151
|
Interleukin-7 Availability Is Maintained by a Hematopoietic Cytokine Sink Comprising Innate Lymphoid Cells and T Cells. Immunity 2017; 47:171-182.e4. [DOI: 10.1016/j.immuni.2017.07.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/05/2017] [Accepted: 07/05/2017] [Indexed: 01/09/2023]
|
152
|
Yi P, Liang Y, Yuan DMK, Jie Z, Kwota Z, Chen Y, Cong Y, Fan X, Sun J. A tightly regulated IL-22 response maintains immune functions and homeostasis in systemic viral infection. Sci Rep 2017; 7:3857. [PMID: 28634408 PMCID: PMC5478593 DOI: 10.1038/s41598-017-04260-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/11/2017] [Indexed: 12/19/2022] Open
Abstract
Interleukin-22 (IL-22) plays an important role in host immunity and tissue homeostasis in infectious and inflammatory diseases. However, the function and regulation of IL-22 in viral infection remain largely unknown. Here, we report that viral infection triggered early IL-22 production from the liver and lymphoid organs. γδ T cells are the main immune cells to produce IL-22 in the liver, a process mediated by the IL-23/phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin complex 1 (mTORC1) signaling pathway. In the presence of IL-23, IL-22 production is independent of aryl hydrocarbon receptor (AhR) signaling. In acute and persistent viral infections, IL-22 deficiency resulted in thymic and splenic hypertrophy, while excessive IL-22 induced atrophy in these lymphoid organs. Moreover, IL-22 deficiency enhanced T cell responses to promote viral clearance, but increased IL-22 in vivo decreased T cell numbers and functions in the liver and lymphoid tissues. Together, our findings reveal a significant effect of the IL-23/PI3K/mTORC1 axis on regulating IL-22 production and also identify a novel role of IL-22 in controlling antiviral T cell responses in the non-lymphoid and lymphoid organs during acute and persistent viral infections.
Collapse
Affiliation(s)
- Panpan Yi
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Hunan, China
- Department of Microbiology and Immunology, University of Texas Medical Branch, Texas, USA
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Texas, USA
| | - Denley Ming Kee Yuan
- Department of Microbiology and Immunology, University of Texas Medical Branch, Texas, USA
| | - Zuliang Jie
- Department of Microbiology and Immunology, University of Texas Medical Branch, Texas, USA
| | - Zakari Kwota
- Department of Microbiology and Immunology, University of Texas Medical Branch, Texas, USA
| | - Yan Chen
- Department of Ophthalmology, University of Texas Medical Branch, Texas, USA
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Texas, USA
- Department of Pathology, University of Texas Medical Branch, Texas, USA
| | - Xuegong Fan
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan, Xiangya Hospital, Central South University, Hunan, China.
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Texas, USA.
- Department of Pathology, University of Texas Medical Branch, Texas, USA.
| |
Collapse
|
153
|
Ferrara JL, Smith CM, Sheets J, Reddy P, Serody JS. Altered homeostatic regulation of innate and adaptive immunity in lower gastrointestinal tract GVHD pathogenesis. J Clin Invest 2017; 127:2441-2451. [PMID: 28581444 DOI: 10.1172/jci90592] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lower gastrointestinal (GI) tract graft-versus-host disease (GVHD) is the predominant cause of morbidity and mortality from GVHD after allogeneic stem cell transplantation. Recent data indicate that lower GI tract GVHD is a complicated process mediated by donor/host antigenic disparities. This process is exacerbated by significant changes to the microbiome, and innate and adaptive immune responses that are critical to the induction of disease, persistence of inflammation, and a lack of response to therapy. Here, we discuss new insights into the biology of lower GI tract GVHD and focus on intrinsic pathways and regulatory mechanisms crucial to normal intestinal function. We then describe multiple instances in which these homeostatic mechanisms are altered by donor T cells or conditioning therapy, resulting in exacerbation of GVHD. We also discuss data suggesting that some of these mechanisms produce biomarkers that could be informative as to the severity of GVHD and its response to therapy. Finally, novel therapies that might restore homeostasis in the GI tract during GVHD are highlighted.
Collapse
Affiliation(s)
- James Lm Ferrara
- Departments of Medicine, Pediatrics, and Academic Informatics and Technology, Icahn School of Medicine at Mount Sinai and Tisch Cancer Institute, New York, New York, USA
| | - Christopher M Smith
- Departments of Medicine, Pediatrics, and Academic Informatics and Technology, Icahn School of Medicine at Mount Sinai and Tisch Cancer Institute, New York, New York, USA
| | - Julia Sheets
- University of North Carolina Hospital, Chapel Hill, North Carolina, USA
| | - Pavan Reddy
- Department of Medicine and University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Jonathan S Serody
- Department of Medicine and UNC Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
154
|
Innate lymphoid cells as regulators of immunity, inflammation and tissue homeostasis. Nat Immunol 2017; 17:765-74. [PMID: 27328006 DOI: 10.1038/ni.3489] [Citation(s) in RCA: 683] [Impact Index Per Article: 97.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/10/2016] [Indexed: 12/14/2022]
Abstract
Research over the last 7 years has led to the formal identification of innate lymphoid cells (ILCs), increased the understanding of their tissue distribution and has established essential functions of ILCs in diverse physiological processes. These include resistance to pathogens, the regulation of autoimmune inflammation, tissue remodeling, cancer and metabolic homeostasis. Notably, many ILC functions appear to be regulated by mechanisms distinct from those of other innate and adaptive immune cells. In this Review, we focus on how group 2 ILC (ILC2) and group 3 ILC (ILC3) responses are regulated and how these cells interact with other immune and non-immune cells to mediate their functions. We highlight experimental evidence from mouse models and patient-based studies that have elucidated the effects of ILCs on the maintenance of tissue homeostasis and the consequences for health and disease.
Collapse
|
155
|
Bando JK, Colonna M. Innate lymphoid cell function in the context of adaptive immunity. Nat Immunol 2017; 17:783-9. [PMID: 27328008 DOI: 10.1038/ni.3484] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/04/2016] [Indexed: 12/12/2022]
Abstract
Innate lymphoid cells (ILCs) are a family of innate immune cells that have diverse functions during homeostasis and disease. Subsets of ILCs have phenotypes that mirror those of polarized helper T cell subsets in their expression of core transcription factors and effector cytokines. Given the similarities between these two classes of lymphocytes, it is important to understand which functions of ILCs are specialized and which are redundant with those of T cells. Here we discuss genetic mouse models that have been used to delineate the contributions of ILCs versus those of T cells and review the current understanding of the specialized in vivo functions of ILCs.
Collapse
Affiliation(s)
- Jennifer K Bando
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
156
|
Abstract
This review aims to address the mechanisms of compromised immune tolerance contributing to the development and maintenance of Alopecia Areata (AA). Our goal is to also highlight future treatment opportunities and therapeutics that will safely and efficiently restore hair growth and maintain patients in remission. AA is a presumptive autoimmune disorder that coincides and genetically clusters to several other autoimmune diseases. In this review, we pay attention to the learnings from the mechanistic research and drug development in these other autoimmune conditions. Interestingly, most of these diseases have been linked to compromised central and peripheral tolerance, and increased intestinal inflammation with enhanced gut permeability. Break of tolerance and priming of the autoreactive T-cells to attack antigenic epitopes in the hair follicle most likely requires several steps which include escape from negative selection and compromised peripheral tolerance. Local skin-related changes are also of importance due to the patchy manifestation of the skin areas with loss of hair, particularly in the early disease. Here, we discuss the defective mechanisms of tolerance, both central and peripheral, and hypothesize that the disease is driven by areas of tolerance break, and that these could be targeted for successful therapeutic interventions.
Collapse
|
157
|
Abstract
Intrathymic T cell development is a complex process that depends upon continuous guidance from thymus stromal cell microenvironments. The thymic epithelium within the thymic stroma comprises highly specialized cells with a high degree of anatomic, phenotypic, and functional heterogeneity. These properties are collectively required to bias thymocyte development toward production of self-tolerant and functionally competent T cells. The importance of thymic epithelial cells (TECs) is evidenced by clear links between their dysfunction and multiple diseases where autoimmunity and immunodeficiency are major components. Consequently, TECs are an attractive target for cell therapies to restore effective immune system function. The pathways and molecular regulators that control TEC development are becoming clearer, as are their influences on particular stages of T cell development. Here, we review both historical and the most recent advances in our understanding of the cellular and molecular mechanisms controlling TEC development, function, dysfunction, and regeneration.
Collapse
Affiliation(s)
- Jakub Abramson
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Graham Anderson
- MRC Centre for Immune Regulation, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| |
Collapse
|
158
|
Fischer JC, Bscheider M, Eisenkolb G, Lin CC, Wintges A, Otten V, Lindemans CA, Heidegger S, Rudelius M, Monette S, Porosnicu Rodriguez KA, Calafiore M, Liebermann S, Liu C, Lienenklaus S, Weiss S, Kalinke U, Ruland J, Peschel C, Shono Y, Docampo M, Velardi E, Jenq RR, Hanash AM, Dudakov JA, Haas T, van den Brink MRM, Poeck H. RIG-I/MAVS and STING signaling promote gut integrity during irradiation- and immune-mediated tissue injury. Sci Transl Med 2017; 9:eaag2513. [PMID: 28424327 PMCID: PMC5604790 DOI: 10.1126/scitranslmed.aag2513] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 05/30/2016] [Accepted: 01/18/2017] [Indexed: 12/25/2022]
Abstract
The molecular pathways that regulate the tissue repair function of type I interferon (IFN-I) during acute tissue damage are poorly understood. We describe a protective role for IFN-I and the RIG-I/MAVS signaling pathway during acute tissue damage in mice. Mice lacking mitochondrial antiviral-signaling protein (MAVS) were more sensitive to total body irradiation- and chemotherapy-induced intestinal barrier damage. These mice developed worse graft-versus-host disease (GVHD) in a preclinical model of allogeneic hematopoietic stem cell transplantation (allo-HSCT) than did wild-type mice. This phenotype was not associated with changes in the intestinal microbiota but was associated with reduced gut epithelial integrity. Conversely, targeted activation of the RIG-I pathway during tissue injury promoted gut barrier integrity and reduced GVHD. Recombinant IFN-I or IFN-I expression induced by RIG-I promoted growth of intestinal organoids in vitro and production of the antimicrobial peptide regenerating islet-derived protein 3 γ (RegIIIγ). Our findings were not confined to RIG-I/MAVS signaling because targeted engagement of the STING (stimulator of interferon genes) pathway also protected gut barrier function and reduced GVHD. Consistent with this, STING-deficient mice suffered worse GVHD after allo-HSCT than did wild-type mice. Overall, our data suggest that activation of either RIG-I/MAVS or STING pathways during acute intestinal tissue injury in mice resulted in IFN-I signaling that maintained gut epithelial barrier integrity and reduced GVHD severity. Targeting these pathways may help to prevent acute intestinal injury and GVHD during allogeneic transplantation.
Collapse
Affiliation(s)
- Julius C Fischer
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Michael Bscheider
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Gabriel Eisenkolb
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chia-Ching Lin
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Alexander Wintges
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Vera Otten
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Caroline A Lindemans
- Pediatric Blood and Bone Marrow Transplant Program, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Simon Heidegger
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, University of Wuerzburg and Comprehensive Cancer Center Mainfranken, Wuerzburg, Germany
| | - Sébastien Monette
- Tri-Institutional Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, Rockefeller University, and Weill Cornell Medical College, New York, NY 10065, USA
| | | | - Marco Calafiore
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sophie Liebermann
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, New Jersey Medical School and Robert Wood Johnson Medical School, Rutgers University, Newark, NJ 08903, USA
| | - Stefan Lienenklaus
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Siegfried Weiss
- Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Christian Peschel
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Yusuke Shono
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Melissa Docampo
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Enrico Velardi
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Robert R Jenq
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alan M Hanash
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jarrod A Dudakov
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tobias Haas
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Marcel R M van den Brink
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hendrik Poeck
- III. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
- Department of Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
159
|
Ansari AR, Liu H. Acute Thymic Involution and Mechanisms for Recovery. Arch Immunol Ther Exp (Warsz) 2017; 65:401-420. [PMID: 28331940 DOI: 10.1007/s00005-017-0462-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 03/12/2017] [Indexed: 12/14/2022]
Abstract
Acute thymic involution (ATI) is usually regarded as a virulence trait. It is caused by several infectious agents (bacteria, viruses, parasites, fungi) and other factors, including stress, pregnancy, malnutrition and chemotherapy. However, the complex mechanisms that operate during ATI differ substantially from each other depending on the causative agent. For instance, a transient reduction in the size and weight of the thymus and depletion of populations of T cell subsets are hallmarks of ATI in many cases, whereas severe disruption of the anatomical structure of the organ is also associated with some factors, including fungal, parasitic and viral infections. However, growing evidence shows that ATI may be therapeutically halted or reversed. In this review, we highlight the current progress in this field with respect to numerous pathological factors and discuss the possible mechanisms. Moreover, these new observations also show that ATI can be mechanistically reversed.
Collapse
Affiliation(s)
- Abdur Rahman Ansari
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, Hubei, China.,Section of Anatomy and Histology, Department of Basic Sciences, College of Veterinary and Animal Sciences (CVAS), Jhang, Pakistan.,University of Veterinary and Animal Sciences (UVAS), Lahore, Pakistan
| | - Huazhen Liu
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, Hubei, China.
| |
Collapse
|
160
|
Establishment and function of tissue-resident innate lymphoid cells in the skin. Protein Cell 2017; 8:489-500. [PMID: 28271445 PMCID: PMC5498338 DOI: 10.1007/s13238-017-0388-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 02/15/2017] [Indexed: 02/07/2023] Open
Abstract
Innate lymphoid cells (ILCs) are a newly classified family of immune cells of the lymphoid lineage. While they could be found in both lymphoid organs and non-lymphoid tissues, ILCs are preferentially enriched in barrier tissues such as the skin, intestine, and lung where they could play important roles in maintenance of tissue integrity and function and protection against assaults of foreign agents. On the other hand, dysregulated activation of ILCs could contribute to tissue inflammatory diseases. In spite of recent progress towards understanding roles of ILCs in the health and disease, mechanisms regulating specific establishment, activation, and function of ILCs in barrier tissues are still poorly understood. We herein review the up-to-date understanding of tissue-specific relevance of ILCs. Particularly we will focus on resident ILCs of the skin, the outmost barrier tissue critical in protection against various foreign hazardous agents and maintenance of thermal and water balance. In addition, we will discuss remaining outstanding questions yet to be addressed.
Collapse
|
161
|
Pathogenic and Protective Autoantibodies in Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy (APECED). Antibodies (Basel) 2017; 6:antib6010001. [PMID: 31548517 PMCID: PMC6698825 DOI: 10.3390/antib6010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 11/22/2016] [Accepted: 01/13/2017] [Indexed: 12/31/2022] Open
Abstract
Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is a rare disorder caused by mutations in the autoimmune regulator (AIRE) gene, leading to defects in T cell selection. The disease manifestations include both autoimmune tissue destruction and immunodeficiency, with specific susceptibility to chronic mucocutaneous candidiasis. Studies have demonstrated a wide repertoire of high affinity tissue- and cytokine-specific antibodies in patients with APECED. Here, we review the antigenic targets and function of these disease-causing and disease-ameliorating antibodies.
Collapse
|
162
|
Eberl G. RORγt, a multitask nuclear receptor at mucosal surfaces. Mucosal Immunol 2017; 10:27-34. [PMID: 27706126 DOI: 10.1038/mi.2016.86] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/30/2016] [Indexed: 02/07/2023]
Abstract
RORγt is a nuclear hormone receptor that has followed an exponential success carrier. Its modest origins as an orphan receptor cloned from human pancreas blossomed within 15 years into a critical regulator of anti-microbial immunity and a major target in the fight against inflammatory pathologies. Here, I review its role as a transcription factor required for the generation of type 3 lymphoid cells, which induce the development of lymphoid tissues, provide resistance of epithelial stem cells to injury, maintain homeostasis with the symbiotic microbiota, orchestrate defense against extracellular microbes, and regulate allergic responses. RORγt is also an intriguing molecule that is regulated by the circadian rhythm and includes cholesterol metabolites as ligands. RORγt therefore links anti-microbial immunity with circadian rhythms and steroids, the logic of which remains to be understood.
Collapse
Affiliation(s)
- G Eberl
- Institut Pasteur, Microenvironment & Immunity Unit, Department of Immunology, Paris, France.,INSERM U1224, Paris, France
| |
Collapse
|
163
|
The Innate Immune Response in Myocardial Infarction, Repair, and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1003:251-272. [PMID: 28667562 DOI: 10.1007/978-3-319-57613-8_12] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Following myocardial infarction (MI), resident innate immune cells such as macrophages, innate lymphoid cells, and mast cells rapidly coordinate their function to contain inflammation by removing dying cells and promoting cardiomyocyte replenishment. To sustain local tissue repair functions, hematopoietic progenitors are mobilized from the bone marrow to the spleen to generate subsequent myeloid cells such as monocytes and neutrophils, which are rapidly recruited at the site of MI. A finely tuned balance between local adaptation and recruitment controls the overall outcome of the cardiac tissue regeneration versus repair and scar formation.In this chapter, the (potential) roles of the innate immune system residing in the heart are discussed in the context of recent findings about macrophage ontogeny and their homeostasis with circulating monocytes during cardiac tissue growth and after myocardial infarction. Their interactions with other members of the innate immune system are also discussed with a particular emphasis on the potential involvement of mast cells and innate lymphoid cells during MI, largely underestimated until recently. Understanding the development and the functions of the different protagonists responding to MI as well as their potential cross talk could help design new strategies for regenerative medicine intervention.
Collapse
|
164
|
Abstract
The family of innate lymphoid cells (ILCs) has attracted attention in recent years as its members are important regulators of immunity, while they can also cause pathology. In both mouse and man, ILCs were initially discovered in developing lymph nodes as lymphoid tissue inducer (LTi) cells. These cells form the prototypic members of the ILC family and play a central role in the formation of secondary lymphoid organs (SLOs). In the absence of LTi cells, lymph nodes (LN) and Peyer's Patches (PP) fail to form in mice, although the splenic white pulp can develop normally. Besides LTi cells, the ILC family encompasses helper-like ILCs with functional distinctions as seen by T-helper cells, as well as cytotoxic natural killer (NK) cells. ILCs are still present in adult SLOs where they have been shown to play a role in lymphoid tissue regeneration. Furthermore, ILCs were implicated to interact with adaptive lymphocytes and influence the adaptive immune response. Here, we review the recent literature on the role of ILCs in secondary lymphoid tissue from the formation of SLOs to mature SLOs in adults, during homeostasis and pathology.
Collapse
Affiliation(s)
- Yotam E Bar-Ephraïm
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
165
|
Kugler DG, Flomerfelt FA, Costa DL, Laky K, Kamenyeva O, Mittelstadt PR, Gress RE, Rosshart SP, Rehermann B, Ashwell JD, Sher A, Jankovic D. Systemic toxoplasma infection triggers a long-term defect in the generation and function of naive T lymphocytes. J Exp Med 2016; 213:3041-3056. [PMID: 27849554 PMCID: PMC5154934 DOI: 10.1084/jem.20151636] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 08/05/2016] [Accepted: 10/18/2016] [Indexed: 12/12/2022] Open
Abstract
Kugler et al. show that systemic infection with Toxoplasma gondii triggers a long-term impairment in thymic function, which leads to an immunodeficient state reflected in decreased antimicrobial resistance. Because antigen-stimulated naive T cells either die as effectors or enter the activated/memory pool, continuous egress of new T lymphocytes from thymus is essential for maintenance of peripheral immune homeostasis. Unexpectedly, we found that systemic infection with the protozoan Toxoplasma gondii triggers not only a transient increase in activated CD4+ Th1 cells but also a persistent decrease in the size of the naive CD4+ T lymphocyte pool. This immune defect is associated with decreased thymic output and parasite-induced destruction of the thymic epithelium, as well as disruption of the overall architecture of that primary lymphoid organ. Importantly, the resulting quantitative and qualitative deficiency in naive CD4+ T cells leads to an immunocompromised state that both promotes chronic toxoplasma infection and leads to decreased resistance to challenge with an unrelated pathogen. These findings reveal that systemic infectious agents, such as T. gondii, can induce long-term immune alterations associated with impaired thymic function. When accumulated during the lifetime of the host, such events, even when occurring at low magnitude, could be a contributing factor in immunological senescence.
Collapse
Affiliation(s)
- David G Kugler
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Francis A Flomerfelt
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Diego L Costa
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Karen Laky
- T Cell Development Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Olena Kamenyeva
- Biological Imaging, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Paul R Mittelstadt
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Ronald E Gress
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Stephan P Rosshart
- Immunology Section, Liver Diseases Branch, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jonathan D Ashwell
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Dragana Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
166
|
El-Jawhari JJ, Jones E, Giannoudis PV. The roles of immune cells in bone healing; what we know, do not know and future perspectives. Injury 2016; 47:2399-2406. [PMID: 27809990 DOI: 10.1016/j.injury.2016.10.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Key events occurring during the bone healing include well-orchestrated and complex interactions between immune cells, multipotential stromal cells (MSCs), osteoblasts and osteoclasts. Through three overlapping phases of this physiological process, innate and adaptive immune cells, cytokines and chemokines have a significant role to play. The aim of the escalating immune response is to achieve an osseous healing in the shortest time and with the least complications facilitating the restoration of function. The uninterrupted progression of these biological events in conjunction with a favourable mechanical environment (stable fracture fixation) remains the hallmark of successful fracture healing. When failure occurs, either the biological environment or the mechanical one could have been disrupted. Not infrequently both may be compromised. Consequently, regenerative treatments involving the use of bone autograft, allograft or synthetic matrices supplemented with MSCs are increasingly used. A better understanding of the bone biology and osteoimmunology can help to improve these evolving cell-therapy based strategies. Herein, an up to date status of the role of immune cells during the different phases of bone healing is presented. Additionally, the known and yet to know events about immune cell interactions with MSCs and osteoblasts and osteoclasts and the therapeutic implications are being discussed.
Collapse
Affiliation(s)
- Jehan J El-Jawhari
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St. James Hospital, University of Leeds, UK; NIHR Biomedical Research Unit, Chapel Allerton Hospital, University of Leeds, UK; Clinical Pathology Department, Faculty of Medicine, Mansoura University, Egypt
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St. James Hospital, University of Leeds, UK; NIHR Biomedical Research Unit, Chapel Allerton Hospital, University of Leeds, UK
| | - Peter V Giannoudis
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St. James Hospital, University of Leeds, UK; NIHR Biomedical Research Unit, Chapel Allerton Hospital, University of Leeds, UK.
| |
Collapse
|
167
|
Scalea JR, Hickman JB, Moore DJ, Brayman KL. An overview of the necessary thymic contributions to tolerance in transplantation. Clin Immunol 2016; 173:S1521-6616(16)30382-5. [PMID: 27989896 DOI: 10.1016/j.clim.2016.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 10/04/2016] [Accepted: 10/22/2016] [Indexed: 02/08/2023]
Abstract
The thymus is important for the development of the immune system. However, aging leads to predictable involution of the thymus and immunodeficiency. These immunodeficiencies may be rectified with thymic rejuvenation. Atrophy of the thymus is governed by a complex interplay of molecular, cytokine and hormonal factors. Herein we review the interaction of these factors across age and how they may be targeted for thymic rejuvenation. We further discuss the growing pre-clinical evidence defining the necessary and sufficient contributions of the thymus to successful tolerance induction in transplantation.
Collapse
Affiliation(s)
- Joseph R Scalea
- Division of Transplantation, Department of Surgery, University of Maryland, United States.
| | - John B Hickman
- School of Medicine, University of Virginia, United States
| | - Daniel J Moore
- Division of Endocrinology, Department of Pediatrics, Department of Pathology, Microbiology and Immunology, Vanderbilt University, United States
| | - Kenneth L Brayman
- School of Medicine, University of Virginia, United States; Division of Transplantation, Department of Surgery, University of Virginia, United States
| |
Collapse
|
168
|
Tait Wojno ED, Artis D. Emerging concepts and future challenges in innate lymphoid cell biology. J Exp Med 2016; 213:2229-2248. [PMID: 27811053 PMCID: PMC5068238 DOI: 10.1084/jem.20160525] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 09/26/2016] [Indexed: 12/15/2022] Open
Abstract
Innate lymphoid cells (ILCs) are innate immune cells that are ubiquitously distributed in lymphoid and nonlymphoid tissues and enriched at mucosal and barrier surfaces. Three major ILC subsets are recognized in mice and humans. Each of these subsets interacts with innate and adaptive immune cells and integrates cues from the epithelium, the microbiota, and pathogens to regulate inflammation, immunity, tissue repair, and metabolic homeostasis. Although intense study has elucidated many aspects of ILC development, phenotype, and function, numerous challenges remain in the field of ILC biology. In particular, recent work has highlighted key new questions regarding how these cells communicate with their environment and other cell types during health and disease. This review summarizes new findings in this rapidly developing field that showcase the critical role ILCs play in directing immune responses through their ability to interact with a variety of hematopoietic and nonhematopoietic cells. In addition, we define remaining challenges and emerging questions facing the field. Finally, this review discusses the potential application of basic studies of ILC biology to the development of new treatments for human patients with inflammatory and infectious diseases in which ILCs play a role.
Collapse
Affiliation(s)
- Elia D Tait Wojno
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853 .,Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10065.,Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065.,Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065
| |
Collapse
|
169
|
Masters AR, Haynes L, Su DM, Palmer DB. Immune senescence: significance of the stromal microenvironment. Clin Exp Immunol 2016; 187:6-15. [PMID: 27529161 DOI: 10.1111/cei.12851] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2016] [Indexed: 12/12/2022] Open
Abstract
The immune system undergoes age-associated changes known as immunosenescence, resulting in increased susceptibility to infections, cancers and autoimmunity in the aged. The basis of our understanding of immunosenescence has been derived primarily from studies examining intrinsic defects within many of the cells of the immune system. While these studies have provided insight into the mechanisms of immunosenescence, a picture is now emerging that the stromal microenvironment within lymphoid organs also contributes significantly to the age-associated decline of immune function. These extrinsic defects appear to impact the functional activity of immune cells and may offer a potential target to recover immune activity. Indeed, rejuvenation studies which have targeted the stromal niche have restored immune function in aged successfully, highlighting the impact of the microenvironment towards the aetiology of immunosenescence.
Collapse
Affiliation(s)
- A R Masters
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, USA.,Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - L Haynes
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, USA.,Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - D-M Su
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX, USA
| | - D B Palmer
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, UK
| |
Collapse
|
170
|
Hoover AR, Dozmorov I, MacLeod J, Du Q, de la Morena MT, Forbess J, Guleserian K, Cleaver OB, van Oers NSC. MicroRNA-205 Maintains T Cell Development following Stress by Regulating Forkhead Box N1 and Selected Chemokines. J Biol Chem 2016; 291:23237-23247. [PMID: 27646003 DOI: 10.1074/jbc.m116.744508] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Indexed: 12/27/2022] Open
Abstract
The thymus, an organ responsible for T cell development, is one of the more stress-sensitive tissues in the body. Stress, in the form of infections, radiation exposure, and steroids, impairs thymic epithelial cell (TEC) functions and induces the programmed cell death of immature thymocytes. MicroRNAs are small noncoding RNAs involved in tissue repair and homeostasis, with several supporting T cell development. We report that miR-205, an epithelial-specific miR, maintains thymopoiesis following inflammatory perturbations. Thus, the activation of diverse pattern recognition receptors in mice causes a more severe thymic hypoplasia and delayed T cell recovery when miR-205 is conditionally ablated in TECs. Gene expression comparisons in the TECs with/without miR-205 revealed a significant differential regulation of chemokine/chemokine receptor pathways, antigen processing components, and changes in the Wnt signaling system. This was partly a consequence of reduced expression of the transcriptional regulator of epithelial cell function, Forkhead Box N1 (Foxn1), and its two regulated targets, stem cell factor and ccl25, following stress. miR-205 mimics supplemented into miR-205-deficient fetal thymic organ cultures restored Foxn1 expression along with ccl25 and stem cell factor A number of putative targets of miR-205 were up-regulated in TECs lacking miR-205, consistent with an important role for this miR in supporting T cell development in response to stress.
Collapse
Affiliation(s)
| | | | | | | | | | - Joseph Forbess
- Internal Medicine.,Children's Health, Dallas, Texas 75235
| | | | | | - Nicolai S C van Oers
- From the Departments of Immunology, .,Pediatrics.,Microbiology, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9093 and
| |
Collapse
|
171
|
Yoon J, Leyva-Castillo JM, Wang G, Galand C, Oyoshi MK, Kumar L, Hoff S, He R, Chervonsky A, Oppenheim JJ, Kuchroo VK, van den Brink MRM, Malefyt RDW, Tessier PA, Fuhlbrigge R, Rosenstiel P, Terhorst C, Murphy G, Geha RS. IL-23 induced in keratinocytes by endogenous TLR4 ligands polarizes dendritic cells to drive IL-22 responses to skin immunization. J Exp Med 2016; 213:2147-66. [PMID: 27551155 PMCID: PMC5032726 DOI: 10.1084/jem.20150376] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/25/2016] [Indexed: 12/12/2022] Open
Abstract
Atopic dermatitis (AD) is a Th2-dominated inflammatory skin disease characterized by epidermal thickening. Serum levels of IL-22, a cytokine known to induce keratinocyte proliferation, are elevated in AD, and Th22 cells infiltrate AD skin lesions. We show that application of antigen to mouse skin subjected to tape stripping, a surrogate for scratching, induces an IL-22 response that drives epidermal hyperplasia and keratinocyte proliferation in a mouse model of skin inflammation that shares many features of AD. DC-derived IL-23 is known to act on CD4(+) T cells to induce IL-22 production. However, the mechanisms that drive IL-23 production by skin DCs in response to cutaneous sensitization are not well understood. We demonstrate that IL-23 released by keratinocytes in response to endogenous TLR4 ligands causes skin DCs, which selectively express IL-23R, to up-regulate their endogenous IL-23 production and drive an IL-22 response in naive CD4(+) T cells that mediates epidermal thickening. We also show that IL-23 is released in human skin after scratching and polarizes human skin DCs to drive an IL-22 response, supporting the utility of IL-23 and IL-22 blockade in AD.
Collapse
Affiliation(s)
- Juhan Yoon
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Juan Manuel Leyva-Castillo
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Guoxing Wang
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Claire Galand
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Michiko K Oyoshi
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Lalit Kumar
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Sabine Hoff
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Rui He
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | | | - Joost J Oppenheim
- Laboratory of Molecular Immunoregulation, National Cancer Institute, Frederick, MD 21702
| | - Vijay K Kuchroo
- Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Marcel R M van den Brink
- Department of Immunology and Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | | | - Philippe A Tessier
- Centre de Recherche du Centre Hospitalier de l'Université Laval, Sainte-Foy, Quebec QC G1V 4G2, Canada
| | - Robert Fuhlbrigge
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - George Murphy
- Department of Dermatology, Harvard Medical School, Boston, MA 02115 Division of Dermatopathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Raif S Geha
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
172
|
The Immune System in Tissue Environments Regaining Homeostasis after Injury: Is "Inflammation" Always Inflammation? Mediators Inflamm 2016; 2016:2856213. [PMID: 27597803 PMCID: PMC4997018 DOI: 10.1155/2016/2856213] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 07/08/2016] [Accepted: 07/19/2016] [Indexed: 12/31/2022] Open
Abstract
Inflammation is a response to infections or tissue injuries. Inflammation was once defined by clinical signs, later by the presence of leukocytes, and nowadays by expression of "proinflammatory" cytokines and chemokines. But leukocytes and cytokines often have rather anti-inflammatory, proregenerative, and homeostatic effects. Is there a need to redefine "inflammation"? In this review, we discuss the functions of "inflammatory" mediators/regulators of the innate immune system that determine tissue environments to fulfill the need of the tissue while regaining homeostasis after injury.
Collapse
|
173
|
Fan X, Rudensky AY. Hallmarks of Tissue-Resident Lymphocytes. Cell 2016; 164:1198-1211. [PMID: 26967286 DOI: 10.1016/j.cell.2016.02.048] [Citation(s) in RCA: 269] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Indexed: 01/20/2023]
Abstract
Although they are classically viewed as continuously recirculating through the lymphoid organs and blood, lymphocytes also establish residency in non-lymphoid tissues, most prominently at barrier sites, including the mucosal surfaces and skin. These specialized tissue-resident lymphocyte subsets span the innate-adaptive continuum and include innate lymphoid cells (ILCs), unconventional T cells (e.g., NKT, MAIT, γδ T cells, and CD8αα(+) IELs), and tissue-resident memory T (T(RM)) cells. Although these diverse cell types differ in the particulars of their biology, they nonetheless exhibit important shared features, including a role in the preservation of tissue integrity and function during homeostasis, infection, and non-infectious perturbations. In this Review, we discuss the hallmarks of tissue-resident innate, innate-like, and adaptive lymphocytes, as well as their potential functions in non-lymphoid organs.
Collapse
Affiliation(s)
- Xiying Fan
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan-Kettering Cancer Center, 417 East 68(th) Street, New York, NY 10065, USA.
| | - Alexander Y Rudensky
- Howard Hughes Medical Institute and Immunology Program, Memorial Sloan-Kettering Cancer Center, 417 East 68(th) Street, New York, NY 10065, USA; Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, 417 East 68(th) Street, New York, NY 10065, USA.
| |
Collapse
|
174
|
Meyer S, Woodward M, Hertel C, Vlaicu P, Haque Y, Kärner J, Macagno A, Onuoha SC, Fishman D, Peterson H, Metsküla K, Uibo R, Jäntti K, Hokynar K, Wolff ASB, Krohn K, Ranki A, Peterson P, Kisand K, Hayday A. AIRE-Deficient Patients Harbor Unique High-Affinity Disease-Ameliorating Autoantibodies. Cell 2016; 166:582-595. [PMID: 27426947 PMCID: PMC4967814 DOI: 10.1016/j.cell.2016.06.024] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/24/2016] [Accepted: 06/10/2016] [Indexed: 01/01/2023]
Abstract
APS1/APECED patients are defined by defects in the autoimmune regulator (AIRE) that mediates central T cell tolerance to many self-antigens. AIRE deficiency also affects B cell tolerance, but this is incompletely understood. Here we show that most APS1/APECED patients displayed B cell autoreactivity toward unique sets of approximately 100 self-proteins. Thereby, autoantibodies from 81 patients collectively detected many thousands of human proteins. The loss of B cell tolerance seemingly occurred during antibody affinity maturation, an obligatorily T cell-dependent step. Consistent with this, many APS1/APECED patients harbored extremely high-affinity, neutralizing autoantibodies, particularly against specific cytokines. Such antibodies were biologically active in vitro and in vivo, and those neutralizing type I interferons (IFNs) showed a striking inverse correlation with type I diabetes, not shown by other anti-cytokine antibodies. Thus, naturally occurring human autoantibodies may actively limit disease and be of therapeutic utility.
Collapse
Affiliation(s)
- Steffen Meyer
- ImmunoQure AG, Königsallee 90, 2012 Düsseldorf, Germany
| | - Martin Woodward
- Peter Gorer Department of Immunobiology, King's College, London SE19RT, UK
| | | | - Philip Vlaicu
- ImmunoQure AG, Königsallee 90, 2012 Düsseldorf, Germany
| | - Yasmin Haque
- Peter Gorer Department of Immunobiology, King's College, London SE19RT, UK
| | - Jaanika Kärner
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, Tartu 50411, Estonia
| | - Annalisa Macagno
- ImmunoQure Research AG, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Shimobi C Onuoha
- ImmunoQure Research AG, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Dmytro Fishman
- Institute of Computer Science, University of Tartu, Liivi 2, Tartu 50409, Estonia; Quretec Ltd., Ülikooli 6A, Tartu 51003, Estonia
| | - Hedi Peterson
- Institute of Computer Science, University of Tartu, Liivi 2, Tartu 50409, Estonia; Quretec Ltd., Ülikooli 6A, Tartu 51003, Estonia
| | - Kaja Metsküla
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, Tartu 50411, Estonia
| | - Raivo Uibo
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, Tartu 50411, Estonia
| | - Kirsi Jäntti
- Clinical Research Institute HUCH Ltd., Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Kati Hokynar
- Clinical Research Institute HUCH Ltd., Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Anette S B Wolff
- Department of Clinical Science, University of Bergen, Laboratory Building, 8th floor, 5021 Bergen, Norway
| | - Kai Krohn
- Clinical Research Institute HUCH Ltd., Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Annamari Ranki
- Department of Dermatology, Allergology and Venereology, Institute of Clinical Medicine, University of Helsinki, Skin and Allergy Hospital, Helsinki University Central Hospital, Meilahdentie 2, 00250 Helsinki, Finland
| | - Pärt Peterson
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, Tartu 50411, Estonia
| | - Kai Kisand
- Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, Tartu 50411, Estonia.
| | - Adrian Hayday
- Peter Gorer Department of Immunobiology, King's College, London SE19RT, UK.
| |
Collapse
|
175
|
Lu Z, Liu R, Huang E, Chu Y. MicroRNAs: New regulators of IL-22. Cell Immunol 2016; 304-305:1-8. [PMID: 27221197 DOI: 10.1016/j.cellimm.2016.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/11/2016] [Accepted: 05/16/2016] [Indexed: 12/18/2022]
Abstract
Interleukin-22 (IL-22) is a cytokine that belongs to the IL-10 family of interleukins. It can be produced by T helper 22 (Th22) cells, T helper 1 (Th1) cells, T helper 17 (Th17) cells, natural killer 22 (NK22) cells, natural killer T (NKT) cells, innate lymphoid cells (ILCs), and γδ T cells. IL-22 acts via binding to a heterodimeric transmembrane receptor complex that consists of IL-22R1 and IL-10R2 and mainly contributes to the tissue repair and host defense. Transcription factors such as retinoid orphan receptor γt (RORγt) and signal transducer and activator of transcription 3 (STAT3), have been reported to play important roles in regulation of IL-22 expression. Recently, it has been demonstrated in several studies that microRNAs (miRNAs) potently regulate expression of interleukins, including production of IL-22. Here, we review current knowledge about regulators of IL-22 expression with a particular emphasis on the role of miRNAs.
Collapse
Affiliation(s)
- Zhou Lu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, People's Republic of China
| | - Ronghua Liu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, People's Republic of China
| | - Enyu Huang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, People's Republic of China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, People's Republic of China; Biotherapy Research Center, Fudan University, Shanghai 200032, People's Republic of China.
| |
Collapse
|
176
|
Tajima A, Pradhan I, Trucco M, Fan Y. Restoration of Thymus Function with Bioengineered Thymus Organoids. CURRENT STEM CELL REPORTS 2016; 2:128-139. [PMID: 27529056 PMCID: PMC4982700 DOI: 10.1007/s40778-016-0040-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The thymus is the primary site for the generation of a diverse repertoire of T-cells that are essential to the efficient function of adaptive immunity. Numerous factors varying from aging, chemotherapy, radiation exposure, virus infection and inflammation contribute to thymus involution, a phenomenon manifested as loss of thymus cellularity, increased stromal fibrosis and diminished naïve T-cell output. Rejuvenating thymus function is a challenging task since it has limited regenerative capability and we still do not know how to successfully propagate thymic epithelial cells (TECs), the predominant population of the thymic stromal cells making up the thymic microenvironment. Here, we will discuss recent advances in thymus regeneration and the prospects of applying bioengineered artificial thymus organoids in regenerative medicine and solid organ transplantation.
Collapse
Affiliation(s)
- Asako Tajima
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA 15212
| | - Isha Pradhan
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA 15212
| | - Massimo Trucco
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA 15212
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19104
| | - Yong Fan
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA 15212
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19104
| |
Collapse
|
177
|
Vacca P, Montaldo E, Croxatto D, Moretta F, Bertaina A, Vitale C, Locatelli F, Mingari MC, Moretta L. NK Cells and Other Innate Lymphoid Cells in Hematopoietic Stem Cell Transplantation. Front Immunol 2016; 7:188. [PMID: 27242795 PMCID: PMC4870263 DOI: 10.3389/fimmu.2016.00188] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/02/2016] [Indexed: 01/03/2023] Open
Abstract
Natural killer (NK) cells play a major role in the T-cell depleted haploidentical hematopoietic stem cell transplantation (haplo-HSCT) to cure high-risk leukemias. NK cells belong to the expanding family of innate lymphoid cells (ILCs). At variance with NK cells, the other ILC populations (ILC1/2/3) are non-cytolytic, while they secrete different patterns of cytokines. ILCs provide host defenses against viruses, bacteria, and parasites, drive lymphoid organogenesis, and contribute to tissue remodeling. In haplo-HSCT patients, the extensive T-cell depletion is required to prevent graft-versus-host disease (GvHD) but increases risks of developing a wide range of life-threatening infections. However, these patients may rely on innate defenses that are reconstituted more rapidly than the adaptive ones. In this context, ILCs may represent important players in the early phases following transplantation. They may contribute to tissue homeostasis/remodeling and lymphoid tissue reconstitution. While the reconstitution of NK cell repertoire and its role in haplo-HSCT have been largely investigated, little information is available on ILCs. Of note, CD34+ cells isolated from different sources of HSC may differentiate in vitro toward various ILC subsets. Moreover, cytokines released from leukemia blasts (e.g., IL-1β) may alter the proportions of NK cells and ILC3, suggesting the possibility that leukemia may skew the ILC repertoire. Further studies are required to define the timing of ILC development and their potential protective role after HSCT.
Collapse
Affiliation(s)
- Paola Vacca
- Department of Experimental Medicine, University of Genova , Genova , Italy
| | - Elisa Montaldo
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute , Milano , Italy
| | - Daniele Croxatto
- Department of Experimental Medicine, University of Genova , Genova , Italy
| | - Francesca Moretta
- Department of Internal Medicine, University of Verona, Verona, Italy; Ospedale Sacro Cuore Negrar, Verona, Italy
| | - Alice Bertaina
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| | - Chiara Vitale
- Department of Experimental Medicine, University of Genova, Genova, Italy; U.O. Immunology IRCCS AOU San Martino-IST, Genova, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy; Department of Pediatrics, University of Pavia, Pavia, Italy
| | - Maria Cristina Mingari
- Department of Experimental Medicine, University of Genova, Genova, Italy; U.O. Immunology IRCCS AOU San Martino-IST, Genova, Italy; Centro di Eccellenza per la Ricerca Biomedica - CEBR, Genova, Italy
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital , Rome , Italy
| |
Collapse
|
178
|
Banuelos J, Lu NZ. A gradient of glucocorticoid sensitivity among helper T cell cytokines. Cytokine Growth Factor Rev 2016; 31:27-35. [PMID: 27235091 DOI: 10.1016/j.cytogfr.2016.05.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 12/19/2022]
Abstract
Helper T (Th) cells secret specific cytokines that promote immune responses whereas glucocorticoids limit the extent of immune responses by inhibiting cytokine secretion and other functions of Th cells. However, glucocorticoid resistance develops in subgroups of patients with Th cell-driven diseases such as asthma and Crohn's disease. Recent evidence supports that Th1, Th2, and Th17 cells have distinct glucocorticoid sensitivity. Th1 cells are sensitive to glucocorticoid-induced apoptosis and cytokine suppression while Th2 cells are sensitive to the latter but not the former and Th17 cells are resistant to both. This gradient of glucocorticoid sensitivity of Th cells corresponds to the glucocorticoid sensitivity of the diseases they underlie. We identify the mechanisms contributing to distinct glucocorticoid sensitivity of Th cells and their cytokines in the literature, as this information is useful to improve treatment strategies for glucocorticoid resistant immunological disorders.
Collapse
Affiliation(s)
- Jesus Banuelos
- Division of Allergy-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States, United States
| | - Nicholas Z Lu
- Division of Allergy-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States, United States.
| |
Collapse
|
179
|
Abstract
As the primary site of T-cell development, the thymus plays a key role in the generation of a strong yet self-tolerant adaptive immune response, essential in the face of the potential threat from pathogens or neoplasia. As the importance of the role of the thymus has grown, so too has the understanding that it is extremely sensitive to both acute and chronic injury. The thymus undergoes rapid degeneration following a range of toxic insults, and also involutes as part of the aging process, albeit at a faster rate than many other tissues. The thymus is, however, capable of regenerating, restoring its function to a degree. Potential mechanisms for this endogenous thymic regeneration include keratinocyte growth factor (KGF) signaling, and a more recently described pathway in which innate lymphoid cells produce interleukin-22 (IL-22) in response to loss of double positive thymocytes and upregulation of IL-23 by dendritic cells. Endogenous repair is unable to fully restore the thymus, particularly in the aged population, and this paves the way toward the need for exogenous strategies to help regenerate or even replace thymic function. Therapies currently in clinical trials include KGF, use of the cytokines IL-7 and IL-22, and hormonal modulation including growth hormone administration and sex steroid inhibition. Further novel strategies are emerging in the preclinical setting, including the use of precursor T cells and thymus bioengineering. The use of such strategies offers hope that for many patients, the next regeneration of their thymus is a step closer.
Collapse
Affiliation(s)
- Mohammed S Chaudhry
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Enrico Velardi
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jarrod A Dudakov
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Marcel R M van den Brink
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
180
|
Perusina Lanfranca M, Lin Y, Fang J, Zou W, Frankel T. Biological and pathological activities of interleukin-22. J Mol Med (Berl) 2016; 94:523-34. [PMID: 26923718 PMCID: PMC4860114 DOI: 10.1007/s00109-016-1391-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/17/2015] [Accepted: 01/21/2016] [Indexed: 12/16/2022]
Abstract
Interleukin (IL)-22, a member of the IL-10 family, is a cytokine secreted by several types of immune cells including IL-22(+)CD4(+) T cells (Th22) and IL-22 expressing innate leukocytes (ILC22). Recent studies have demonstrated that IL-22 is a key component in mucosal barrier defense, tissue repair, epithelial cell survival, and proliferation. Furthermore, accumulating evidence has defined both protective and pathogenic properties of IL-22 in a number of conditions including autoimmune disease, infection, and malignancy. In this review, we summarize the expression and signaling pathway and functional characteristics of the IL-22 and IL-22 receptor axis in physiological and pathological scenarios and discuss the potential to target IL-22 signaling to treat human diseases.
Collapse
Affiliation(s)
- Mirna Perusina Lanfranca
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
| | - Yanwei Lin
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai, 200001, China
| | - Jingyuan Fang
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai, 200001, China
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
- The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
- Graduate Programs in Immunology and Tumor Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Timothy Frankel
- Department of Surgery, University of Michigan School of Medicine, 109 Zina Pitcher Place, Ann Arbor, MI, 48109, USA.
- The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
181
|
Lamarthée B, Malard F, Saas P, Mohty M, Gaugler B. Interleukin-22 in Graft-Versus-Host Disease after Allogeneic Stem Cell Transplantation. Front Immunol 2016; 7:148. [PMID: 27148267 PMCID: PMC4836046 DOI: 10.3389/fimmu.2016.00148] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/04/2016] [Indexed: 01/20/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potential curative treatment for hematologic malignancies and non-malignant diseases. Because of the lower toxicity of reduced intensity conditioning, the number of transplants is in constant increase. However, allo-HSCT is still limited by complications, such as graft-versus-host disease (GVHD), which is associated with important morbidity and mortality. Acute GVHD is an exacerbated inflammatory response that leads to the destruction of healthy host tissues by donor immune cells. Recently, the contribution of innate immunity in GVHD triggering has been investigated by several groups and resulted in the identification of new cellular and molecular effectors involved in GVHD pathogenesis. Interleukin-22 (IL-22) is produced by both immune and adaptive cells and has both protective and inflammatory properties. Its role in GVHD processes has been investigated, and the data suggest that its effect depends on the timing, the target tissue, and the origin of the producing cells (donor/host). In this review, we discuss the role of IL-22 in allo-HSCT and GVHD.
Collapse
Affiliation(s)
- Baptiste Lamarthée
- Centre de Recherche Saint Antoine, INSERM UMR 938, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Florent Malard
- Centre de Recherche Saint Antoine, INSERM UMR 938, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Philippe Saas
- INSERM UMR1098, Besançon, France; UMR 1098, SFR FED 4234, Université de Bourgogne Franche-Comté, Besançon, France; UMR 1098, Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France
| | - Mohamad Mohty
- Centre de Recherche Saint Antoine, INSERM UMR 938, Paris, France; Université Pierre et Marie Curie, Paris, France; Service d'Hématologie Clinique, Hôpital Saint-Antoine, Paris, France
| | - Béatrice Gaugler
- Centre de Recherche Saint Antoine, INSERM UMR 938, Paris, France; Université Pierre et Marie Curie, Paris, France
| |
Collapse
|
182
|
Ohigashi I, Kozai M, Takahama Y. Development and developmental potential of cortical thymic epithelial cells. Immunol Rev 2016; 271:10-22. [DOI: 10.1111/imr.12404] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Izumi Ohigashi
- Division of Experimental Immunology; Institute for Genome Research; University of Tokushima; Tokushima Japan
| | - Mina Kozai
- Division of Experimental Immunology; Institute for Genome Research; University of Tokushima; Tokushima Japan
| | - Yousuke Takahama
- Division of Experimental Immunology; Institute for Genome Research; University of Tokushima; Tokushima Japan
| |
Collapse
|
183
|
Al‐Chami E, Tormo A, Pasquin S, Kanjarawi R, Ziouani S, Rafei M. Interleukin-21 administration to aged mice rejuvenates their peripheral T-cell pool by triggering de novo thymopoiesis. Aging Cell 2016; 15:349-60. [PMID: 26762709 PMCID: PMC4783337 DOI: 10.1111/acel.12440] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2015] [Indexed: 12/16/2022] Open
Abstract
The vaccination efficacy in the elderly is significantly reduced compared to younger populations due to thymic involution and age‐related intrinsic changes affecting their naïve T‐cell compartment. Interleukin (IL)‐21 was recently shown to display thymostimulatory properties. Therefore, we hypothesized that its administration to ageing hosts may improve T‐cell output and thus restore a competent peripheral T‐cell compartment. Indeed, an increase in the production of recent thymic emigrants (RTEs) attributable to intrathymic expansion of early thymic progenitors (ETPs), double‐negative (DN), and double‐positive (DP) thymocytes as well as thymic epithelial cell (TEC) was observed in recombinant (r)IL‐21‐treated aged mice. In sharp contrast, no alterations in the frequency of bone marrow (BM)‐derived progenitors were detected following rIL‐21 administration. Enhanced production of naïve T cells improved the T‐cell receptor (TCR) repertoire diversity and re‐established a pool of T cells exhibiting higher levels of miR‐181a and diminished amounts of the TCR‐inhibiting phosphatases SHP‐2 and DUSP5/6. As a result, stimulation of T cells derived from rIL‐21‐treated aged mice displayed enhanced activation of Lck, ZAP‐70, and ERK, which ultimately boosted their IL‐2 production, CD25 expression, and proliferation capabilities in comparison with T cells derived from control aged mice. Consequently, aged rIL‐21‐treated mice vaccinated using a tyrosinase‐related protein 2 (Trp2)‐derived peptide exhibited a substantial delay in B16 tumor growth and improved survival. The results of this study highlight the immunorestorative function of rIL‐21 paving its use as a strategy for the re‐establishment of effective immunity in the elderly.
Collapse
Affiliation(s)
- E. Al‐Chami
- Department of Pharmacology Université de Montréal Montréal QC H3C 1J7 Canada
| | - A. Tormo
- Department of Pharmacology Université de Montréal Montréal QC H3C 1J7 Canada
| | - S. Pasquin
- Department of Pharmacology Université de Montréal Montréal QC H3C 1J7 Canada
| | - R. Kanjarawi
- Department of Pharmacology Université de Montréal Montréal QC H3C 1J7 Canada
| | - S. Ziouani
- Université Paris‐Sud, Faculté de Pharmacie 5 rue J.B. Clément 92296 Châtenay‐Malabry Cedex France
| | - M. Rafei
- Department of Pharmacology Université de Montréal Montréal QC H3C 1J7 Canada
| |
Collapse
|
184
|
Alloantigen presentation and graft-versus-host disease: fuel for the fire. Blood 2016; 127:2963-70. [PMID: 27030390 DOI: 10.1182/blood-2016-02-697250] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/05/2016] [Indexed: 12/16/2022] Open
Abstract
Allogeneic stem cell transplantation (SCT) is a unique procedure, primarily in patients with hematopoietic malignancies, involving chemoradiotherapy followed by the introduction of donor hematopoietic and immune cells into an inflamed and lymphopenic environment. Interruption of the process by which recipient alloantigen is presented to donor T cells to generate graft-versus-host disease (GVHD) represents an attractive therapeutic strategy to prevent morbidity and mortality after SCT and has been increasingly studied in the last 15 years. However, the immune activation resulting in GVHD has no physiological equivalent in nature; alloantigen is ubiquitous, persists indefinitely, and can be presented by multiple cell types at numerous sites, often on incompatible major histocompatibility complex, and occurs in the context of intense inflammation early after SCT. The recognition that alloantigen presentation is also critical to the development of immunological tolerance via both deletional and regulatory mechanisms further adds to this complexity. Finally, GVHD itself appears capable of inhibiting the presentation of microbiological antigens by donor dendritic cells late after SCT that is mandatory for the establishment of effective pathogen-specific immunity. Here, we review our current understanding of alloantigen, its presentation by various antigen-presenting cells, subsequent recognition by donor T cells, and the potential of therapeutic strategies interrupting this disease-initiating process to modify transplant outcome.
Collapse
|
185
|
Ulyanchenko S, O'Neill KE, Medley T, Farley AM, Vaidya HJ, Cook AM, Blair NF, Blackburn CC. Identification of a Bipotent Epithelial Progenitor Population in the Adult Thymus. Cell Rep 2016; 14:2819-32. [PMID: 26997270 PMCID: PMC4819909 DOI: 10.1016/j.celrep.2016.02.080] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 12/28/2015] [Accepted: 02/21/2016] [Indexed: 11/16/2022] Open
Abstract
Thymic epithelial cells (TECs) are critically required for T cell development, but the cellular mechanisms that maintain adult TECs are poorly understood. Here, we show that a previously unidentified subpopulation, EpCam(+)UEA1(-)Ly-51(+)PLET1(+)MHC class II(hi), which comprises <0.5% of adult TECs, contains bipotent TEC progenitors that can efficiently generate both cortical (c) TECs and medullary (m) TECs. No other adult TEC population tested in this study contains this activity. We demonstrate persistence of PLET1(+)Ly-51(+) TEC-derived cells for 9 months in vivo, suggesting the presence of thymic epithelial stem cells. Additionally, we identify cTEC-restricted short-term progenitor activity but fail to detect high efficiency mTEC-restricted progenitors in the adult thymus. Our data provide a phenotypically defined adult thymic epithelial progenitor/stem cell that is able to generate both cTECs and mTECs, opening avenues for improving thymus function in patients.
Collapse
Affiliation(s)
- Svetlana Ulyanchenko
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, 5, Little France Drive, Edinburgh EH16 4UU, UK
| | - Kathy E O'Neill
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, 5, Little France Drive, Edinburgh EH16 4UU, UK
| | - Tanya Medley
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, 5, Little France Drive, Edinburgh EH16 4UU, UK
| | - Alison M Farley
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, 5, Little France Drive, Edinburgh EH16 4UU, UK
| | - Harsh J Vaidya
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, 5, Little France Drive, Edinburgh EH16 4UU, UK
| | - Alistair M Cook
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, 5, Little France Drive, Edinburgh EH16 4UU, UK
| | - Natalie F Blair
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, 5, Little France Drive, Edinburgh EH16 4UU, UK
| | - C Clare Blackburn
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, 5, Little France Drive, Edinburgh EH16 4UU, UK.
| |
Collapse
|
186
|
Velardi E, Dudakov JA, van den Brink MRM. Sex steroid ablation: an immunoregenerative strategy for immunocompromised patients. Bone Marrow Transplant 2016; 50 Suppl 2:S77-81. [PMID: 26039214 DOI: 10.1038/bmt.2015.101] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Age-related decline in thymic function is a well-described process that results in reduced T-cell development and thymic output of new naïve T cells. Thymic involution leads to reduced response to vaccines and new pathogens in otherwise healthy individuals; however, reduced thymic function is particularly detrimental in clinical scenarios where the immune system is profoundly depleted such as after chemotherapy, radiotherapy, infection and shock. Poor thymic function and restoration of immune competence has been correlated with an increased risk of opportunistic infections, tumor relapse and autoimmunity. Apart from their primary role in sex dimorphism, sex steroid levels profoundly affect the immune system in general and, in fact, age-related thymic involution has been at least partially attributed to the increase in sex steroids at puberty. Subsequently it has been demonstrated that the removal of sex steroids, or sex steroid ablation (SSA), triggers physiologic changes that ultimately lead to thymic re-growth and improved T-cell reconstitution in settings of hematopoietic stem cell transplant (HSCT). Although the cellular and molecular process underlying these regenerative effects are still poorly understood, SSA clearly represents an attractive therapeutic approach to enhance thymic function and restore immune competence in immunodeficient individuals.
Collapse
Affiliation(s)
- E Velardi
- 1] Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA [2] Department of Clinical and Experimental Medicine, University of Perugia, Perugia, Italy
| | - J A Dudakov
- 1] Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA [2] Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Melbourne, Victoria, Australia
| | - M R M van den Brink
- 1] Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA [2] Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
187
|
Yang Q, Bhandoola A. The development of adult innate lymphoid cells. Curr Opin Immunol 2016; 39:114-20. [PMID: 26871595 DOI: 10.1016/j.coi.2016.01.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 01/21/2023]
Abstract
Innate lymphoid cells (ILC) are a specialized family of effector lymphocytes that transcriptionally and functionally mirror effector subsets of T cells, but differ from T cells in that they lack clonally distributed adaptive antigen receptors. Our understanding of this family of lymphocytes is still in its infancy. In this review, we summarize current understanding and discuss recent insights into the cellular and molecular events that occur during early ILC development in adult mice. We discuss how these events overlap and diverge with the early development of adaptive T cells, and how they may influence the molecular and functional properties of mature ILC.
Collapse
Affiliation(s)
- Qi Yang
- T-Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Avinash Bhandoola
- T-Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States.
| |
Collapse
|
188
|
Bredenkamp N, Jin X, Liu D, O'Neill KE, Manley NR, Blackburn CC. Construction of a functional thymic microenvironment from pluripotent stem cells for the induction of central tolerance. Regen Med 2016; 10:317-29. [PMID: 25933240 DOI: 10.2217/rme.15.8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The thymus is required for generation of a self-tolerant, self-restricted T-cell repertoire. The capacity to manipulate or replace thymus function therapeutically would be beneficial in a variety of clinical settings, including for improving recovery following bone marrow transplantation, restoring immune system function in the elderly and promoting tolerance to transplanted organs or cells. An attractive strategy would be transplantation of thymus organoids generated from cells produced in vitro, for instance from pluripotent stem cells. Here, we review recent progress toward this goal, focusing on advances in directing differentiation of pluripotent stem cells to thymic epithelial cells, a key cell type of the thymic stroma, and related direct reprogramming strategies.
Collapse
Affiliation(s)
- Nicholas Bredenkamp
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, SCRM Building, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | | | | | | | | | | |
Collapse
|
189
|
Naumnik W, Naumnik B, Niklińska W, Ossolińska M, Chyczewska E. Clinical Implications of Hepatocyte Growth Factor, Interleukin-20, and Interleukin-22 in Serum and Bronchoalveolar Fluid of Patients with Non-Small Cell Lung Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 952:41-49. [PMID: 27573644 DOI: 10.1007/5584_2016_66] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hepatocyte growth factor (HGF) is involved in tumorigenesis, interleukin-20 (IL-20) is an inhibitor of angiogenesis, and interleukin-22 (IL-22) stimulates tumor growth. The aim of this study was to determine the level of HGF, IL-20, and IL-22 in both serum and bronchoalveolar lavage fluid (BALF) of non-small cell lung cancer (NSCLC) patients before onset of chemotherapy, the nature of the interrelationships between these markers, and their prognostic significance regarding post-chemotherapy survival time. We studied 46 NSCLC patients and 15 healthy subjects as a control group. We found significantly higher serum levels of HGF and IL-22 in the NSCLC patients than those in controls [pg/ml: HGF - 1911 (693-6510) vs. 1333 (838-3667), p = 0.0004; IL-22 - 10.66 (1.44-70.34) vs. 4.69 (0.35-12.29), p = 0.0007]. In contrast, concentrations of HGF and IL-22 in BALF were lower in NSCLC patients than those in controls [pg/ml: HGF - 72 (6-561) vs. 488 (14-2003), p = 0.0002; IL-22 - 2.28 (0.70-6.52) vs. 3.72 (2.76-5.64), p = 0.002]. In the NSCLC patients, there was a negative correlation between the serum level of IL-20 and time to tumor progression (r = -0.405, p = 0.04) and between the serum level of HGF and survival time (r = -0.41, p = 0.005). In addition, a higher serum level of HGF and a higher BALF level of IL-22 in patients were linked with a shorter overall survival. We conclude that HGF, IL-20, and IL-22 in the serum and BALF of NSCLC patients before chemotherapy may be a prognostic of cancer progression.
Collapse
Affiliation(s)
- W Naumnik
- Department of Lung Diseases, Medical University of Bialystok, 14 Zurawia Street, 15-540, Bialystok, Poland.
- Department of Clinical Molecular Biology, Medical University of Bialystok, Bialystok, Poland.
| | - B Naumnik
- First Department of Nephrology and Transplantation with Dialysis Unit, Medical University of Bialystok, Bialystok, Poland
| | - W Niklińska
- Department of Histology and Embryology, Medical University of Bialystok, Bialystok, Poland
| | - M Ossolińska
- Department of Lung Diseases, Medical University of Bialystok, 14 Zurawia Street, 15-540, Bialystok, Poland
| | - E Chyczewska
- Department of Lung Diseases, Medical University of Bialystok, 14 Zurawia Street, 15-540, Bialystok, Poland
| |
Collapse
|
190
|
Lindemans CA, Calafiore M, Mertelsmann AM, O'Connor MH, Dudakov JA, Jenq RR, Velardi E, Young LF, Smith OM, Lawrence G, Ivanov JA, Fu YY, Takashima S, Hua G, Martin ML, O'Rourke KP, Lo YH, Mokry M, Romera-Hernandez M, Cupedo T, Dow L, Nieuwenhuis EE, Shroyer NF, Liu C, Kolesnick R, van den Brink MRM, Hanash AM. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature 2015; 528:560-564. [PMID: 26649819 PMCID: PMC4720437 DOI: 10.1038/nature16460] [Citation(s) in RCA: 767] [Impact Index Per Article: 85.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 11/18/2015] [Indexed: 12/16/2022]
Abstract
Epithelial regeneration is critical for barrier maintenance and organ function after intestinal injury. The intestinal stem cell (ISC) niche provides Wnt, Notch and epidermal growth factor (EGF) signals supporting Lgr5(+) crypt base columnar ISCs for normal epithelial maintenance. However, little is known about the regulation of the ISC compartment after tissue damage. Using ex vivo organoid cultures, here we show that innate lymphoid cells (ILCs), potent producers of interleukin-22 (IL-22) after intestinal injury, increase the growth of mouse small intestine organoids in an IL-22-dependent fashion. Recombinant IL-22 directly targeted ISCs, augmenting the growth of both mouse and human intestinal organoids, increasing proliferation and promoting ISC expansion. IL-22 induced STAT3 phosphorylation in Lgr5(+) ISCs, and STAT3 was crucial for both organoid formation and IL-22-mediated regeneration. Treatment with IL-22 in vivo after mouse allogeneic bone marrow transplantation enhanced the recovery of ISCs, increased epithelial regeneration and reduced intestinal pathology and mortality from graft-versus-host disease. ATOH1-deficient organoid culture demonstrated that IL-22 induced epithelial regeneration independently of the Paneth cell niche. Our findings reveal a fundamental mechanism by which the immune system is able to support the intestinal epithelium, activating ISCs to promote regeneration.
Collapse
Affiliation(s)
- Caroline A Lindemans
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
- Department of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marco Calafiore
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Anna M Mertelsmann
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Margaret H O'Connor
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jarrod A Dudakov
- Department of Immunology, Memorial Sloan-Kettering Cancer Center, New York, New York
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Robert R Jenq
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Enrico Velardi
- Department of Immunology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Lauren F Young
- Department of Immunology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Odette M Smith
- Department of Immunology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Gillian Lawrence
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Juliet A Ivanov
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Ya-Yuan Fu
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Shuichiro Takashima
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Guoqiang Hua
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Maria L Martin
- Department of Molecular Pharmacology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Kevin P O'Rourke
- Department of Cancer Biology & Genetics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Yuan-Hung Lo
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Michal Mokry
- Department of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Tom Cupedo
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lukas Dow
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Edward E Nieuwenhuis
- Department of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Noah F Shroyer
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Chen Liu
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Richard Kolesnick
- Department of Molecular Pharmacology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Marcel R M van den Brink
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
- Department of Immunology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Alan M Hanash
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
191
|
Lucarelli B, Merli P, Bertaina V, Locatelli F. Strategies to accelerate immune recovery after allogeneic hematopoietic stem cell transplantation. Expert Rev Clin Immunol 2015; 12:343-58. [PMID: 26588325 DOI: 10.1586/1744666x.2016.1123091] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The interplay existing between immune reconstitution and patient outcome has been extensively demonstrated in allogeneic hematopoietic stem cell transplantation. One of the leading causes of infection-related mortality is the slow recovery of T-cell immunity due to the conditioning regimen and/or age-related thymus damage, poor naïve T-cell output, and restricted T-cell receptor (TCR) repertoires. With the aim of improving posttransplantation immune reconstitution, several immunotherapy approaches have been explored. Donor leukocyte infusions are widely used to accelerate immune recovery, but they carry the risk of provoking graft-versus-host disease. This review will focus on sophisticated strategies of thymus function-recovery, adoptive infusion of donor-derived, allodepleted T cells, T-cell lines/clones specific for life-threatening pathogens, regulatory T cells, and of T cells transduced with suicide genes.
Collapse
Affiliation(s)
- Barbarella Lucarelli
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy
| | - Pietro Merli
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy
| | - Valentina Bertaina
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy
| | - Franco Locatelli
- a Department of Pediatric Hematology-Oncology , IRCCS, Bambino Gesù Children's Hospital , Rome , Italy.,b Department of Pediatrics , University of Pavia , Pavia , Italy
| |
Collapse
|
192
|
Denkinger MD, Leins H, Schirmbeck R, Florian MC, Geiger H. HSC Aging and Senescent Immune Remodeling. Trends Immunol 2015; 36:815-824. [PMID: 26611154 PMCID: PMC4710174 DOI: 10.1016/j.it.2015.10.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 10/19/2015] [Accepted: 10/19/2015] [Indexed: 01/10/2023]
Abstract
Aging-associated changes in the function of the immune system are referred to as senescent immune remodeling (SIR). Here we review the current understanding on the cellular and molecular mechanisms underlying SIR. We focus on aging-associated changes in T and B cells, and discuss recent evidence supporting the notion that aging of the hematopoietic stem cell (HSC) compartment directly contributes to SIR due to aging-associated alterations in stem cell differentiation. We conclude by outlining strategies to attenuate SIR, including approaches to rejuvenate HSCs, which may open new avenues for targeting SIR in the clinic.
Collapse
Affiliation(s)
- Michael D Denkinger
- Institute for Molecular Medicine, Stem Cells and Aging, Ulm University, Ulm, Germany; aging research center, Ulm University, Ulm, Germany; AGAPLESION Bethesda Clinic, Geriatric Medicine, Ulm University, Ulm, Germany; Geriatric Center Ulm/Alb-Donau, Ulm, Germany
| | - Hanna Leins
- AGAPLESION Bethesda Clinic, Geriatric Medicine, Ulm University, Ulm, Germany; Department of Internal Medicine I, University Hospital of Ulm, Ulm, Germany
| | | | - Maria Carolina Florian
- Institute for Molecular Medicine, Stem Cells and Aging, Ulm University, Ulm, Germany; aging research center, Ulm University, Ulm, Germany
| | - Hartmut Geiger
- Institute for Molecular Medicine, Stem Cells and Aging, Ulm University, Ulm, Germany; aging research center, Ulm University, Ulm, Germany; Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
193
|
Avitabile S, Odorisio T, Madonna S, Eyerich S, Guerra L, Eyerich K, Zambruno G, Cavani A, Cianfarani F. Interleukin-22 Promotes Wound Repair in Diabetes by Improving Keratinocyte Pro-Healing Functions. J Invest Dermatol 2015; 135:2862-2870. [PMID: 26168231 DOI: 10.1038/jid.2015.278] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/08/2015] [Accepted: 06/18/2015] [Indexed: 11/09/2022]
Abstract
Impaired re-epithelialization, imbalanced expression of cytokines and growth factors, and vascular disease contribute to healing impairment in diabetes. IL-22, a pro-inflammatory cytokine mediating a cross-talk between immune system and epithelial cells, has been shown to have a role in repair processes. In this study we aimed to investigate IL-22 regenerative potential in the poor healing context of diabetic wounds. By using streptozotocin-induced diabetic mice, we demonstrated that IL-22 wound treatment significantly accelerated the healing process, by promoting re-epithelialization, granulation tissue formation, and vascularization. Improved re-epithelialization was associated with increased keratinocyte proliferation and signal transducer and activator of transcription 3 (STAT3) activation. We showed that endogenous IL-22 content was reduced at both mRNA and protein level during the inflammatory phase of diabetic wounds, with fewer IL-22-positive cells infiltrating the granulation tissue. We demonstrated that IL-22 treatment promoted proliferation and injury repair of hyperglycemic keratinocytes and induced activation of STAT3 and extracellular signal-regulated kinase transduction pathways in keratinocytes grown in hyperglycemic condition or isolated from diabetic patients. Finally, we demonstrated that IL-22 treatment was able to inhibit diabetic keratinocyte differentiation while promoting vascular endothelial growth factor release. Our data indicate a pro-healing role of IL-22 in diabetic wounds, suggesting a therapeutic potential for this cytokine in diabetic ulcer management.
Collapse
Affiliation(s)
- Simona Avitabile
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Teresa Odorisio
- Laboratory of Biochemistry, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Stefania Madonna
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Stefanie Eyerich
- ZAUM - Center of Allergy and Environment, Technische Universität and Helmholtz Center, Munich, Germany
| | - Liliana Guerra
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Kilian Eyerich
- Department of Dermatology and Allergy, Technische Universität, Munich, Germany
| | - Giovanna Zambruno
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy
| | - Andrea Cavani
- Laboratory of Experimental Immunology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy.
| | - Francesca Cianfarani
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata IRCCS, Rome, Italy.
| |
Collapse
|
194
|
Farese AM, Hankey KG, Cohen MV, MacVittie TJ. Lymphoid and Myeloid Recovery in Rhesus Macaques Following Total Body X-Irradiation. HEALTH PHYSICS 2015; 109:414-26. [PMID: 26425902 PMCID: PMC4593069 DOI: 10.1097/hp.0000000000000348] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Recovery from severe immunosuppression requires hematopoietic stem cell reconstitution and effective thymopoiesis to restore a functional immune cell repertoire. Herein, a model of immune cell reconstitution consequent to potentially lethal doses of irradiation is described, which may be valuable in evaluating potential medical countermeasures. Male rhesus macaques were total body irradiated by exposure to 6.00 Gy 250 kVp x-radiation (midline tissue dose, 0.13 Gy min), resulting in an approximate LD10/60 (n = 5/59). Animals received medical management, and hematopoietic and immune cell recovery was assessed (n ≤ 14) through 370 d post exposure. A subset of animals (n ≤ 8) was examined through 700 d. Myeloid recovery was assessed by neutrophil and platelet-related parameters. Lymphoid recovery was assessed by the absolute lymphocyte count and FACS-based phenotyping of B- and T-cell subsets. Recent thymic emigrants were identified by T cell receptor excision circle quantification. Severe neutropenia, lymphopenia, and thrombocytopenia resolved within 30 d. Total CD3+ cells μL required 60 d to reach values 60% of normal, followed by subsequent slow recovery to approximately normal by 180 d post irradiation. Recovery of CD3+4+ and CD3+8+ cell memory and naïve subsets were markedly different. Memory populations were ≥ 100% of normal by day 60, whereas naïve populations were only 57% normal at 180 d and never fully recovered to baseline post irradiation. Total (CD20+) B cells μL were within normal levels by 77 d post exposure. This animal model elucidates the variable T- and B-cell subset recovery kinetics after a potentially lethal dose of total-body irradiation that are dependent on marrow-derived stem and progenitor cell recovery, peripheral homeostatic expansion, and thymopoiesis.
Collapse
Affiliation(s)
- Ann M. Farese
- University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD
| | - Kim G. Hankey
- University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD
| | | | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD
| |
Collapse
|
195
|
Konya V, Mjösberg J. Innate lymphoid cells in graft-versus-host disease. Am J Transplant 2015; 15:2795-801. [PMID: 26228632 PMCID: PMC4973689 DOI: 10.1111/ajt.13394] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/20/2015] [Accepted: 05/23/2015] [Indexed: 01/25/2023]
Abstract
Innate lymphoid cells (ILC) are lymphocytes lacking rearranged antigen receptors such as those expressed by T and B cells. ILC are important effector and regulatory cells of the innate immune system, controlling lymphoid organogenesis, tissue inflammation, and homeostasis. The family of ILC consists of cytotoxic NK cells and the more recently described noncytotoxic group 1, 2, and 3 ILC. The classification of noncytotoxic ILC-in many aspects-mirrors that of T helper cells, which is based on the expression of master transcription factors and signature cytokines specific for each subset. The IL-22 producing RORγt(+) ILC3 subset was recently found to be critical in the prevention of intestinal graft-versus-host disease (GVHD) following allogeneic hematopoietic cell transplantation (HCT) via strengthening the intestinal mucosal barrier. In this review, we summarize the current view of the immunological functions of human noncytotoxic ILC subsets and discuss the potentially beneficial features of IL-22 producing ILC3 in improving allo-HCT efficacy by attenuating susceptibility to GVHD. In addition, we explore the possibility of other ILC subsets playing a role in GVHD.
Collapse
Affiliation(s)
- V Konya
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - J Mjösberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
196
|
Tateishi R, Akiyama N, Miyauchi M, Yoshinaga R, Sasanuma H, Kudo T, Shimbo M, Shinohara M, Obata K, Inoue JI, Shirakawa M, Shiba D, Asahara H, Yoshida N, Takahashi S, Morita H, Akiyama T. Hypergravity Provokes a Temporary Reduction in CD4+CD8+ Thymocyte Number and a Persistent Decrease in Medullary Thymic Epithelial Cell Frequency in Mice. PLoS One 2015; 10:e0141650. [PMID: 26513242 PMCID: PMC4626100 DOI: 10.1371/journal.pone.0141650] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 10/12/2015] [Indexed: 11/18/2022] Open
Abstract
Gravity change affects many immunological systems. We investigated the effects of hypergravity (2G) on murine thymic cells. Exposure of mice to 2G for three days reduced the frequency of CD4+CD8+ thymocytes (DP) and mature medullary thymic epithelial cells (mTECs), accompanied by an increment of keratin-5 and keratin-8 double-positive (K5+K8+) TECs that reportedly contain TEC progenitors. Whereas the reduction of DP was recovered by a 14-day exposure to 2G, the reduction of mature mTECs and the increment of K5+K8+ TEC persisted. Interestingly, a surgical lesion of the inner ear’s vestibular apparatus inhibited these hypergravity effects. Quantitative PCR analysis revealed that the gene expression of Aire and RANK that are critical for mTEC function and development were up-regulated by the 3-day exposure and subsequently down-regulated by the 14-day exposure to 2G. Unexpectedly, this dynamic change in mTEC gene expression was independent of the vestibular apparatus. Overall, data suggest that 2G causes a temporary reduction of DP and a persistent reduction of mature mTECs in a vestibular system-dependent manner, and also dysregulates mTEC gene expression without involving the vestibular system. These data might provide insight on the impact of gravity change on thymic functions during spaceflight and living.
Collapse
Affiliation(s)
- Ryosuke Tateishi
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
| | - Nobuko Akiyama
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
| | - Maki Miyauchi
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
| | - Riko Yoshinaga
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
| | - Hiroki Sasanuma
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- Laboratory of Developmental Genetics, Center for Experimental Medicine and Systems Biology, The University of Tokyo, Tokyo, Japan
| | - Takashi Kudo
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Miki Shimbo
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masahiro Shinohara
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- JST, PRESTO, Kawaguchi, Saitama, Japan
| | - Koji Obata
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- Department of Physiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Jun-ichiro Inoue
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masaki Shirakawa
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Tsukuba, Ibaraki, Japan
| | - Dai Shiba
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- JEM Utilization Center, Human Spaceflight Technology Directorate, JAXA, Tsukuba, Ibaraki, Japan
| | - Hiroshi Asahara
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Nobuaki Yoshida
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- Laboratory of Developmental Genetics, Center for Experimental Medicine and Systems Biology, The University of Tokyo, Tokyo, Japan
| | - Satoru Takahashi
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hironobu Morita
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- Department of Physiology, Gifu University Graduate School of Medicine, Gifu, Japan
- * E-mail: (TA); (HM)
| | - Taishin Akiyama
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Mouse epigenetics project, ISS/Kibo experiment, Japan Aerospace Exploration Agency, JAXA, Tsukuba, Japan
- * E-mail: (TA); (HM)
| |
Collapse
|
197
|
Peters CP, Mjösberg JM, Bernink JH, Spits H. Innate lymphoid cells in inflammatory bowel diseases. Immunol Lett 2015; 172:124-31. [PMID: 26470815 DOI: 10.1016/j.imlet.2015.10.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 09/28/2015] [Accepted: 10/07/2015] [Indexed: 12/29/2022]
Abstract
It is generally believed that inflammatory bowel diseases (IBD) are caused by an aberrant immune response to environmental triggers in genetically susceptible individuals. The exact contribution of the adaptive and innate immune system has not been elucidated. However, recent advances in treatments targeting key inflammatory mediators such as tumour necrosis factor highlight the crucial role of the innate immune system in IBD. Innate lymphoid cells (ILCs) have recently been identified to play an important role in immune mediated inflammatory diseases. In this review we recapitulate the current knowledge on ILCs in IBD.
Collapse
Affiliation(s)
- C P Peters
- Department of Gastroenterology and Hepatology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| | - J M Mjösberg
- Center for Infectious Medicine, Department of Medicine, Karolinska University Hospital Huddinge Karolinska Institutet, CIM, F59, S-14186 Stockholm, Sweden.
| | - J H Bernink
- Department of Celbiology and Histology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| | - H Spits
- Department of Celbiology and Histology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
198
|
Sun L, Sun C, Liang Z, Li H, Chen L, Luo H, Zhang H, Ding P, Sun X, Qin Z, Zhao Y. FSP1(+) fibroblast subpopulation is essential for the maintenance and regeneration of medullary thymic epithelial cells. Sci Rep 2015; 5:14871. [PMID: 26445893 PMCID: PMC4597222 DOI: 10.1038/srep14871] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/10/2015] [Indexed: 12/11/2022] Open
Abstract
Thymic epithelial cells (TECs) form a 3-dimentional network supporting thymocyte development and maturation. Besides epithelium and thymocytes, heterogeneous fibroblasts are essential components in maintaining thymic microenvironments. However, thymic fibroblast characteristics, development and function remain to be determined. We herein found that thymic non-hematopoietic CD45(-)FSP1(+) cells represent a unique Fibroblast specific protein 1 (FSP1)(-)fibroblast-derived cell subset. Deletion of these cells in FSP1-TK transgenic mice caused thymus atrophy due to the loss of TECs, especially mature medullary TECs (MHCII(high), CD80(+) and Aire(+)). In a cyclophosphamide-induced thymus injury and regeneration model, lack of non-hematopoietic CD45(-)FSP1(+) fibroblast subpopulation significantly delayed thymus regeneration. In fact, thymic FSP1(+) fibroblasts released more IL-6, FGF7 and FSP1 in the culture medium than their FSP1(-) counterparts. Further experiments showed that the FSP1 protein could directly enhance the proliferation and maturation of TECs in the in vitro culture systems. FSP1 knockout mice had significantly smaller thymus size and less TECs than their control. Collectively, our studies reveal that thymic CD45(-)FSP1(+) cells are a subpopulation of fibroblasts, which is crucial for the maintenance and regeneration of TECs especially medullary TECs through providing IL-6, FGF7 and FSP1.
Collapse
Affiliation(s)
- Lina Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chenming Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhanfeng Liang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hongran Li
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lin Chen
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Haiying Luo
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hongmei Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Pengbo Ding
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiaoning Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhihai Qin
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
199
|
Akiyama T, Tateishi R, Akiyama N, Yoshinaga R, Kobayashi TJ. Positive and Negative Regulatory Mechanisms for Fine-Tuning Cellularity and Functions of Medullary Thymic Epithelial Cells. Front Immunol 2015; 6:461. [PMID: 26441966 PMCID: PMC4568481 DOI: 10.3389/fimmu.2015.00461] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/24/2015] [Indexed: 01/10/2023] Open
Abstract
Self-tolerant T cells and regulatory T cells develop in the thymus. A wide variety of cell-cell interactions in the thymus is required for the differentiation, proliferation, and repertoire selection of T cells. Various secreted and cell surface molecules expressed in thymic epithelial cells (TECs) mediate these processes. Moreover, cytokines expressed by cells of hematopoietic origin regulate the cellularity of TECs. Tumor necrosis factor (TNF) family RANK ligand, lymphotoxin, and CD40 ligand, expressed in T cells and innate lymphoid cells (ILCs), promote the differentiation and proliferation of medullary TECs (mTECs) that play critical roles in the induction of immune tolerance. A recent study suggests that interleukin-22 (IL-22) produced by ILCs promotes regeneration of TECs after irradiation. Intriguingly, tumor growth factor-β and osteoprotegerin limit cellularity of mTECs, thereby attenuating regulatory T cell generation. We will review recent insights into the molecular basis for cell-cell interactions regulating differentiation and proliferation of mTECs and also discuss about a perspective on use of mathematical models for understanding this complicated system.
Collapse
Affiliation(s)
- Taishin Akiyama
- Division of Cellular and Molecular Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | - Ryosuke Tateishi
- Division of Cellular and Molecular Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | - Nobuko Akiyama
- Division of Cellular and Molecular Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | - Riko Yoshinaga
- Division of Cellular and Molecular Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | | |
Collapse
|
200
|
Khan IS, Park CY, Mavropoulos A, Shariat N, Pollack JL, Barczak AJ, Erle DJ, McManus MT, Anderson MS, Jeker LT. Identification of MiR-205 As a MicroRNA That Is Highly Expressed in Medullary Thymic Epithelial Cells. PLoS One 2015; 10:e0135440. [PMID: 26270036 PMCID: PMC4535774 DOI: 10.1371/journal.pone.0135440] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/22/2015] [Indexed: 11/30/2022] Open
Abstract
Thymic epithelial cells (TECs) support T cell development in the thymus. Cortical thymic epithelial cells (cTECs) facilitate positive selection of developing thymocytes whereas medullary thymic epithelial cells (mTECs) facilitate the deletion of self-reactive thymocytes in order to prevent autoimmunity. The mTEC compartment is highly dynamic with continuous maturation and turnover, but the genetic regulation of these processes remains poorly understood. MicroRNAs (miRNAs) are important regulators of TEC genetic programs since miRNA-deficient TECs are severely defective. However, the individual miRNAs important for TEC maintenance and function and their mechanisms of action remain unknown. Here, we demonstrate that miR-205 is highly and preferentially expressed in mTECs during both thymic ontogeny and in the postnatal thymus. This distinct expression is suggestive of functional importance for TEC biology. Genetic ablation of miR-205 in TECs, however, neither revealed a role for miR-205 in TEC function during homeostatic conditions nor during recovery from thymic stress conditions. Thus, despite its distinct expression, miR-205 on its own is largely dispensable for mTEC biology.
Collapse
Affiliation(s)
- Imran S. Khan
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
| | - Chong Y. Park
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- WM Keck Center for Noncoding RNAs, University of California San Francisco, San Francisco, California, United States of America
| | - Anastasia Mavropoulos
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Nikki Shariat
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- WM Keck Center for Noncoding RNAs, University of California San Francisco, San Francisco, California, United States of America
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, United States of America
| | - Joshua L. Pollack
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Andrea J. Barczak
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - David J. Erle
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Michael T. McManus
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- WM Keck Center for Noncoding RNAs, University of California San Francisco, San Francisco, California, United States of America
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, United States of America
| | - Mark S. Anderson
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (MSA); (LTJ)
| | - Lukas T. Jeker
- UCSF Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
- * E-mail: (MSA); (LTJ)
| |
Collapse
|