancing the regenerative capacity of nephrons might be a promising strategy to improve long-term outcomes in patients with acute kidney injury and/or chronic kidney disease.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München-Innenstadt, Ziemssenstrasse 1, 80336 Munich, Germany
| |
Collapse
|
237
|
Abstract
Innate lymphoid cells (ILCs) are lymphoid cells that do not express rearranged receptors and have important effector and regulatory functions in innate immunity and tissue remodeling. ILCs are categorized into 3 groups based on their distinct patterns of cytokine production and the requirement of particular transcription factors for their development and function. Group 1 ILCs (ILC1s) produce interferon γ and depend on Tbet, group 2 ILCs (ILC2s) produce type 2 cytokines like interleukin-5 (IL-5) and IL-13 and require GATA3, and group 3 ILCs (ILC3s) include lymphoid tissue inducer cells, produce IL-17 and/or IL-22, and are dependent on RORγt. Whereas ILCs play essential roles in the innate immune system, uncontrolled activation and proliferation of ILCs can contribute to inflammatory autoimmune diseases. In this review, we provide an overview of the characteristics of ILCs in the context of health and disease. We will focus on human ILCs but refer to mouse studies if needed to clarify aspects of ILC biology.
Collapse
|
238
|
Image-guided intrathymic injection of multipotent stem cells supports lifelong T-cell immunity and facilitates targeted immunotherapy. Blood 2014; 123:2797-805. [PMID: 24652996 DOI: 10.1182/blood-2013-10-535401] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
T-cell deficiency related to disease, medical treatment, or aging represents a major clinical challenge and is associated with significant morbidity and mortality in cancer and bone marrow transplantation recipients. This study describes several innovative and clinically relevant strategies to manipulate thymic function based on an interventional radiology technique for intrathymic injection of cells or drugs. We show that intrathymic injection of multipotent hematopoietic stem/progenitor cells into irradiated syngeneic or allogeneic young or aged recipients resulted in efficient and long-lasting generation of functional donor T cells. Persistence of intrathymic donor cells was associated with intrathymic presence of cells resembling long-term hematopoietic stem cells, suggesting a self-renewal capacity of the intrathymically injected cells. Furthermore, our approach enabled the induction of long-term antigen-specific T-cell-mediated antitumor immunity following intrathymic injection of progenitor cells harboring a transgenic T-cell receptor gene. The intrathymic injection of interleukin-7 prior to irradiation conferred radioprotection. In addition, thymopoiesis of aged mice improved with a single intrathymic administration of low-dose keratinocyte growth factor, an effect that was sustained even in the setting of radiation-induced injury. Taken together, we established a preclinical framework for the development of novel clinical protocols to establish lifelong antigen-specific T-cell immunity.
Collapse
|
239
|
Tanriver Y, Diefenbach A. Transcription factors controlling development and function of innate lymphoid cells. Int Immunol 2014; 26:119-28. [DOI: 10.1093/intimm/dxt063] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
240
|
Eidenschenk C, Rutz S, Liesenfeld O, Ouyang W. Role of IL-22 in microbial host defense. Curr Top Microbiol Immunol 2014; 380:213-36. [PMID: 25004820 DOI: 10.1007/978-3-662-43492-5_10] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Interleukin (IL)-22 is a member of the IL-10 family of cytokines, which, besides IL-10, contains seven additional cytokines. Although the founding member IL-10 is an important immunoregulatory cytokine that represses both innate and adaptive immunity, the other family members preferentially target epithelial cells and enhance innate host defense mechanisms against various pathogens such as bacteria, yeast, and viruses. Based on their functions, the IL-10 family can be further divided into three subgroups, IL-10 itself, the IL-20 subfamily, and the IFNλ subfamily. IL-22 is the best-studied member of the IL-20 subfamily, and exemplifies the diverse biological effects of this subfamily. IL-22 elicits various innate immune responses from epithelial cells and is essential for host defense against several invading pathogens, including Citrobacter rodentium and Klebsiella pneumonia. IL-22 also protects tissue integrity and maintains the mucosal homeostasis. On the other hand, IL-22 is a proinflammatory cytokine with the capacity to amplify inflammatory responses, which might result in tissue damage, e.g., the IL-22-dependent necrosis of the small intestine during Toxoplasma gondii infection.
Collapse
Affiliation(s)
- Celine Eidenschenk
- Department of Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA,
| | | | | | | |
Collapse
|
241
|
Pan B, Liu J, Zhang Y, Sun Y, Wu Q, Zhao K, Zeng L, Xu K. Acute ablation of DP thymocytes induces up-regulation of IL-22 and Foxn1 in TECs. Clin Immunol 2014; 150:101-8. [PMID: 24333537 DOI: 10.1016/j.clim.2013.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 10/18/2013] [Accepted: 11/01/2013] [Indexed: 12/20/2022]
Abstract
Thymic epithelial cells (TECs) provide the basic architecture for the development of thymocytes. TEC is regenerative after impairment in thymus of young mice. However, how this regeneration program is governed remains unclear. Transcription factor Foxn1 is a central mediator of the differentiation and function of TEC. We examined the relation between thymic injury and Foxn1 in TEC. Total body irradiation (TBI) treatments induced up-regulation of Foxn1 in TEC, which was abolished when thymic function recovered. Specific depletion of double positive (DP) thymocytes triggered the up-regulation of Foxn1. On the other hand, extracellular IL-22 is a potential regulator of homeostasis of TEC. We demonstrated that TBI treatments also induced the up-regulation of intrathymic IL-22. Expression pattern of Foxn1 shares similar characteristics with IL-22. Furthermore, Foxn1 related genes that regulate the function of TEC were also up-regulated. Thus, our data reveal that TBI treatment triggers regeneration program of TEC.
Collapse
Affiliation(s)
- Bin Pan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China
| | - Jie Liu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China
| | - Ying Zhang
- Laboratory of Transplant Immunology, Xuzhou Medical College, Xuzhou, China
| | - Yuqi Sun
- Laboratory of Transplant Immunology, Xuzhou Medical College, Xuzhou, China
| | - Qingyun Wu
- Laboratory of Transplant Immunology, Xuzhou Medical College, Xuzhou, China
| | - Kai Zhao
- Laboratory of Transplant Immunology, Xuzhou Medical College, Xuzhou, China
| | - Lingyu Zeng
- Laboratory of Transplant Immunology, Xuzhou Medical College, Xuzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China; Laboratory of Transplant Immunology, Xuzhou Medical College, Xuzhou, China.
| |
Collapse
|
242
|
Wang X, Ouyang W. Interleukin-22: A Bridge Between Epithelial Innate Host Defense and Immune Cells. CYTOKINE FRONTIERS 2014. [PMCID: PMC7120444 DOI: 10.1007/978-4-431-54442-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Interleukin-22 (IL-22), an IL-10 family cytokine, is produced by various leukocytes. The receptor of IL-22, however, is preferentially detected on peripheral tissue epithelial cells. IL-22 functions as a unique messenger from immune system to tissue epithelial cells and to regulate homeostasis of epithelia. IL-22 is able to directly enhance antimicrobial defense mechanisms in epithelial cells and to facilitate epithelial barrier repair and wound healing process. It, therefore, possesses an irreplaceable role in host defense against certain pathogens that specifically invade epithelial cells. In addition, IL-22 can help to preserve the integrity and homeostasis of various epithelial organs during infection or inflammation. The importance of its tissue-protective function is manifested in many inflammatory situations such as inflammatory bowel diseases (IBD) and hepatitis. On the other hand, as a cytokine, IL-22 is capable of induction of proinflammatory responses, especially in synergy with other cytokines. Consequently, IL-22 contributes to pathogenesis of certain inflammatory diseases for example psoriasis.
Collapse
|
243
|
MacVittie TJ, Bennett AW, V Cohen M, Farese AM, Higgins A, Hankey KG. Immune cell reconstitution after exposure to potentially lethal doses of radiation in the nonhuman primate. HEALTH PHYSICS 2014; 106:84-96. [PMID: 24276552 DOI: 10.1097/hp.0b013e3182a2a9b2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Delayed immune reconstitution remains a major cause of morbidity associated with myelosuppression induced by cytotoxic therapy or myeloablative conditioning for stem cell transplant, as well as potentially lethal doses of total- or partial-body irradiation. Restoration of a functional immune cell repertoire requires hematopoietic stem cell reconstitution for all immune cells and effective thymopoiesis for T cell recovery. There are no medical countermeasures available to mitigate damage consequent to high-dose, potentially lethal irradiation, and there are no well characterized large animal models of prolonged immunosuppression to assess efficacy of potential countermeasures. Herein, the authors describe a model of T and B cell reconstitution following lethal doses of partial-body irradiation with 5% bone marrow sparing that includes full exposure of the thymus. Rhesus macaques (n = 31 male, 5.5-11.3 kg body weight) were exposed to midline tissue doses of 9.0-12.0 Gy using 6 MV LINAC-derived photons at a dose rate of 0.80 Gy min, sparing approximately 5% of bone marrow (tibiae, ankles, and feet). All animals received medical management and were monitored for myeloid and lymphoid suppression and recovery through 180 d post-exposure. Myeloid recovery was assessed by neutrophil and platelet-related hematological parameters. Reconstitution of B and T cell subsets was assessed by flow cytometric immunophenotyping, and recent thymic emigrants were identified by RT-PCR of T cell receptor excision circles. Mortality was recorded through 180 d post-exposure. Acute myelo-suppression was characterized by severe neutropenia and thrombocytopenia, followed by recovery 30-60 d post-exposure. Total T (CD3+) and B (CD20+) cells were reduced significantly following exposure and exhibited differential recovery patterns post-exposure. Both CD4+ and CD8+ subsets of naïve T cells and total CD4+ T cell counts remained significantly lower than baseline through 180 d post-exposure. The failure of recent thymic emigrants and naïve T cell subsets to recover to normal baseline values reflects the severe radiation effects on the recovery of marrow-derived stem and early thymic progenitor cells, their mobilization and seeding of receptive thymic niches, and slow endogenous thymic regeneration.
Collapse
Affiliation(s)
- Thomas J MacVittie
- *University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD; †Integrated Research Facility, Frederick, MD; ‡Naval Medical Research Center, Silver Spring, MD
| | | | | | | | | | | |
Collapse
|
244
|
Kumar V. Innate lymphoid cells: New paradigm in immunology of inflammation. Immunol Lett 2014; 157:23-37. [DOI: 10.1016/j.imlet.2013.11.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 10/20/2013] [Accepted: 11/04/2013] [Indexed: 12/27/2022]
|
245
|
Abstract
Interleukin-22 (IL-22) is a key effector molecule that is produced by activated T cells, including T helper 22 (TH22) cells, TH17 cells and TH1 cells, as well as subsets of innate lymphoid cells. Although IL-22 can act synergistically with IL-17 or tumour necrosis factor, some important functions of IL-22 are unique to this cytokine. Data obtained over the past few years indicate that the IL-22-IL-22 receptor subunit 1 (IL-22R1) system has a high potential clinical relevance in psoriasis, ulcerative colitis, graft-versus-host disease, certain infections and tumours, as well as in liver and pancreas damage. This Review highlights current knowledge of the biology of the IL-22-IL-22R1 system, its role in inflammation, tissue protection, regeneration and antimicrobial defence, as well as the positive and potentially negative consequences of its therapeutic modulation.
Collapse
Affiliation(s)
- Robert Sabat
- 1] Interdisciplinary Group of Molecular Immunopathology, Institute of Medical Immunology, Department of Dermatology and Allergy, University Medicine Charité, Charitéplatz 1, D-10117 Berlin, Germany. [2] Research Center Immunosciences, University Hospital Charité, Hessische Strasse 3-4, D-10115 Berlin, Germany
| | - Wenjun Ouyang
- Department of Immunology, Genentech, 1 DNA Way, South San Francisco, California 94080, USA
| | - Kerstin Wolk
- 1] Interdisciplinary Group of Molecular Immunopathology, Institute of Medical Immunology, Department of Dermatology and Allergy, University Medicine Charité, Charitéplatz 1, D-10117 Berlin, Germany. [2] Research Center Immunosciences, University Hospital Charité, Hessische Strasse 3-4, D-10115 Berlin, Germany
| |
Collapse
|
246
|
Martin JCJ, Bériou G, Heslan M, Chauvin C, Utriainen L, Aumeunier A, Scott CL, Mowat A, Cerovic V, Houston SA, Leboeuf M, Hubert FX, Hémont C, Merad M, Milling S, Josien R. Interleukin-22 binding protein (IL-22BP) is constitutively expressed by a subset of conventional dendritic cells and is strongly induced by retinoic acid. Mucosal Immunol 2014; 7:101-13. [PMID: 23653115 PMCID: PMC4291114 DOI: 10.1038/mi.2013.28] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 04/08/2013] [Indexed: 02/04/2023]
Abstract
Interleukin-22 (IL-22) is mainly produced at barrier surfaces by T cells and innate lymphoid cells and is crucial to maintain epithelial integrity. However, dysregulated IL-22 action leads to deleterious inflammation and is involved in diseases such as psoriasis, intestinal inflammation, and cancer. IL-22 binding protein (IL-22BP) is a soluble inhibitory IL-22 receptor and may represent a crucial regulator of IL-22. We show both in rats and mice that, in the steady state, the main source of IL-22BP is constituted by a subset of conventional dendritic cells (DCs) in lymphoid and non-lymphoid tissues. In mouse intestine, IL-22BP was specifically expressed in lamina propria CD103(+)CD11b(+) DC. In humans, IL-22BP was expressed in immature monocyte-derived DC and strongly induced by retinoic acid but dramatically reduced upon maturation. Our data suggest that a subset of immature DCs may actively participate in the regulation of IL-22 activity in the gut by producing high levels of IL-22BP.
Collapse
Affiliation(s)
- JCJ Martin
- INSERM Center of Research in Transplantation and Immunology, UMR1064, Nantes, F - 44000, France,CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, F-44000, France,CHU Nantes, Laboratoire d’immunologie, Nantes, F-44000, France,Université de Nantes, Faculté de Médecine, Nantes, F-44000, France
| | - G Bériou
- INSERM Center of Research in Transplantation and Immunology, UMR1064, Nantes, F - 44000, France,CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, F-44000, France
| | - M Heslan
- INSERM Center of Research in Transplantation and Immunology, UMR1064, Nantes, F - 44000, France,CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, F-44000, France
| | - C Chauvin
- INSERM Center of Research in Transplantation and Immunology, UMR1064, Nantes, F - 44000, France,CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, F-44000, France
| | - L Utriainen
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - A Aumeunier
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - CL Scott
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - A Mowat
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - V Cerovic
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - SA Houston
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - M Leboeuf
- Department of Gene and Cell medicine and the Department of Medicine, Mount Sinai School of Medicine, New York 10029, USA
| | - FX Hubert
- INSERM Center of Research in Transplantation and Immunology, UMR1064, Nantes, F - 44000, France,CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, F-44000, France,Université de Nantes, Faculté de Médecine, Nantes, F-44000, France
| | - C Hémont
- INSERM Center of Research in Transplantation and Immunology, UMR1064, Nantes, F - 44000, France,CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, F-44000, France,CHU Nantes, Laboratoire d’immunologie, Nantes, F-44000, France,Université de Nantes, Faculté de Médecine, Nantes, F-44000, France
| | - M Merad
- Department of Gene and Cell medicine and the Department of Medicine, Mount Sinai School of Medicine, New York 10029, USA
| | - S Milling
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - R Josien
- INSERM Center of Research in Transplantation and Immunology, UMR1064, Nantes, F - 44000, France,CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), Nantes, F-44000, France,CHU Nantes, Laboratoire d’immunologie, Nantes, F-44000, France,Université de Nantes, Faculté de Médecine, Nantes, F-44000, France
| |
Collapse
|
247
|
Zhao K, Zhao D, Huang D, Song X, Chen C, Pan B, Wu Q, Cao J, Yao Y, Zeng L, Xu K. The identification and characteristics of IL-22-producing T cells in acute graft-versus-host disease following allogeneic bone marrow transplantation. Immunobiology 2013; 218:1505-13. [PMID: 23816304 DOI: 10.1016/j.imbio.2013.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 05/11/2013] [Accepted: 05/12/2013] [Indexed: 01/09/2023]
Abstract
Graft-versus-host disease (GVHD) remains the major obstacle for allogeneic bone marrow transplantation, in which many proinflammatory cytokines secreted by alloreactive donor T cells are involved. Role of IL-22 as a member of IL-10 family in GVHD is still disputed and the properties of IL-22-producing cells are unclear. We demonstrated here that CD4⁺ T cells but not CD8⁺ T cells involved in GVHD were the main cellular source of donor-derived IL-22. Th1 and Th17 cells were detected not only express classical cytokine IFN-γ or IL-17, but also contributed to IL-22 secretion in GVHD. Th22 cells characterized by the independent secretion of IL-22 were identified and occupied almost half percentage of IL-22-producing CD4⁺ T cells. The frequency of IL-22-producing CD4⁺ T cells showed dynamic changes with the development of GVHD. Finally, we observed that IL-22-producing CD4⁺ T cells in GVHD mouse carried CD62L⁻CD44(high/low) surface markers. In conclusion, we illuminate the characteristics of donor-derived IL-22-producing CD4⁺ T cells, which may have potent implication for further study of pathogenesis of GVHD.
Collapse
Affiliation(s)
- Kai Zhao
- Laboratory of Transplantation and Immunology, Xuzhou Medical College, No. 99 West Huaihai Road, Xuzhou 221002, Jiangsu, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, No. 99 West Huaihai Road, Xuzhou 221002, Jiangsu, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Thymic epithelial cell expansion through matricellular protein CYR61 boosts progenitor homing and T-cell output. Nat Commun 2013; 4:2842. [DOI: 10.1038/ncomms3842] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 10/29/2013] [Indexed: 12/30/2022] Open
|
249
|
Abstract
Age-related regression of the thymus is associated with a decline in naïve T cell output. This is thought to contribute to the reduction in T cell diversity seen in older individuals and linked with increased susceptibility to infection, autoimmune disease, and cancer. Thymic involution is one of the most dramatic and ubiquitous changes seen in the aging immune system, but the mechanisms which underlying this process are poorly understood. However, a picture is emerging, implicating the involvement of both extrinsic and intrinsic factors. In this review we assess the role of the thymic microenvironment as a potential target that regulates thymic involution, question whether thymocyte development in the aged thymus is functionally impaired, and explore the kinetics of thymic involution.
Collapse
Affiliation(s)
- Donald B Palmer
- Infection and Immunity Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London , London , UK
| |
Collapse
|
250
|
Boehm T, Swann JB. Thymus involution and regeneration: two sides of the same coin? Nat Rev Immunol 2013; 13:831-8. [DOI: 10.1038/nri3534] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|