101
|
Balakrishnan A, Jama B, Morris GP. Endogenous co‐expression of two T cell receptors promotes lymphopenia‐induced proliferation via increased affinity for self‐antigen. J Leukoc Biol 2018; 104:1097-1104. [DOI: 10.1002/jlb.1ab0618-214rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 11/11/2022] Open
Affiliation(s)
- Amritha Balakrishnan
- Department of PathologyUniversity of California San Diego La Jolla California USA
| | - Burhan Jama
- Department of PathologyUniversity of California San Diego La Jolla California USA
| | - Gerald P. Morris
- Department of PathologyUniversity of California San Diego La Jolla California USA
| |
Collapse
|
102
|
Browning LM, Pietrzak M, Kuczma M, Simms CP, Kurczewska A, Refugia JM, Lowery DJ, Rempala G, Gutkin D, Ignatowicz L, Muranski P, Kraj P. TGF-β-mediated enhancement of T H17 cell generation is inhibited by bone morphogenetic protein receptor 1α signaling. Sci Signal 2018; 11:eaar2125. [PMID: 30154100 PMCID: PMC8713300 DOI: 10.1126/scisignal.aar2125] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
The cytokines of the transforming growth factor-β (TGF-β) family promote the growth and differentiation of multiple tissues, but the role of only the founding member, TGF-β, in regulating the immune responses has been extensively studied. TGF-β is critical to prevent the spontaneous activation of self-reactive T cells and sustain immune homeostasis. In contrast, in the presence of proinflammatory cytokines, TGF-β promotes the differentiation of effector T helper 17 (TH17) cells. Abrogating TGF-β receptor signaling prevents the development of interleukin-17 (IL-17)-secreting cells and protects mice from TH17 cell-mediated autoimmunity. We found that the receptor of another member of TGF-β family, bone morphogenetic protein receptor 1α (BMPR1α), regulates T helper cell activation. We found that the differentiation of TH17 cells from naive CD4+ T cells was inhibited in the presence of BMPs. Abrogation of BMPR1α signaling during CD4+ T cell activation induced a developmental program that led to the generation of inflammatory effector cells expressing large amounts of IL-17, IFN-γ, and TNF family cytokines and transcription factors defining the TH17 cell lineage. We found that TGF-β and BMPs cooperated to establish effector cell functions and the cytokine profile of activated CD4+ T cells. Together, our data provide insight into the immunoregulatory function of BMPs.
Collapse
Affiliation(s)
- Lauren M Browning
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Maciej Pietrzak
- Department of Biomedical Informatics, Ohio State University, Columbus, OH 43210, USA
| | - Michal Kuczma
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Colin P Simms
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Agnieszka Kurczewska
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Justin M Refugia
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Dustin J Lowery
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Grzegorz Rempala
- College of Public Health, Ohio State University, Columbus, OH 43210, USA
| | - Dmitriy Gutkin
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15240, USA
| | - Leszek Ignatowicz
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Pawel Muranski
- Columbia University Medical Center, New York, NY 10032, USA
| | - Piotr Kraj
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA.
| |
Collapse
|
103
|
Commensal microflora-induced T cell responses mediate progressive neurodegeneration in glaucoma. Nat Commun 2018; 9:3209. [PMID: 30097565 PMCID: PMC6086830 DOI: 10.1038/s41467-018-05681-9] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/18/2018] [Indexed: 01/18/2023] Open
Abstract
Glaucoma is the most prevalent neurodegenerative disease and a leading cause of blindness worldwide. The mechanisms causing glaucomatous neurodegeneration are not fully understood. Here we show, using mice deficient in T and/or B cells and adoptive cell transfer, that transient elevation of intraocular pressure (IOP) is sufficient to induce T-cell infiltration into the retina. This T-cell infiltration leads to a prolonged phase of retinal ganglion cell degeneration that persists after IOP returns to a normal level. Heat shock proteins (HSP) are identified as target antigens of T-cell responses in glaucomatous mice and human glaucoma patients. Furthermore, retina-infiltrating T cells cross-react with human and bacterial HSPs; mice raised in the absence of commensal microflora do not develop glaucomatous T-cell responses or the associated neurodegeneration. These results provide compelling evidence that glaucomatous neurodegeneration is mediated in part by T cells that are pre-sensitized by exposure to commensal microflora.
Collapse
|
104
|
Jones R, Cosway EJ, Willis C, White AJ, Jenkinson WE, Fehling HJ, Anderson G, Withers DR. Dynamic changes in intrathymic ILC populations during murine neonatal development. Eur J Immunol 2018; 48:1481-1491. [PMID: 29851080 PMCID: PMC6174991 DOI: 10.1002/eji.201847511] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/19/2018] [Accepted: 05/25/2018] [Indexed: 12/29/2022]
Abstract
Members of the innate lymphoid cell (ILC) family have been implicated in the development of thymic microenvironments and the recovery of this architecture after damage. However, a detailed characterization of this family in the thymus is lacking. To better understand the thymic ILC compartment, we have utilized multiple in vivo models including the fate mapping of inhibitor of DNA binding‐2 (Id2) expression and the use of Id2 reporter mice. Our data demonstrate that ILCs are more prominent immediately after birth, but were rapidly diluted as the T‐cell development program increased. As observed in the embryonic thymus, CCR6+NKp46− lymphoid tissue inducer (LTi) cells were the main ILC3 population present, but numbers of these cells swiftly declined in the neonate and ILC3 were barely detectable in adult thymus. This loss of ILC3 means ILC2 are the dominant ILC population in the thymus. Thymic ILC2 were able to produce IL‐5 and IL‐13, were located within the medulla, and did not result from ILC3 plasticity. Furthermore, in WT mice, thymic ILC2 express little RANKL (receptor activator of nuclear factor kappa‐B ligand) arguing that functionally, these cells provide different signals to LTi cells in the thymus. Collectively, these data reveal a dynamic switch in the ILC populations of the thymus during neonatal development.
Collapse
Affiliation(s)
- Rhys Jones
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Emilie J Cosway
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Claire Willis
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andrea J White
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - William E Jenkinson
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Hans J Fehling
- Institute of Immunology, University of Ulm, Ulm, Germany
| | - Graham Anderson
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - David R Withers
- Institute of Immunology & Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
105
|
Dann SM, Le CHY, Hanson EM, Ross MC, Eckmann L. Giardia Infection of the Small Intestine Induces Chronic Colitis in Genetically Susceptible Hosts. THE JOURNAL OF IMMUNOLOGY 2018; 201:548-559. [PMID: 29898958 DOI: 10.4049/jimmunol.1700824] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 05/07/2018] [Indexed: 01/01/2023]
Abstract
The lumen-dwelling protozoan Giardia is an important parasitic cause of diarrheal disease worldwide. Infection can persist over extended periods with minimal intestinal inflammation, suggesting that Giardia may attenuate host responses to ensure its survival, although clearance eventually occurs in most cases. IL-10 is an anti-inflammatory regulator critical for intestinal homeostasis and controlling host responses to bacterial exposure, yet its potential role in coordinating antiprotozoal host defense in the intestine is not known. In this study, we found that murine infection with the natural enteric pathogen Giardia muris induced a transient IL-10 response after 2-4 wk at the primary site of infection in the upper small intestine, but parasite colonization and eradication were not affected by the absence of the cytokine in gene-targeted mice. However, IL-10 was critical for controlling infection-associated immunological sequelae in the colon because severe and persistent diarrhea and colitis were observed in IL-10-deficient mice within 1-2 wk postinfection but not in uninfected littermate controls. Inflammation was characterized by epithelial hyperplasia, neutrophil and macrophage expansion, and Th1 induction and could be prevented by blockade of IL-12/IL-23 p40 but not depletion of CD11c+ dendritic cells. Furthermore, the intestinal microbiota underwent characteristic shifts in composition and was required for disease because antibiotics and loss of TLR signaling in MyD88-deficient mice protected against colitis. Together, our data suggest that transient infection by a luminal and seemingly noninflammatory pathogen can trigger sustained colitis in genetically susceptible hosts, which has broader implications for understanding postinfectious syndromes and other chronic intestinal inflammatory conditions.
Collapse
Affiliation(s)
- Sara M Dann
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Christine H Y Le
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Elaine M Hanson
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| | - Matthew C Ross
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030
| | - Lars Eckmann
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093; and
| |
Collapse
|
106
|
Wirasinha RC, Singh M, Archer SK, Chan A, Harrison PF, Goodnow CC, Daley SR. αβ T-cell receptors with a central CDR3 cysteine are enriched in CD8αα intraepithelial lymphocytes and their thymic precursors. Immunol Cell Biol 2018; 96:553-561. [PMID: 29726044 DOI: 10.1111/imcb.12047] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/09/2018] [Accepted: 03/23/2018] [Indexed: 01/04/2023]
Abstract
The thymus plays a crucial role in immune tolerance by exposing developing T cells (thymocytes) to a myriad of self-antigens. Strong T-cell receptor (TCR) engagement induces tolerance in self-reactive thymocytes by stimulating apoptosis or selection into specialized T-cell lineages, including intestinal TCRαβ+ CD8αα+ intraepithelial lymphocytes (IEL). TCR-intrinsic amino acid motifs that can be used to predict whether a TCR will be strongly self-reactive remain elusive. Here, a novel TCR sequence alignment approach revealed that T-cell lineages in C57BL/6 mice had divergent usage of cysteine within two positions of the amino acid at the apex of the complementarity-determining region 3 (CDR3) of the TCRα or TCRβ chain. Compared to pre-selection thymocytes, central CDR3 cysteine usage was increased in IEL and Type A IEL precursors (IELp) and markedly decreased in Foxp3+ regulatory T cells (T-reg) and naïve T cells. These findings reveal a TCR-intrinsic motif that distinguishes Type A IELp and IEL from T-reg and naïve T cells.
Collapse
Affiliation(s)
- Rushika C Wirasinha
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Mandeep Singh
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Stuart K Archer
- Monash Bioinformatics Platform, Monash University, Melbourne, VIC, 3800, Australia
| | - Anna Chan
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Paul F Harrison
- Monash Bioinformatics Platform, Monash University, Melbourne, VIC, 3800, Australia
| | - Christopher C Goodnow
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Stephen R Daley
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| |
Collapse
|
107
|
Gon S, Loosveld M, Crouzet T, Potier D, Bonnet M, Morin SO, Michel G, Vey N, Nunès JA, Malissen B, Roncagalli R, Nadel B, Payet-Bornet D. Fit αβ T-cell receptor suppresses leukemogenesis of Pten-deficient thymocytes. Haematologica 2018; 103:999-1007. [PMID: 29567770 PMCID: PMC6058769 DOI: 10.3324/haematol.2018.188359] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/15/2018] [Indexed: 12/25/2022] Open
Abstract
Signaling through the αβT cell receptor (TCR) is a crucial determinant of T-cell fate and can induce two opposite outcomes during thymocyte development: cell death or survival and differentiation. To date, the role played by T-cell receptor in the oncogenic transformation of developing T cells remains unclear. Here we show that human primary T-cell acute lymphoblastic leukemias expressing an αβT cell receptor are frequently deficient for phosphatase and tensin homolog protein (PTEN), and fail to respond strongly to T-cell receptor activation. Using Pten-deficient T-cell acute lymphoblastic leukemia mouse models, we confirm that T-cell receptor signaling is involved in leukemogenesis. We show that abrogation of T-cell receptor expression accelerated tumor onset, while enforced expression of a fit transgenic T-cell receptor led to the development of T-cell receptor-negative lymphoma and delayed tumorigenesis. We further demonstrate that pre-tumoral Pten-deficient thymocytes harboring fit T-cell receptors undergo early clonal deletion, thus preventing their malignant transformation, while cells with unfit T-cell receptors that should normally be deleted during positive selection, pass selection and develop T-cell acute lymphoblastic leukemias. Altogether, our data show that fit T-cell receptor signaling suppresses tumor development mediated by Pten loss-of-function and point towards a role of Pten in positive selection.
Collapse
Affiliation(s)
- Stéphanie Gon
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Marie Loosveld
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France.,APHM, Hôpital La Timone, Laboratoire d'Hématologie, CRCM, Marseille, France
| | - Thomas Crouzet
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Delphine Potier
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Mélanie Bonnet
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Stéphanie O Morin
- Aix-Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Gérard Michel
- APHM, Hôpital La Timone, Service d'Hématologie et d'Oncologie Pédiatrique, Marseille, France
| | - Norbert Vey
- Aix-Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France.,Institut Paoli-Calmettes, Hematology Department, Marseille, France
| | - Jacques A Nunès
- Aix-Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | | | | | - Bertrand Nadel
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | | |
Collapse
|
108
|
Abstract
The immune system is remarkably responsive to a myriad of invading microorganisms and provides continuous surveillance against tissue damage and developing tumor cells. To achieve these diverse functions, multiple soluble and cellular components must react in an orchestrated cascade of events to control the specificity, magnitude and persistence of the immune response. Numerous catabolic and anabolic processes are involved in this process, and prominent roles for l-arginine and l-glutamine catabolism have been described, as these amino acids serve as precursors of nitric oxide, creatine, agmatine, tricarboxylic acid cycle intermediates, nucleotides and other amino acids, as well as for ornithine, which is used to synthesize putrescine and the polyamines spermidine and spermine. Polyamines have several purported roles and high levels of polyamines are manifest in tumor cells as well in autoreactive B- and T-cells in autoimmune diseases. In the tumor microenvironment, l-arginine catabolism by both tumor cells and suppressive myeloid cells is known to dampen cytotoxic T-cell functions suggesting there might be links between polyamines and T-cell suppression. Here, we review studies suggesting roles of polyamines in normal immune cell function and highlight their connections to autoimmunity and anti-tumor immune cell function.
Collapse
Affiliation(s)
- Rebecca S Hesterberg
- University of South Florida Cancer Biology Graduate Program, University of South Florida, 4202 East Fowler Ave, Tampa, FL 33620, USA.
- Department Immunology, PharmD, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, 23033 SRB, Tampa, FL 33612, USA.
| | - John L Cleveland
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | - Pearlie K Epling-Burnette
- Department Immunology, PharmD, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, 23033 SRB, Tampa, FL 33612, USA.
| |
Collapse
|
109
|
Cerqueira-Rodrigues B, Mendes A, Correia-Neves M, Nobrega C. Ag85-focused T-cell immune response controls Mycobacterium avium chronic infection. PLoS One 2018; 13:e0193596. [PMID: 29499041 PMCID: PMC5834192 DOI: 10.1371/journal.pone.0193596] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/14/2018] [Indexed: 01/09/2023] Open
Abstract
CD4+ T cells are essential players for the control of mycobacterial infections. Several mycobacterial antigens have been identified for eliciting a relevant CD4+ T cell mediated-immune response, and numerous studies explored this issue in the context of Mycobacterium tuberculosis infection. Antigen 85 (Ag85), a highly conserved protein across Mycobacterium species, is secreted at the early phase of M. tuberculosis infection leading to the proliferation of Ag85-specific CD4+ T cells. However, in the context of Mycobacterium avium infection, little is known about the expression of this antigen and the elicited immune response. In the current work, we investigated if a T cell receptor (TCR) repertoire mostly, but not exclusively, directed at Ag85 is sufficient to mount a protective immune response against M. avium. We show that P25 mice, whose majority of T cells express a transgenic TCR specific for Ag85, control M. avium infection at the same level as wild type (WT) mice up to 20 weeks post-infection (wpi). During M. avium infection, Ag85 antigen is easily detected in the liver of 20 wpi mice by immunohistochemistry. In spite of the propensity of P25 CD4+ T cells to produce higher amounts of interferon-gamma (IFNγ) upon ex vivo stimulation, no differences in serum IFNγ levels are detected in P25 compared to WT mice, nor enhanced immunopathology is detected in P25 mice. These results indicate that a T cell response dominated by Ag85-specific T cells is appropriate to control M. avium infection with no signs of immunopathology.
Collapse
Affiliation(s)
- Bruno Cerqueira-Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Mendes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Claudia Nobrega
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal
- * E-mail:
| |
Collapse
|
110
|
Host defense against oral microbiota by bone-damaging T cells. Nat Commun 2018; 9:701. [PMID: 29453398 PMCID: PMC5816021 DOI: 10.1038/s41467-018-03147-6] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 01/24/2018] [Indexed: 12/18/2022] Open
Abstract
The immune system evolved to efficiently eradicate invading bacteria and terminate inflammation through balancing inflammatory and regulatory T-cell responses. In autoimmune arthritis, pathogenic TH17 cells induce bone destruction and autoimmune inflammation. However, whether a beneficial function of T-cell-induced bone damage exists is unclear. Here, we show that bone-damaging T cells have a critical function in the eradication of bacteria in a mouse model of periodontitis, which is the most common infectious disease. Bacterial invasion leads to the generation of specialized TH17 cells that protect against bacteria by evoking mucosal immune responses as well as inducing bone damage, the latter of which also inhibits infection by removing the tooth. Thus, bone-damaging T cells, which may have developed to stop local infection by inducing tooth loss, function as a double-edged sword by protecting against pathogens while also inducing skeletal tissue degradation.
Collapse
|
111
|
Dolence JJ, Kobayashi T, Iijima K, Krempski J, Drake LY, Dent AL, Kita H. Airway exposure initiates peanut allergy by involving the IL-1 pathway and T follicular helper cells in mice. J Allergy Clin Immunol 2017; 142:1144-1158.e8. [PMID: 29247716 DOI: 10.1016/j.jaci.2017.11.020] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/02/2017] [Accepted: 11/08/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Little is currently known regarding the immunologic mechanism(s) that initiate peanut allergy. Notably, peanut proteins have been detected in house dust, and their levels correlate with peanut allergy prevalence. OBJECTIVE This study aimed to develop a new mouse model for peanut allergy and to investigate the immunologic mechanisms involved in peanut allergen sensitization. METHODS To mimic environmental exposure, naive mice were exposed to peanut flour by inhalation for up to 4 weeks. We then analyzed serum levels of IgE antibody and challenged mice with peanut proteins. Immunological mechanisms involved in sensitization were analyzed using cytokine reporter mice, an adoptive cell transfer model, and gene knockout mice. RESULTS When exposed to peanut flour by inhalation, both BALB/c and C57BL/6 mice developed peanut allergy, as demonstrated by the presence of peanut-specific IgE antibodies and manifestation of acute anaphylaxis on challenge. A large number of follicular helper T (Tfh) cells were also detected in draining lymph nodes of allergic mice. These cells produced IL-4 and IL-21, and they more robustly promoted peanut-specific IgE production than Th2 cells did. Genetic depletion of Tfh cells decreased IgE antibody levels and protected mice from anaphylaxis, without affecting Th2 cells. Furthermore, peanut flour exposure increased lung levels of IL-1α and IL-1β, and mice deficient in the receptor for these cytokines showed a significant decrease in Tfh cells compared with in wild-type mice. CONCLUSIONS Tfh cells play a key role in peanut allergy, and the IL-1 pathway is involved in the Tfh response to peanut allergen exposure.
Collapse
Affiliation(s)
- Joseph J Dolence
- Department of Medicine and Immunology, Mayo Clinic Rochester, Rochester, Minn
| | - Takao Kobayashi
- Department of Medicine and Immunology, Mayo Clinic Rochester, Rochester, Minn
| | - Koji Iijima
- Department of Medicine and Immunology, Mayo Clinic Rochester, Rochester, Minn
| | - James Krempski
- Department of Medicine and Immunology, Mayo Clinic Rochester, Rochester, Minn; Mayo Graduate School, Rochester, Minn
| | - Li Y Drake
- Department of Medicine and Immunology, Mayo Clinic Rochester, Rochester, Minn
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Ind
| | - Hirohito Kita
- Department of Medicine and Immunology, Mayo Clinic Rochester, Rochester, Minn.
| |
Collapse
|
112
|
Muro R, Nitta T, Nakano K, Okamura T, Takayanagi H, Suzuki H. γδTCR recruits the Syk/PI3K axis to drive proinflammatory differentiation program. J Clin Invest 2017; 128:415-426. [PMID: 29202478 DOI: 10.1172/jci95837] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/31/2017] [Indexed: 12/14/2022] Open
Abstract
γδT cells produce inflammatory cytokines and have been implicated in the pathogenesis of cancer, infectious diseases, and autoimmunity. The T cell receptor (TCR) signal transduction that specifically regulates the development of IL-17-producing γδT (γδT17) cells largely remains unclear. Here, we showed that the receptor proximal tyrosine kinase Syk is essential for γδTCR signal transduction and development of γδT17 in the mouse thymus. Zap70, another tyrosine kinase essential for the development of αβT cells, failed to functionally substitute for Syk in the development of γδT17. Syk induced the activation of the PI3K/Akt pathway upon γδTCR stimulation. Mice deficient in PI3K signaling exhibited a complete loss of γδT17, without impaired development of IFN-γ-producing γδT cells. Moreover, γδT17-dependent skin inflammation was ameliorated in mice deficient in RhoH, an adaptor known to recruit Syk. Thus, we deciphered lineage-specific TCR signaling and identified the Syk/PI3K pathway as a critical determinant of proinflammatory γδT cell differentiation.
Collapse
Affiliation(s)
- Ryunosuke Muro
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba, Japan.,Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Tadashi Okamura
- Department of Laboratory Animal Medicine, and.,Section of Animal Models, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Harumi Suzuki
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba, Japan
| |
Collapse
|
113
|
Greczmiel U, Kräutler NJ, Pedrioli A, Bartsch I, Agnellini P, Bedenikovic G, Harker J, Richter K, Oxenius A. Sustained T follicular helper cell response is essential for control of chronic viral infection. Sci Immunol 2017; 2:2/18/eaam8686. [DOI: 10.1126/sciimmunol.aam8686] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 09/25/2017] [Indexed: 12/15/2022]
|
114
|
Guo Y, MacIsaac KD, Chen Y, Miller RJ, Jain R, Joyce-Shaikh B, Ferguson H, Wang IM, Cristescu R, Mudgett J, Engstrom L, Piers KJ, Baltus GA, Barr K, Zhang H, Mehmet H, Hegde LG, Hu X, Carter LL, Aicher TD, Glick G, Zaller D, Hawwari A, Correll CC, Jones DC, Cua DJ. Inhibition of RORγT Skews TCRα Gene Rearrangement and Limits T Cell Repertoire Diversity. Cell Rep 2017; 17:3206-3218. [PMID: 28009290 DOI: 10.1016/j.celrep.2016.11.073] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/20/2016] [Accepted: 11/23/2016] [Indexed: 02/08/2023] Open
Abstract
Recent studies have elucidated the molecular mechanism of RORγT transcriptional regulation of Th17 differentiation and function. RORγT was initially identified as a transcription factor required for thymopoiesis by maintaining survival of CD4+CD8+ (DP) thymocytes. While RORγ antagonists are currently being developed to treat autoimmunity, it remains unclear how RORγT inhibition may impact thymocyte development. In this study, we show that in addition to regulating DP thymocytes survival, RORγT also controls genes that regulate thymocyte migration, proliferation, and T cell receptor (TCR)α selection. Strikingly, pharmacological inhibition of RORγ skews TCRα gene rearrangement, limits T cell repertoire diversity, and inhibits development of autoimmune encephalomyelitis. Thus, targeting RORγT not only inhibits Th17 cell development and function but also fundamentally alters thymic-emigrant recognition of self and foreign antigens. The analysis of RORγ inhibitors has allowed us to gain a broader perspective of the diverse function of RORγT and its impact on T cell biology.
Collapse
Affiliation(s)
- Yanxia Guo
- Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, USA
| | - Kenzie D MacIsaac
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Yi Chen
- Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, USA
| | - Richard J Miller
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Renu Jain
- Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, USA
| | | | - Heidi Ferguson
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - I-Ming Wang
- Merck Research Laboratories, 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Razvan Cristescu
- Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, USA
| | - John Mudgett
- Merck Research Laboratories, 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Laura Engstrom
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kyle J Piers
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Gretchen A Baltus
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kenneth Barr
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Hongjun Zhang
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Huseyin Mehmet
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | | | - Xiao Hu
- Lycera Corp, 2600 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Laura L Carter
- Lycera Corp, 2600 Plymouth Road, Ann Arbor, MI 48109, USA
| | | | - Gary Glick
- Lycera Corp, 2600 Plymouth Road, Ann Arbor, MI 48109, USA
| | - Dennis Zaller
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Abbas Hawwari
- King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City Hospital, Ministry of National Guard Health Affairs, Mail Code 520, P.O. Box 6664, Al Hasa 31982, Kingdom of Saudi Arabia
| | - Craig C Correll
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Dallas C Jones
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Daniel J Cua
- Merck Research Laboratories, 901 California Avenue, Palo Alto, CA 94304, USA.
| |
Collapse
|
115
|
Ohta T, Yoshikawa S, Tabakawa Y, Yamaji K, Ishiwata K, Shitara H, Taya C, Oh-Hora M, Kawano Y, Miyake K, Yamanishi Y, Yonekawa H, Watanabe N, Kanuka H, Karasuyama H. Skin CD4 + Memory T Cells Play an Essential Role in Acquired Anti-Tick Immunity through Interleukin-3-Mediated Basophil Recruitment to Tick-Feeding Sites. Front Immunol 2017; 8:1348. [PMID: 29085376 PMCID: PMC5650685 DOI: 10.3389/fimmu.2017.01348] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/03/2017] [Indexed: 12/14/2022] Open
Abstract
Ticks, blood-sucking arthropods, serve as vectors for transmission of infectious diseases including Lyme borreliosis. After tick infestation, several animal species can develop resistance to subsequent infestations, reducing the risk of transmission. In a mouse model, basophils reportedly infiltrate tick-feeding sites during the second but not first infestation and play a crucial role in the expression of acquired tick resistance. However, the mechanism underlying basophil recruitment to the second tick-feeding site remains ill-defined. Here, we investigated cells and their products responsible for the basophil recruitment. Little or no basophil infiltration was detected in T-cell-deficient mice, and adoptive transfer of CD4+ but not CD8+ T cells reconstituted it. Il3 gene expression was highly upregulated at the second tick-feeding site, and adoptive transfer of interleukin-3 (IL-3)-sufficient but not IL-3-deficient CD4+ T cells conferred the basophil infiltration on T-cell-deficient mice, indicating that the CD4+ T-cell-derived IL-3 is essential for the basophil recruitment. Notably, IL-3+ resident CD4+ memory T cells were detected even before the second infestation in previously uninfested skin distant from the first tick-feeding site. Taken together, IL-3 produced locally by skin CD4+ memory T cells appears to play a crucial role in basophil recruitment to the second tick-feeding site.
Collapse
Affiliation(s)
- Takuya Ohta
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Soichiro Yoshikawa
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuya Tabakawa
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kayoko Yamaji
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kenji Ishiwata
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroshi Shitara
- Laboratory for Transgenic Technology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Choji Taya
- Laboratory for Transgenic Technology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masatsugu Oh-Hora
- Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yohei Kawano
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kensuke Miyake
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshinori Yamanishi
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiromichi Yonekawa
- Laboratory for Transgenic Technology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Bio-Oriented Technology Research Advancement Institution (BRAIN), Saitama, Japan
| | - Naohiro Watanabe
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hirotaka Kanuka
- Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Hajime Karasuyama
- Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
116
|
Nevers T, Salvador AM, Velazquez F, Ngwenyama N, Carrillo-Salinas FJ, Aronovitz M, Blanton RM, Alcaide P. Th1 effector T cells selectively orchestrate cardiac fibrosis in nonischemic heart failure. J Exp Med 2017; 214:3311-3329. [PMID: 28970239 PMCID: PMC5679176 DOI: 10.1084/jem.20161791] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 06/13/2017] [Accepted: 08/21/2017] [Indexed: 12/20/2022] Open
Abstract
Despite emerging data indicating a role for T cells in profibrotic cardiac repair and healing after ischemia, little is known about whether T cells directly impact cardiac fibroblasts (CFBs) to promote cardiac fibrosis (CF) in nonischemic heart failure (HF). Recently, we reported increased T cell infiltration in the fibrotic myocardium of nonischemic HF patients, as well as the protection from CF and HF in TCR-α-/- mice. Here, we report that T cells activated in such a context are mainly IFN-γ+, adhere to CFB, and induce their transition into myofibroblasts. Th1 effector cells selectively drive CF both in vitro and in vivo, whereas adoptive transfer of Th1 cells, opposite to activated IFN-γ-/- Th cells, partially reconstituted CF and HF in TCR-α-/- recipient mice. Mechanistically, Th1 cells use integrin α4 to adhere to and induce TGF-β in CFB in an IFN-γ-dependent manner. Our findings identify a previously unrecognized role for Th1 cells as integrators of perivascular CF and cardiac dysfunction in nonischemic HF.
Collapse
Affiliation(s)
- Tania Nevers
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Ane M Salvador
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Francisco Velazquez
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Njabulo Ngwenyama
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | | | - Mark Aronovitz
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | - Robert M Blanton
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | - Pilar Alcaide
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| |
Collapse
|
117
|
Yu Y, Feng X, Vieten G, Dippel S, Imvised T, Gueler F, Ure BM, Kuebler JF, Klemann C. Conventional alpha beta (αβ) T cells do not contribute to acute intestinal ischemia-reperfusion injury in mice. PLoS One 2017; 12:e0181326. [PMID: 28704542 PMCID: PMC5509314 DOI: 10.1371/journal.pone.0181326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/29/2017] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Ischemia-reperfusion injury (IRI) is associated with significant patient mortality and morbidity. The complex cascade of IRI is incompletely understood, but inflammation is known to be a key mediator. In addition to the predominant innate immune responses, previous research has also indicated that αβ T cells contribute to IRI in various organ models. The aim of this study was to clarify the role αβ T cells play in IRI to the gut. METHODS Adult wild-type (WT) and αβ T cell-deficient mice were subjected to acute intestinal IRI with 30min ischemia followed by 4h reperfusion. The gene expression of pro-inflammatory cytokines was measured by qPCR, and the influx of leukocyte subpopulations in the gut was assessed via flow cytometry and histology. Pro-inflammatory cytokines in the serum were measured, and transaminases were assessed as an indicator of distant organ IRI. RESULTS Intestinal IRI led to an increased expression of pro-inflammatory cytokines in the gut tissue and an influx of leukocytes that predominantly consisted of neutrophils and macrophages. Furthermore, intestinal IRI increased serum IL-6, TNF-α, and ALT/AST levels. The αβ T cell-deficient mice did not exhibit a more significant increase in pro-inflammatory cytokines in the gut or serum following IR than the WT mice. There was also no difference between WT- and αβ T cell-deficient mice in terms of neutrophil infiltration or macrophage activation. Furthermore, the increase in transaminases was equal in both groups indicating that the level of distant organ injury was comparable. CONCLUSION An increasing body of evidence demonstrates that αβ T cells play a key role in IRI. In the gut, however, αβ T cells are not pivotal in the first hours following acute IRI as deficiency does not impact cytokine production, neutrophil recruitment, macrophage activation, or distant organ injury. Thus, αβ T cells may be considered innocent bystanders during the acute phase of intestinal IRI.
Collapse
Affiliation(s)
- Yi Yu
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Xiaoyan Feng
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Gertrud Vieten
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Stephanie Dippel
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Tawan Imvised
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Faikah Gueler
- Department of Nephrology, Hannover Medical School, Hanover, Germany
| | - Benno M. Ure
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Jochen F. Kuebler
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
| | - Christian Klemann
- Department of Pediatric Surgery, Center of Surgery, Hannover Medical School, Hanover, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
118
|
Dong M, Artusa P, Kelly SA, Fournier M, Baldwin TA, Mandl JN, Melichar HJ. Alterations in the Thymic Selection Threshold Skew the Self-Reactivity of the TCR Repertoire in Neonates. THE JOURNAL OF IMMUNOLOGY 2017; 199:965-973. [PMID: 28659353 DOI: 10.4049/jimmunol.1602137] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/02/2017] [Indexed: 12/19/2022]
Abstract
Neonatal and adult T cells differ in their effector functions. Although it is known that cell-intrinsic differences in mature T cells contribute to this phenomenon, the factors involved remain unclear. Given emerging evidence that the binding strength of a TCR for self-peptide presented by MHC (self-pMHC) impacts T cell function, we sought to determine whether altered thymic selection influences the self-reactivity of the TCR repertoire during ontogeny. We found that conventional and regulatory T cell subsets in the thymus of neonates and young mice expressed higher levels of cell surface CD5, a surrogate marker for TCR avidity for self-pMHC, as compared with their adult counterparts, and this difference in self-reactivity was independent of the germline bias of the neonatal TCR repertoire. The increased binding strength of the TCR repertoire for self-pMHC in neonates was not solely due to reported defects in clonal deletion. Rather, our data suggest that thymic selection is altered in young mice such that thymocytes bearing TCRs with low affinity for self-peptide are not efficiently selected into the neonatal repertoire, and stronger TCR signals accompany both conventional and regulatory T cell selection. Importantly, the distinct levels of T cell self-reactivity reflect physiologically relevant differences based on the preferential expansion of T cells from young mice to fill a lymphopenic environment. Therefore, differences in thymic selection in young versus adult mice skew the TCR repertoire, and the relatively higher self-reactivity of the T cell pool may contribute to the distinct immune responses observed in neonates.
Collapse
Affiliation(s)
- Mengqi Dong
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada.,Department of Microbiology, Infectious Diseases, and Immunology, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Patricio Artusa
- Department of Physiology and McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Stephanie A Kelly
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and
| | - Marilaine Fournier
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada
| | - Troy A Baldwin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and
| | - Judith N Mandl
- Department of Physiology and McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Heather J Melichar
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada; .,Department of Medicine, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| |
Collapse
|
119
|
Abstract
Body weight regain often causes failure of obesity therapies while the underlying mechanism remains largely unknown. In this study, we report that immune cells, especially CD4+ T cells, mediate the ‘memory’ of previous obese status. In a weight gain-loss-regain model, we found that C57BL/6J mice with an obesity history showed a much faster rate of body weight regain. This obesity memory could last for at least 2 months after previously obese mice were kept at the same body weight as non-obese mice. Surprisingly, such obesity memory was abrogated by dexamethasone treatment, whereas immunodeficient Rag1−/− and H2A−/− mice failed to establish such memory. Rag1−/− mice repossessed the obesity memory when immune cells or CD4+ T cells isolated from previously obese mice were transferred. Furthermore, depletion of CD4+ T cells led to obesity memory ablation. Taken together, we conclude that CD4+ T cells mediate obesity memory and promote weight regain.
Collapse
|
120
|
Nitta T, Kochi Y, Muro R, Tomofuji Y, Okamura T, Murata S, Suzuki H, Sumida T, Yamamoto K, Takayanagi H. Human thymoproteasome variations influence CD8 T cell selection. Sci Immunol 2017; 2:2/12/eaan5165. [PMID: 28783658 DOI: 10.1126/sciimmunol.aan5165] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/24/2017] [Indexed: 12/13/2022]
Abstract
The proteasome is a multi-subunit protease complex essential for housekeeping protein degradation and the production of the major histocompatibility complex (MHC) class I-bound antigen peptides that are essential for recognition by CD8 T cells. MHC variations dramatically contribute to T cell selection and autoimmunity, but genetic variations of peptide processing machinery including proteasome genes have been poorly explored in this context. In the computational analysis of human proteasome gene variation, we documented that PSMB11 was highly enriched for nucleotide changes that interfere with protein function. This gene encodes β5t, a thymus-specific catalytic subunit that regulates positive selection of CD8 T cells by producing a distinct set of MHC class I-bound peptides. The introduction of PSMB11 variations into the mouse genome by genome-editing revealed that these variations impaired the development of CD8 T cells in vivo. One of the PSMB11 polymorphisms altered the CD8 T cell repertoire in mice and was associated with a higher risk of an autoimmune disease in humans. Our findings suggest that, in addition to the MHC haplotype, proteasome variations influence T cell repertoire selection and may contribute to the difference in individual susceptibility to autoimmunity.
Collapse
Affiliation(s)
- Takeshi Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Yuta Kochi
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan.,Research Team for Autoimmune Diseases, Research Program for Intractable Disease of the Ministry of Health, Labour and Welfare, Tokyo, Japan
| | - Ryunosuke Muro
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Yoshihiko Tomofuji
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan.,Section of Animal Models, Department of Infectious Diseases, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Shigeo Murata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Harumi Suzuki
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba 272-8516, Japan
| | - Takayuki Sumida
- Research Team for Autoimmune Diseases, Research Program for Intractable Disease of the Ministry of Health, Labour and Welfare, Tokyo, Japan.,Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan.,Research Team for Autoimmune Diseases, Research Program for Intractable Disease of the Ministry of Health, Labour and Welfare, Tokyo, Japan.,Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
121
|
GATA3 Abundance Is a Critical Determinant of T Cell Receptor β Allelic Exclusion. Mol Cell Biol 2017; 37:MCB.00052-17. [PMID: 28320875 DOI: 10.1128/mcb.00052-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/14/2017] [Indexed: 12/30/2022] Open
Abstract
Allelic exclusion describes the essential immunological process by which feedback repression of sequential DNA rearrangements ensures that only one autosome expresses a functional T or B cell receptor. In wild-type mammals, approximately 60% of cells have recombined the DNA of one T cell receptor β (TCRβ) V-to-DJ-joined allele in a functional configuration, while the second allele has recombined only the DJ sequences; the other 40% of cells have recombined the V to the DJ segments on both alleles, with only one of the two alleles predicting a functional TCRβ protein. Here we report that the transgenic overexpression of GATA3 leads predominantly to biallelic TCRβ gene (Tcrb) recombination. We also found that wild-type immature thymocytes can be separated into distinct populations based on intracellular GATA3 expression and that GATA3LO cells had almost exclusively recombined only one Tcrb locus (that predicted a functional receptor sequence), while GATA3HI cells had uniformly recombined both Tcrb alleles (one predicting a functional and the other predicting a nonfunctional rearrangement). These data show that GATA3 abundance regulates the recombination propensity at the Tcrb locus and provide new mechanistic insight into the historic immunological conundrum for how Tcrb allelic exclusion is mediated.
Collapse
|
122
|
El Khassawna T, Serra A, Bucher CH, Petersen A, Schlundt C, Könnecke I, Malhan D, Wendler S, Schell H, Volk HD, Schmidt-Bleek K, Duda GN. T Lymphocytes Influence the Mineralization Process of Bone. Front Immunol 2017; 8:562. [PMID: 28596766 PMCID: PMC5442173 DOI: 10.3389/fimmu.2017.00562] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/26/2017] [Indexed: 12/17/2022] Open
Abstract
Bone is a unique organ able to regenerate itself after injuries. This regeneration requires the local interplay between different biological systems such as inflammation and matrix formation. Structural reconstitution is initiated by an inflammatory response orchestrated by the host immune system. However, the individual role of T cells and B cells in regeneration and their relationship to bone tissue reconstitution remain unknown. Comparing bone and fracture healing in animals with and without mature T and B cells revealed the essential role of these immune cells in determining the tissue mineralization and thus the bone quality. Bone without mature T and B cells is stiffer when compared to wild-type bone thus lacking the elasticity that helps to absorb forces, thus preventing fractures. In-depth analysis showed dysregulations in collagen deposition and osteoblast distribution upon lack of mature T and B cells. These changes in matrix deposition have been correlated with T cells rather than B cells within this study. This work presents, for the first time, a direct link between immune cells and matrix formation during bone healing after fracture. It illustrates specifically the role of T cells in the collagen organization process and the lack thereof in the absence of T cells.
Collapse
Affiliation(s)
- Thaqif El Khassawna
- Experimental Trauma Surgery, Faculty of Medicine, Justus-Liebig University, Giessen, Germany
| | | | - Christian H Bucher
- Julius Wolff Institute, Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ansgar Petersen
- Julius Wolff Institute, Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Schlundt
- Julius Wolff Institute, Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ireen Könnecke
- Julius Wolff Institute, Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Deeksha Malhan
- Experimental Trauma Surgery, Faculty of Medicine, Justus-Liebig University, Giessen, Germany
| | - Sebastian Wendler
- Julius Wolff Institute, Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Hanna Schell
- Julius Wolff Institute, Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute, Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute, Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
123
|
Pelly VS, Coomes SM, Kannan Y, Gialitakis M, Entwistle LJ, Perez-Lloret J, Czieso S, Okoye IS, Rückerl D, Allen JE, Brombacher F, Wilson MS. Interleukin 4 promotes the development of ex-Foxp3 Th2 cells during immunity to intestinal helminths. J Exp Med 2017; 214:1809-1826. [PMID: 28507062 PMCID: PMC5460998 DOI: 10.1084/jem.20161104] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 01/03/2017] [Accepted: 03/31/2017] [Indexed: 12/31/2022] Open
Abstract
Pelly et al. use novel mouse reporter systems to show that a proportion of Th2 cells develop from Foxp3-expressing cells in an IL-4–dependent manner, highlighting the potential to subvert T reg cell–mediated suppression in favor of type 2 immunity. Immunity to intestinal helminth infections requires the rapid activation of T helper 2 cells (Th2 cells). However, simultaneous expansion of CD4+Foxp3+ regulatory T cells (T reg cells) impedes protective responses, resulting in chronic infections. The ratio between T reg and effector T cells can therefore determine the outcome of infection. The redifferentiation of T reg cells into Th cells has been identified in hyperinflammatory diseases. In this study, we asked whether ex–T reg Th2 cells develop and contribute to type-2 immunity. Using multigene reporter and fate-reporter systems, we demonstrate that a significant proportion of Th2 cells derive from Foxp3+ cells after Heligmosomoides polygyrus infection and airway allergy. Ex-Foxp3 Th2 cells exhibit characteristic Th2 effector functions and provide immunity to H. polygyrus. Through selective deletion of Il4ra on Foxp3+ cells, we further demonstrate IL-4 is required for the development of ex-Foxp3 Th2 cells. Collectively, our findings indicate that converting T reg cells into Th2 cells could concomitantly enhance Th2 cells and limit T reg cell–mediated suppression.
Collapse
Affiliation(s)
- Victoria S Pelly
- Allergy and Anti-Helminth Immunity Laboratory, Mill Hill Laboratory, The Francis Crick Institute, London NW7 1AA, England, UK
| | - Stephanie M Coomes
- Allergy and Anti-Helminth Immunity Laboratory, Mill Hill Laboratory, The Francis Crick Institute, London NW7 1AA, England, UK
| | - Yashaswini Kannan
- Allergy and Anti-Helminth Immunity Laboratory, Mill Hill Laboratory, The Francis Crick Institute, London NW7 1AA, England, UK
| | - Manolis Gialitakis
- Ahr Immunity Laboratory, Mill Hill Laboratory, The Francis Crick Institute, London NW7 1AA, England, UK
| | - Lewis J Entwistle
- Allergy and Anti-Helminth Immunity Laboratory, Mill Hill Laboratory, The Francis Crick Institute, London NW7 1AA, England, UK
| | - Jimena Perez-Lloret
- Allergy and Anti-Helminth Immunity Laboratory, Mill Hill Laboratory, The Francis Crick Institute, London NW7 1AA, England, UK
| | - Stephanie Czieso
- Allergy and Anti-Helminth Immunity Laboratory, Mill Hill Laboratory, The Francis Crick Institute, London NW7 1AA, England, UK
| | - Isobel S Okoye
- Allergy and Anti-Helminth Immunity Laboratory, Mill Hill Laboratory, The Francis Crick Institute, London NW7 1AA, England, UK
| | - Dominik Rückerl
- Faculty of Life Sciences (3IR), University of Manchester, Manchester M13 9PT, England, UK
| | - Judith E Allen
- Faculty of Life Sciences (3IR), University of Manchester, Manchester M13 9PT, England, UK
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology, University of Cape Town, Institute of Infectious Disease and Molecular Medicine and South African Medical Research Council, 7925 Cape Town, South Africa
| | - Mark S Wilson
- Allergy and Anti-Helminth Immunity Laboratory, Mill Hill Laboratory, The Francis Crick Institute, London NW7 1AA, England, UK
| |
Collapse
|
124
|
Shinde P, Liu W, Ménoret A, Luster AD, Vella AT. Optimal CD4 T cell priming after LPS-based adjuvanticity with CD134 costimulation relies on CXCL9 production. J Leukoc Biol 2017; 102:57-69. [PMID: 28432083 DOI: 10.1189/jlb.1a0616-261rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 03/29/2017] [Accepted: 04/02/2017] [Indexed: 12/29/2022] Open
Abstract
LPS is a powerful adjuvant, and although LPS-mediated TLR4 signaling has been exquisitely delineated, the in vivo mechanism of how TLR4 responses impact T cell priming is far less clear. Besides costimulation, TNF and type 1 IFN are dominant cytokines released after TLR4 activation and can shape T cell responses, but other downstream factors have not been examined extensively. Depending on context, we show that IFNαR1 blockade resulted in minor to major effects on specific CD4 T cell clonal expansion. To help explain these differences, it was hypothesized that IFNαR1 blockade would inhibit specific T cell migration by reducing chemokine receptor signaling, but specific CD4 T cells from IFNαR1-blocked mice were readily able to migrate in response to specific chemokines. Next, we examined downstream factors and found that type 1 IFN signaling was necessary for chemokine production, even when mice were immunized with specific Ag with LPS and CD134 costimulation. IFNαR1 signaling promoted CXCL9 and CXCL10 synthesis, suggesting that these chemokines might be involved in the LPS and CD134 costimulation response. After immunization, we show that CXCL9 blockade inhibited CD4 T cell accumulation in the liver but also in LNs, even in the presence of elevated serum IFN-β levels. Thus, whereas type 1 IFN might have direct effects on primed CD4 T cells, the downstream chemokines that play a role during migration also impact accumulation. In sum, CXCL9 production is a key benchmark for productive CD4 T cell vaccination strategies.
Collapse
Affiliation(s)
- Paurvi Shinde
- Department of Immunology, School of Medicine, University of Connecticut Health, Farmington, Connecticut, USA
| | - Wenhai Liu
- Department of Immunology, School of Medicine, University of Connecticut Health, Farmington, Connecticut, USA
| | - Antoine Ménoret
- Department of Immunology, School of Medicine, University of Connecticut Health, Farmington, Connecticut, USA.,Institute for Systems Genomics, University of Connecticut School of Medicine, Farmington, Connecticut, USA; and
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anthony T Vella
- Department of Immunology, School of Medicine, University of Connecticut Health, Farmington, Connecticut, USA;
| |
Collapse
|
125
|
Chen YT, Su YC, Chang ML, Tsai PF, Kung JT. Low-Level MHC Class II Expression Leads to Suboptimal Th Cell Response, Increased Autoaggression, and Heightened Cytokine Inducibility. THE JOURNAL OF IMMUNOLOGY 2017; 198:1928-1943. [DOI: 10.4049/jimmunol.1600967] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 12/26/2016] [Indexed: 01/27/2023]
|
126
|
Gfi1-Foxo1 axis controls the fidelity of effector gene expression and developmental maturation of thymocytes. Proc Natl Acad Sci U S A 2016; 114:E67-E74. [PMID: 27994150 DOI: 10.1073/pnas.1617669114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Double-positive (DP) thymocytes respond to intrathymic T-cell receptor (TCR) signals by undergoing positive selection and lineage differentiation into single-positive (SP) mature cells. Concomitant with these well-characterized events is the acquisition of a mature T-cell gene expression program characterized by the induction of the effector molecules IL-7Rα, S1P1, and CCR7, but the underlying mechanism remains elusive. We report here that transcription repressor Growth factor independent 1 (Gfi1) orchestrates the fidelity of the DP gene expression program and developmental maturation into SP cells. Loss of Gfi1 resulted in premature induction of effector genes and the transcription factors forkhead box protein O1 (Foxo1) and Klf2 in DP thymocytes and the accumulation of postselection intermediate populations and accelerated transition into SP cells. Strikingly, partial loss of Foxo1 function, but not restored survival fitness, rectified the dysregulated gene expression and thymocyte maturation in Gfi1-deficient mice. Our results establish the Gfi1-Foxo1 axis and the transcriptional circuitry that actively maintain DP identity and shape the proper generation of mature T cells.
Collapse
|
127
|
Wang Y, Sun SN, Liu Q, Yu YY, Guo J, Wang K, Xing BC, Zheng QF, Campa MJ, Patz EF, Li SY, He YW. Autocrine Complement Inhibits IL10-Dependent T-cell-Mediated Antitumor Immunity to Promote Tumor Progression. Cancer Discov 2016; 6:1022-35. [PMID: 27297552 DOI: 10.1158/2159-8290.cd-15-1412] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 06/06/2016] [Indexed: 12/23/2022]
Abstract
UNLABELLED In contrast to its inhibitory effects on many cells, IL10 activates CD8(+) tumor-infiltrating lymphocytes (TIL) and enhances their antitumor activity. However, CD8(+) TILs do not routinely express IL10, as autocrine complement C3 inhibits IL10 production through complement receptors C3aR and C5aR. CD8(+) TILs from C3-deficient mice, however, express IL10 and exhibit enhanced effector function. C3-deficient mice are resistant to tumor development in a T-cell- and IL10-dependent manner; human TILs expanded with IL2 plus IL10 increase the killing of primary tumors in vitro compared with IL2-treated TILs. Complement-mediated inhibition of antitumor immunity is independent of the programmed death 1/programmed death ligand 1 (PD-1/PD-L1) immune checkpoint pathway. Our findings suggest that complement receptors C3aR and C5aR expressed on CD8(+) TILs represent a novel class of immune checkpoints that could be targeted for tumor immunotherapy. Moreover, incorporation of IL10 in the expansion of TILs and in gene-engineered T cells for adoptive cell therapy enhances their antitumor efficacy. SIGNIFICANCE Our data suggest novel strategies to enhance immunotherapies: a combined blockade of complement signaling by antagonists to C3aR, C5aR, and anti-PD-1 to enhance anti-PD-1 efficacy; a targeted IL10 delivery to CD8(+) TILs using anti-PD-1-IL10 or anti-CTLA4-IL10 fusion proteins; and the addition of IL10 in TIL expansion for adoptive cellular therapy. Cancer Discov; 6(9); 1022-35. ©2016 AACR.See related commentary by Peng et al., p. 953This article is highlighted in the In This Issue feature, p. 932.
Collapse
Affiliation(s)
- Yu Wang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina
| | - Sheng-Nan Sun
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Qing Liu
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - Yang-Yang Yu
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - Jian Guo
- Department of Immunology, Duke University Medical Center, Durham, North Carolina
| | - Kun Wang
- Hepatopancreatobiliary Surgery Department, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Bao-Cai Xing
- Hepatopancreatobiliary Surgery Department, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Qing-Feng Zheng
- Thoracic Surgery Department, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, China
| | - Michael J Campa
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| | - Edward F Patz
- Department of Radiology, Duke University Medical Center, Durham, North Carolina. Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Shi-You Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Chaoyang District, Beijing, China
| | - You-Wen He
- Department of Immunology, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
128
|
Follicular helper T cells mediate IgE antibody response to airborne allergens. J Allergy Clin Immunol 2016; 139:300-313.e7. [PMID: 27325434 DOI: 10.1016/j.jaci.2016.04.021] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/18/2016] [Accepted: 04/25/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND TH2 cells have long been believed to play a pivotal role in allergic immune responses, including IgE antibody production and type 2 cytokine-mediated inflammation and pathology. A new T-cell subset, follicular helper T (TFH) cells, is specialized in supporting B-cell maturation and antibody production. OBJECTIVE We sought to investigate the roles of TFH cells in allergic immune responses. METHODS Naive mice were exposed to cytokines or natural allergens through the airways. Development of allergic immune responses was analyzed by collecting draining lymph nodes and sera and by challenging the animals. Cytokine reporter mice and gene-deficient mice were used to dissect the immunologic mechanisms. RESULTS We observed the development of IL-4-producing TFH cells and TH2 cells in draining lymph nodes after airway exposure to IL-1 family cytokines or natural allergens. TFH and TH2 cells demonstrated unique phenotypes, tissue localization, and cytokine responses. TFH cells supported the sustained production of IgE antibody in vivo in the absence of other T-cell subsets or even when TH2 cell functions were severely compromised. Conversely, conditional deficiency of the master regulator Bcl6 in CD4+ T cells resulted in a marked reduction in TFH cell numbers and IgE antibody levels, but type 2 cytokine responses and eosinophilic inflammation in the airways remained unaffected. CONCLUSION TFH cells play critical roles in the regulation of IgE antibody production. Allergic immune responses to airborne allergens likely involve 2 distinct subsets of IL-4-producing CD4+ T cells, namely TFH and Th2 cells.
Collapse
|
129
|
Intercellular Protein Transfer from Thymocytes to Thymic Epithelial Cells. PLoS One 2016; 11:e0152641. [PMID: 27022746 PMCID: PMC4811443 DOI: 10.1371/journal.pone.0152641] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/16/2016] [Indexed: 01/22/2023] Open
Abstract
Promiscuous expression of tissue restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs) is crucial for negative selection of self-reactive T cells to establish central tolerance. Intercellular transfer of self-peptide-MHC complexes from mTECs to thymic dendritic cells (DCs) allows DCs to acquire TRAs, which in turn contributes to negative selection and regulatory T cell generation. However, mTECs are unlikely to express all TRAs, such as immunoglobulins generated only in B cells after somatic recombination, hyper-mutation, or class-switches. We report here that both mTECs and cortical TECs can efficiently acquire not only cell surface but also intracellular proteins from thymocytes. This reveals a previously unappreciated intercellular sharing of molecules from thymocytes to TECs, which may broaden the TRA inventory in mTECs for establishing a full spectrum of central tolerance.
Collapse
|
130
|
Morphologic and Histologic Comparison of Hypertrophic Scar in Nude Mice, T-Cell Receptor, and Recombination Activating Gene Knockout Mice. Plast Reconstr Surg 2016; 136:1192-1204. [PMID: 26595016 DOI: 10.1097/prs.0000000000001782] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Proliferative scars in nude mice have demonstrated morphologic and histologic similarities to human hypertrophic scar. Gene knockout technology provides the opportunity to study the effect of deleting immune cells in various disease processes. The authors' objective was to test whether grafting human skin onto T-cell receptor (TCR) αβ-/-γδ-/-, recombination activating gene (RAG)-1-/-, and RAG-2γ-/-c-/- mice results in proliferative scars consistent with human hypertrophic scar and to characterize the morphologic, histologic, and cellular changes that occur after removing immune cells. METHODS Nude TCRαβ-/-γδ-/-, RAG-1-/-, and RAG-2-/-γc-/- mice (n = 20 per strain) were grafted with human skin and euthanized at 30, 60, 120, and 180 days. Controls (n = 5 per strain) were autografted with mouse skin. Scars and normal skin were harvested at each time point. Sections were stained with hematoxylin and eosin, Masson's trichrome, and immunohistochemistry for anti-human leukocyte antigen-ABC, α-smooth muscle actin, decorin, and biglycan. RESULTS TCRαβ-/-γδ-/-, RAG-1-/-, and RAG-2-/-γc-/- mice grafted with human skin developed firm, elevated scars with histologic and immunohistochemical similarities to human hypertrophic scar. Autografted controls showed no evidence of pathologic scarring. Knockout animals demonstrated a capacity for scar remodeling not observed in nude mice where reductions in α-smooth muscle actin staining pattern and scar thickness occurred over time. CONCLUSIONS Human skin transplanted onto TCRαβ-/-γδ-/-, RAG-1-/-, and RAG-2-/-γc-/- mice results in proliferative scars with morphologic and histologic features of human hypertrophic scar. Remodeling of proliferative scars generated in knockout animals is analogous to changes in human hypertrophic scar. These animal models may better represent the natural history of human hypertrophic scar.
Collapse
|
131
|
Bhattacharya A, Hegazy AN, Deigendesch N, Kosack L, Cupovic J, Kandasamy RK, Hildebrandt A, Merkler D, Kühl AA, Vilagos B, Schliehe C, Panse I, Khamina K, Baazim H, Arnold I, Flatz L, Xu HC, Lang PA, Aderem A, Takaoka A, Superti-Furga G, Colinge J, Ludewig B, Löhning M, Bergthaler A. Superoxide Dismutase 1 Protects Hepatocytes from Type I Interferon-Driven Oxidative Damage. Immunity 2016; 43:974-86. [PMID: 26588782 PMCID: PMC4658338 DOI: 10.1016/j.immuni.2015.10.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 05/29/2015] [Accepted: 08/03/2015] [Indexed: 12/23/2022]
Abstract
Tissue damage caused by viral hepatitis is a major cause of morbidity and mortality worldwide. Using a mouse model of viral hepatitis, we identified virus-induced early transcriptional changes in the redox pathways in the liver, including downregulation of superoxide dismutase 1 (Sod1). Sod1(-/-) mice exhibited increased inflammation and aggravated liver damage upon viral infection, which was independent of T and NK cells and could be ameliorated by antioxidant treatment. Type I interferon (IFN-I) led to a downregulation of Sod1 and caused oxidative liver damage in Sod1(-/-) and wild-type mice. Genetic and pharmacological ablation of the IFN-I signaling pathway protected against virus-induced liver damage. These results delineate IFN-I mediated oxidative stress as a key mediator of virus-induced liver damage and describe a mechanism of innate-immunity-driven pathology, linking IFN-I signaling with antioxidant host defense and infection-associated tissue damage. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Anannya Bhattacharya
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Ahmed N Hegazy
- Experimental Immunology, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany; Translational Gastroenterology Unit, Experimental Medicine Division Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, OX3 9DU Oxford, UK
| | - Nikolaus Deigendesch
- Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | - Lindsay Kosack
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Jovana Cupovic
- Institute of Immunobiology, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland
| | - Richard K Kandasamy
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Andrea Hildebrandt
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, Centre Médical Universitaire, 1 rue Michel Servet, 1211 Geneva, Switzerland; Department of Neuropathology, University Medicine Göttingen, Robert-Koch Strasse 40, 37099 Goettingen, Germany
| | - Anja A Kühl
- Department of Medicine I for Gastroenterology, Infectious Disease and Rheumatology, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200 Berlin, Germany
| | - Bojan Vilagos
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Christopher Schliehe
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Isabel Panse
- Experimental Immunology, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany
| | - Kseniya Khamina
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Hatoon Baazim
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Isabelle Arnold
- Translational Gastroenterology Unit, Experimental Medicine Division Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, OX3 9DU Oxford, UK
| | - Lukas Flatz
- Institute of Immunobiology, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland
| | - Haifeng C Xu
- Department of Gastroenterology, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Philipp A Lang
- Department of Gastroenterology, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany; Department of Molecular Medicine II, Heinrich Heine University, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Alan Aderem
- Seattle Biomedical Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, WA 98109-5219, USA
| | - Akinori Takaoka
- Division of Signaling in Cancer and Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido 060-0815, Japan
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria; Center for Physiology and Pharmacology, Medical University of Vienna, Lazarettgasse 14, 1090 Vienna, Austria
| | - Jacques Colinge
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria
| | - Burkhard Ludewig
- Institute of Immunobiology, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, 9007 St. Gallen, Switzerland
| | - Max Löhning
- Experimental Immunology, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; German Rheumatism Research Center (DRFZ), a Leibniz Institute, 10117 Berlin, Germany
| | - Andreas Bergthaler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT25.3, 1090 Vienna, Austria.
| |
Collapse
|
132
|
Moor K, Wotzka SY, Toska A, Diard M, Hapfelmeier S, Slack E. Peracetic Acid Treatment Generates Potent Inactivated Oral Vaccines from a Broad Range of Culturable Bacterial Species. Front Immunol 2016; 7:34. [PMID: 26904024 PMCID: PMC4749699 DOI: 10.3389/fimmu.2016.00034] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/24/2016] [Indexed: 12/28/2022] Open
Abstract
Our mucosal surfaces are the main sites of non-vector-borne pathogen entry, as well as the main interface with our commensal microbiota. We are still only beginning to understand how mucosal adaptive immunity interacts with commensal and pathogenic microbes to influence factors such as infectivity, phenotypic diversity, and within-host evolution. This is in part due to difficulties in generating specific mucosal adaptive immune responses without disrupting the mucosal microbial ecosystem itself. Here, we present a very simple tool to generate inactivated mucosal vaccines from a broad range of culturable bacteria. Oral gavage of 1010 peracetic acid-inactivated bacteria induces high-titer-specific intestinal IgA in the absence of any measurable inflammation or species invasion. As a proof of principle, we demonstrate that this technique is sufficient to provide fully protective immunity in the murine model of invasive non-typhoidal Salmonellosis, even in the face of severe innate immune deficiency.
Collapse
Affiliation(s)
- Kathrin Moor
- Institute for Microbiology, ETH Zürich , Zürich , Switzerland
| | - Sandra Y Wotzka
- Institute for Microbiology, ETH Zürich , Zürich , Switzerland
| | - Albulena Toska
- Institute for Microbiology, ETH Zürich , Zürich , Switzerland
| | - Médéric Diard
- Institute for Microbiology, ETH Zürich , Zürich , Switzerland
| | | | - Emma Slack
- Institute for Microbiology, ETH Zürich , Zürich , Switzerland
| |
Collapse
|
133
|
Kashem SW, Riedl MS, Yao C, Honda CN, Vulchanova L, Kaplan DH. Nociceptive Sensory Fibers Drive Interleukin-23 Production from CD301b+ Dermal Dendritic Cells and Drive Protective Cutaneous Immunity. Immunity 2016; 43:515-26. [PMID: 26377898 DOI: 10.1016/j.immuni.2015.08.016] [Citation(s) in RCA: 316] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/30/2015] [Accepted: 07/28/2015] [Indexed: 12/24/2022]
Abstract
Innate resistance to Candida albicans in mucosal tissues requires the production of interleukin-17A (IL-17A) by tissue-resident cells early during infection, but the mechanism of cytokine production has not been precisely defined. In the skin, we found that dermal γδ T cells were the dominant source of IL-17A during C. albicans infection and were required for pathogen resistance. Induction of IL-17A from dermal γδ T cells and resistance to C. albicans required IL-23 production from CD301b(+) dermal dendritic cells (dDCs). In addition, we found that sensory neurons were directly activated by C. albicans. Ablation of sensory neurons increased susceptibility to C. albicans infection, which could be rescued by exogenous addition of the neuropeptide CGRP. These data define a model in which nociceptive pathways in the skin drive production of IL-23 by CD301b(+) dDCs resulting in IL-17A production from γδ T cells and resistance to cutaneous candidiasis.
Collapse
MESH Headings
- Animals
- Candida albicans/immunology
- Candida albicans/physiology
- Candidiasis/genetics
- Candidiasis/immunology
- Candidiasis/microbiology
- Cells, Cultured
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dermis/cytology
- Flow Cytometry
- Host-Pathogen Interactions/immunology
- Immunity/genetics
- Immunity/immunology
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Interleukin-23/genetics
- Interleukin-23/immunology
- Interleukin-23/metabolism
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Mice, Inbred Strains
- Mice, Knockout
- Mice, Transgenic
- Oligonucleotide Array Sequence Analysis
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Calcitonin Gene-Related Peptide/genetics
- Receptors, Calcitonin Gene-Related Peptide/immunology
- Receptors, Calcitonin Gene-Related Peptide/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sensory Receptor Cells/immunology
- Sensory Receptor Cells/metabolism
- Skin/immunology
- Skin/metabolism
- Skin/microbiology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transcriptome/genetics
- Transcriptome/immunology
Collapse
Affiliation(s)
- Sakeen W Kashem
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Maureen S Riedl
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Chen Yao
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher N Honda
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lucy Vulchanova
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel H Kaplan
- Department of Dermatology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
134
|
Schuldt NJ, Auger JL, Hogquist KA, Binstadt BA. Bi-Allelic TCRα or β Recombination Enhances T Cell Development but Is Dispensable for Antigen Responses and Experimental Autoimmune Encephalomyelitis. PLoS One 2015; 10:e0145762. [PMID: 26693713 PMCID: PMC4687847 DOI: 10.1371/journal.pone.0145762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 12/08/2015] [Indexed: 01/13/2023] Open
Abstract
Dual TCRα-expressing T cells outnumber dual TCRβ-expressing cells by ~10:1. As a result, efforts to understand how dual TCR T cells impact immunity have focused on dual TCRα expression; dual TCRβ expression remains understudied. We recently demonstrated, however, that dual TCRβ expression accelerated disease in a TCR transgenic model of autoimmune arthritis through enhanced positive selection efficiency, indicating that dual TCRβ expression, though rare, can impact thymic selection. Here we generated mice hemizygous for TCRα, TCRβ, or both on the C57BL/6 background to investigate the impact bi-allelic TCR chain recombination has on T cell development, repertoire diversity, and autoimmunity. Lack of bi-allelic TCRα or TCRβ recombination reduced αβ thymocyte development efficiency, and the absence of bi-allelic TCRβ recombination promoted γδ T cell development. However, we observed no differences in the numbers of naïve and expanded antigen-specific T cells between TCRα+/-β+/- and wildtype mice, and TCR repertoire analysis revealed only subtle differences in Vβ gene usage. Finally, the absence of dual TCR T cells did not impact induced experimental autoimmune encephalomyelitis pathogenesis. Thus, despite more stringent allelic exclusion of TCRβ relative to TCRα, bi-allelic TCRβ expression can measurably impact thymocyte development and is necessary for maintaining normal αβ/γδ T cell proportions.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Gene Expression Regulation/immunology
- Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/immunology
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology
- Mice
- Mice, Knockout
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocytes/microbiology
Collapse
Affiliation(s)
- Nathaniel J. Schuldt
- Departments of Pediatrics, University of Minnesota, Minneapolis, United States of America
- Center for Immunology, University of Minnesota, Minneapolis, United States of America
| | - Jennifer L. Auger
- Departments of Pediatrics, University of Minnesota, Minneapolis, United States of America
- Center for Immunology, University of Minnesota, Minneapolis, United States of America
| | - Kristin A. Hogquist
- Departments of Pediatrics, University of Minnesota, Minneapolis, United States of America
- Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, United States of America
- Center for Immunology, University of Minnesota, Minneapolis, United States of America
| | - Bryce A. Binstadt
- Departments of Pediatrics, University of Minnesota, Minneapolis, United States of America
- Center for Immunology, University of Minnesota, Minneapolis, United States of America
- * E-mail:
| |
Collapse
|
135
|
Ghasemlou N, Chiu IM, Julien JP, Woolf CJ. CD11b+Ly6G- myeloid cells mediate mechanical inflammatory pain hypersensitivity. Proc Natl Acad Sci U S A 2015; 112:E6808-17. [PMID: 26598697 PMCID: PMC4679057 DOI: 10.1073/pnas.1501372112] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pain hypersensitivity at the site of inflammation as a result of chronic immune diseases, pathogenic infection, and tissue injury is a common medical condition. However, the specific contributions of the innate and adaptive immune system to the generation of pain during inflammation have not been systematically elucidated. We therefore set out to characterize the cellular and molecular immune response in two widely used preclinical models of inflammatory pain: (i) intraplantar injection of complete Freund's adjuvant (CFA) as a model of adjuvant- and pathogen-based inflammation and (ii) a plantar incisional wound as a model of tissue injury-based inflammation. Our findings reveal differences in temporal patterns of immune cell recruitment and activation states, cytokine production, and pain in these two models, with CFA causing a nonresolving granulomatous inflammatory response whereas tissue incision induced resolving immune and pain responses. These findings highlight the significant differences and potential clinical relevance of the incisional wound model compared with the CFA model. By using various cell-depletion strategies, we find that, whereas lymphocyte antigen 6 complex locus G (Ly)6G(+)CD11b(+) neutrophils and T-cell receptor (TCR) β(+) T cells do not contribute to the development of thermal or mechanical pain hypersensitivity in either model, proliferating CD11b(+)Ly6G(-) myeloid cells were necessary for mechanical hypersensitivity during incisional pain, and, to a lesser extent, CFA-induced inflammation. However, inflammatory (CCR2(+)Ly6C(hi)) monocytes were not responsible for these effects. The finding that a population of proliferating CD11b(+)Ly6G(-) myeloid cells contribute to mechanical inflammatory pain provides a potential cellular target for its treatment in wound inflammation.
Collapse
Affiliation(s)
- Nader Ghasemlou
- F. M. Kirby Neurobiology Center, Boston Children's Hospital & Harvard Medical School, Boston, MA 02115
| | - Isaac M Chiu
- F. M. Kirby Neurobiology Center, Boston Children's Hospital & Harvard Medical School, Boston, MA 02115; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Jean-Pierre Julien
- Research Centre of Institut Universitaire en Santé Mentale de Québec and Department of Psychiatry and Neuroscience, Laval University, Quebec City, QC, Canada G1J 2G3
| | - Clifford J Woolf
- F. M. Kirby Neurobiology Center, Boston Children's Hospital & Harvard Medical School, Boston, MA 02115;
| |
Collapse
|
136
|
Horai R, Zárate-Bladés CR, Dillenburg-Pilla P, Chen J, Kielczewski JL, Silver PB, Jittayasothorn Y, Chan CC, Yamane H, Honda K, Caspi RR. Microbiota-Dependent Activation of an Autoreactive T Cell Receptor Provokes Autoimmunity in an Immunologically Privileged Site. Immunity 2015; 43:343-53. [PMID: 26287682 DOI: 10.1016/j.immuni.2015.07.014] [Citation(s) in RCA: 280] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 04/30/2015] [Accepted: 05/21/2015] [Indexed: 12/30/2022]
Abstract
Activated retina-specific T cells that have acquired the ability to break through the blood-retinal barrier are thought to be causally involved in autoimmune uveitis, a major cause of human blindness. It is unclear where these autoreactive T cells first become activated, given that their cognate antigens are sequestered within the immune-privileged eye. We demonstrate in a novel mouse model of spontaneous uveitis that activation of retina-specific T cells is dependent on gut commensal microbiota. Retina-specific T cell activation involved signaling through the autoreactive T cell receptor (TCR) in response to non-cognate antigen in the intestine and was independent of the endogenous retinal autoantigen. Our findings not only have implications for the etiology of human uveitis, but also raise the possibility that activation of autoreactive TCRs by commensal microbes might be a more common trigger of autoimmune diseases than is currently appreciated.
Collapse
Affiliation(s)
- Reiko Horai
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA
| | | | - Patricia Dillenburg-Pilla
- Oral and Pharyngeal Cancer Branch, National Institutes of Dental and Craniofacial Research, NIH, Bethesda, MD 20892, USA
| | - Jun Chen
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA; State Key of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | | | - Phyllis B Silver
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA
| | | | - Chi-Chao Chan
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Hidehiro Yamane
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Kenya Honda
- Keio University School of Medicine, Tokyo 160-8582, Japan; RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
137
|
Di Giovangiulio M, Stakenborg N, Bosmans G, Meroni E, Farro G, Gomez-Pinilla PJ, Depoortere I, Boeckxstaens GE, Matteoli G. Ghrelin receptor modulates T helper cells during intestinal inflammation. Neurogastroenterol Motil 2015; 27:1542-52. [PMID: 26227790 DOI: 10.1111/nmo.12640] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 06/23/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND The orexigenic peptide ghrelin has anti-inflammatory properties in colitis, however, the mechanism of action and the immune cells targeted remain still to be elucidated. Here, we assessed the possible effect of ghrelin on T helper (Th) cells in a T cell transfer model of chronic colitis. METHODS Disease was induced in the recombination activating gene 1 knockout mice (Rag1(-/-) ) by adoptive transfer of naïve Th cells from ghrelin receptor knockout mice (GRLN-R(-/-) ) or littermate wild-type (WT) mice. The course and severity of colitis was assessed by monitoring body weight, diarrhea score, histological analysis, gene expression, and flow cytometry analysis. The possible effects of ghrelin on Th cell proliferation, polarization, and apoptosis was examined in vitro. KEY RESULTS Our data showed that Rag1(-/-) mice injected with GRLN-R(-/-) Th cells displayed increased severity of colitis compared to mice injected with WT Th cells. In addition, Rag1(-/-) mice injected with GRLN-R(-/-) Th cells had significantly higher intestinal inflammation and increased accumulation of Th1 and Th17 cells in the colon. In vitro, ghrelin directly affected proliferation of Th cells and induced apoptosis whereas it did not influence Th cell polarization. CONCLUSION & INFERENCES Our observations suggest that ghrelin modulates Th effector cells in the gut controlling proliferation and inducing apoptosis. Our findings further support the use of ghrelin as a novel therapeutic option to treat intestinal inflammatory diseases.
Collapse
Affiliation(s)
- M Di Giovangiulio
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - N Stakenborg
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G Bosmans
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - E Meroni
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G Farro
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - P J Gomez-Pinilla
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - I Depoortere
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G E Boeckxstaens
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G Matteoli
- Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| |
Collapse
|
138
|
Kara EE, McKenzie DR, Bastow CR, Gregor CE, Fenix KA, Ogunniyi AD, Paton JC, Mack M, Pombal DR, Seillet C, Dubois B, Liston A, MacDonald KPA, Belz GT, Smyth MJ, Hill GR, Comerford I, McColl SR. CCR2 defines in vivo development and homing of IL-23-driven GM-CSF-producing Th17 cells. Nat Commun 2015; 6:8644. [PMID: 26511769 PMCID: PMC4639903 DOI: 10.1038/ncomms9644] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/15/2015] [Indexed: 12/22/2022] Open
Abstract
IL-17-producing helper T (Th17) cells are critical for host defense against extracellular pathogens but also drive numerous autoimmune diseases. Th17 cells that differ in their inflammatory potential have been described including IL-10-producing Th17 cells that are weak inducers of inflammation and highly inflammatory, IL-23-driven, GM-CSF/IFNγ-producing Th17 cells. However, their distinct developmental requirements, functions and trafficking mechanisms in vivo remain poorly understood. Here we identify a temporally regulated IL-23-dependent switch from CCR6 to CCR2 usage by developing Th17 cells that is critical for pathogenic Th17 cell-driven inflammation in experimental autoimmune encephalomyelitis (EAE). This switch defines a unique in vivo cell surface signature (CCR6−CCR2+) of GM-CSF/IFNγ-producing Th17 cells in EAE and experimental persistent extracellular bacterial infection, and in humans. Using this signature, we identify an IL-23/IL-1/IFNγ/TNFα/T-bet/Eomesodermin-driven circuit driving GM-CSF/IFNγ-producing Th17 cell formation in vivo. Thus, our data identify a unique cell surface signature, trafficking mechanism and T-cell intrinsic regulators of GM-CSF/IFNγ-producing Th17 cells. Little is known regarding migration of Th17 cells that produce distinct cytokines implicated in protection and pathology. Kara et al. show that a switch from CCR6 to CCR2 by Th17 cells defines a signature (CCR6−CCR2+) of GM-CSF+ Th17 cells and drives pathology in a mouse model of autoimmunity.
Collapse
Affiliation(s)
- Ervin E Kara
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Duncan R McKenzie
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Cameron R Bastow
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Carly E Gregor
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Kevin A Fenix
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Abiodun D Ogunniyi
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia.,Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - James C Paton
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia.,Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg 93042, Germany
| | - Diana R Pombal
- Department of Microbiology and Immunology, VIB and University of Leuven, B-3000 Leuven, Belgium
| | - Cyrill Seillet
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Bénédicte Dubois
- Department of Neurosciences, KU-Leuven-University of Leuven, B-3000 Leuven, Belgium
| | - Adrian Liston
- Department of Microbiology and Immunology, VIB and University of Leuven, B-3000 Leuven, Belgium
| | - Kelli P A MacDonald
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Gabrielle T Belz
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia.,School of Medicine, University of Queensland, Herston, Queensland 4006, Australia
| | - Geoffrey R Hill
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia.,The Royal Brisbane and Women's Hospital, Herston, Queensland 4029, Australia
| | - Iain Comerford
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Shaun R McColl
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia.,Centre for Molecular Pathology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
139
|
Mayeux J, Skaug B, Luo W, Russell LM, John S, Saelee P, Abbasi H, Li QZ, Garrett-Sinha LA, Satterthwaite AB. Genetic Interaction between Lyn, Ets1, and Btk in the Control of Antibody Levels. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26209625 DOI: 10.4049/jimmunol.1500165] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tight control of B cell differentiation into plasma cells (PCs) is critical for proper immune responses and the prevention of autoimmunity. The Ets1 transcription factor acts in B cells to prevent PC differentiation. Ets1(-/-) mice accumulate PCs and produce autoantibodies. Ets1 expression is downregulated upon B cell activation through the BCR and TLRs and is maintained by the inhibitory signaling pathway mediated by Lyn, CD22 and SiglecG, and SHP-1. In the absence of these inhibitory components, Ets1 levels are reduced in B cells in a Btk-dependent manner. This leads to increased PCs, autoantibodies, and an autoimmune phenotype similar to that of Ets1(-/-) mice. Defects in inhibitory signaling molecules, including Lyn and Ets1, are associated with human lupus, although the effects are more subtle than the complete deficiency that occurs in knockout mice. In this study, we explore the effect of partial disruption of the Lyn/Ets1 pathway on B cell tolerance and find that Lyn(+/-)Ets1(+/-) mice demonstrate greater and earlier production of IgM, but not IgG, autoantibodies compared with Lyn(+/-) or Ets1(+/-) mice. We also show that Btk-dependent downregulation of Ets1 is important for normal PC homeostasis when inhibitory signaling is intact. Ets1 deficiency restores the decrease in steady state PCs and Ab levels observed in Btk(-/-) mice. Thus, depending on the balance of activating and inhibitory signals to Ets1, there is a continuum of effects on autoantibody production and PC maintenance. This ranges from full-blown autoimmunity with complete loss of Ets1-maintaining signals to reduced PC and Ab levels with impaired Ets1 downregulation.
Collapse
Affiliation(s)
- Jessica Mayeux
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Brian Skaug
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Wei Luo
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203; and
| | - Lisa M Russell
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203; and
| | - Shinu John
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203; and
| | - Prontip Saelee
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203; and
| | - Hansaa Abbasi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Quan-Zhen Li
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Lee Ann Garrett-Sinha
- Department of Biochemistry, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203; and
| | - Anne B Satterthwaite
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390; Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
140
|
Becker AM, Callahan DJ, Richner JM, Choi J, DiPersio JF, Diamond MS, Bhattacharya D. GPR18 Controls Reconstitution of Mouse Small Intestine Intraepithelial Lymphocytes following Bone Marrow Transplantation. PLoS One 2015. [PMID: 26197390 PMCID: PMC4510063 DOI: 10.1371/journal.pone.0133854] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Specific G protein coupled receptors (GPRs) regulate the proper positioning, function, and development of immune lineage subsets. Here, we demonstrate that GPR18 regulates the reconstitution of intraepithelial lymphocytes (IELs) of the small intestine following bone marrow transplantation. Through analysis of transcriptional microarray data, we find that GPR18 is highly expressed in IELs, lymphoid progenitors, and mature follicular B cells. To establish the physiological role of this largely uncharacterized GPR, we generated Gpr18-/- mice. Despite high levels of GPR18 expression in specific hematopoietic progenitors, Gpr18-/- mice have no defects in lymphopoiesis or myelopoiesis. Moreover, antibody responses following immunization with hapten-protein conjugates or infection with West Nile virus are normal in Gpr18-/- mice. Steady-state numbers of IELs are also normal in Gpr18-/- mice. However, competitive bone marrow reconstitution experiments demonstrate that GPR18 is cell-intrinsically required for the optimal restoration of small intestine TCRγδ+ and TCRαβ+ CD8αα+ IELs. In contrast, GPR18 is dispensable for the reconstitution of large intestine IELs. Moreover, Gpr18-/- bone marrow reconstitutes small intestine IELs similarly to controls in athymic recipients. Gpr18-/- chimeras show no changes in susceptibility to intestinal insults such as Citrobacter rodentium infections or graft versus host disease. These data reveal highly specific requirements for GPR18 in the development and reconstitution of thymus-derived intestinal IEL subsets in the steady-state and after bone marrow transplantation.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/cytology
- Bone Marrow Cells/cytology
- Bone Marrow Transplantation
- Citrobacter
- Female
- Graft vs Host Disease
- Hematopoietic Stem Cells/cytology
- Intestinal Mucosa/metabolism
- Intestine, Small/metabolism
- Lymphocytes/cytology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Myelopoiesis
- Oligonucleotide Array Sequence Analysis
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Thymus Gland/metabolism
- Transplantation, Homologous
- West Nile virus
Collapse
Affiliation(s)
- Amy M. Becker
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Derrick J. Callahan
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Justin M. Richner
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jaebok Choi
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Division of Oncology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - John F. DiPersio
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Division of Oncology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Michael S. Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Deepta Bhattacharya
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
141
|
Hernández PP, Mahlakoiv T, Yang I, Schwierzeck V, Nguyen N, Guendel F, Gronke K, Ryffel B, Hoelscher C, Dumoutier L, Renauld JC, Suerbaum S, Staeheli P, Diefenbach A. Interferon-λ and interleukin 22 act synergistically for the induction of interferon-stimulated genes and control of rotavirus infection. Nat Immunol 2015; 16:698-707. [PMID: 26006013 PMCID: PMC4589158 DOI: 10.1038/ni.3180] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/27/2015] [Indexed: 12/13/2022]
Abstract
The epithelium is the main entry point for many viruses, but the processes that protect barrier surfaces against viral infections are incompletely understood. Here we identified interleukin 22 (IL-22) produced by innate lymphoid cell group 3 (ILC3) as an amplifier of signaling via interferon-λ (IFN-λ), a synergism needed to curtail the replication of rotavirus, the leading cause of childhood gastroenteritis. Cooperation between the receptor for IL-22 and the receptor for IFN-λ, both of which were 'preferentially' expressed by intestinal epithelial cells (IECs), was required for optimal activation of the transcription factor STAT1 and expression of interferon-stimulated genes (ISGs). These data suggested that epithelial cells are protected against viral replication by co-option of two evolutionarily related cytokine networks. These data may inform the design of novel immunotherapy for viral infections that are sensitive to interferons.
Collapse
Affiliation(s)
- Pedro P. Hernández
- Research Centre Immunology and Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
- Max-Planck-Institute for Immunobiology and Epigenetics, Stübeweg 51, D-79108 Freiburg, Germany
| | - Tanel Mahlakoiv
- Department of Medical Microbiology and Hygiene, Institute for Virology, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstrasse 19A, D-79104 Freiburg, Germany
| | - Ines Yang
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany and DZIF – German Center for Infection Research, Hannover-Braunschweig Site, D-30625 Hannover, Germany
| | - Vera Schwierzeck
- Research Centre Immunology and Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
| | - Nam Nguyen
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
| | - Fabian Guendel
- Research Centre Immunology and Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
- Research Training Group (GRK1104) of Organogenesis, Hauptstrasse 1, D-79104 Freiburg, Germany
| | - Konrad Gronke
- Research Centre Immunology and Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
- Max-Planck-Institute for Immunobiology and Epigenetics, Stübeweg 51, D-79108 Freiburg, Germany
| | - Bernhard Ryffel
- INEM - UMR7355, Molecular Immunology, University and CNRS, F-45071 Orleans, France and Institute of Infectious Disease, University of Cape Town, RSA
| | - Christoph Hoelscher
- Infection Immunology Research, Research Center Borstel, D-23845 Borstel, Germany
- Cluster of Excellence Inflammation at Interfaces (Borstel-Kiel-Lübeck-Plön)
| | - Laure Dumoutier
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, 74 Avenue Hippocrate, B-1200 Brussels, Belgium
| | - Jean-Christophe Renauld
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, 74 Avenue Hippocrate, B-1200 Brussels, Belgium
| | - Sebastian Suerbaum
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany and DZIF – German Center for Infection Research, Hannover-Braunschweig Site, D-30625 Hannover, Germany
| | - Peter Staeheli
- Department of Medical Microbiology and Hygiene, Institute for Virology, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
| | - Andreas Diefenbach
- Research Centre Immunology and Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, D-55131 Mainz, Germany
- Department of Medical Microbiology and Hygiene, Institute for Medical Microbiology and Hygiene, Freiburg University Medical Centre, Hermann-Herder-Strasse 11, D-79104 Freiburg, Germany
- Research Training Group (GRK1104) of Organogenesis, Hauptstrasse 1, D-79104 Freiburg, Germany
| |
Collapse
|
142
|
López-Rodríguez C, Aramburu J, Berga-Bolaños R. Transcription factors and target genes of pre-TCR signaling. Cell Mol Life Sci 2015; 72:2305-21. [PMID: 25702312 PMCID: PMC11113633 DOI: 10.1007/s00018-015-1864-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/22/2015] [Accepted: 02/16/2015] [Indexed: 11/27/2022]
Abstract
Almost 30 years ago pioneering work by the laboratories of Harald von Boehmer and Susumo Tonegawa provided the first indications that developing thymocytes could assemble a functional TCRβ chain-containing receptor complex, the pre-TCR, before TCRα expression. The discovery and study of the pre-TCR complex revealed paradigms of signaling pathways in control of cell survival and proliferation, and culminated in the recognition of the multifunctional nature of this receptor. As a receptor integrated in a dynamic developmental process, the pre-TCR must be viewed not only in the light of the biological outcomes it promotes, but also in context with those molecular processes that drive its expression in thymocytes. This review article focuses on transcription factors and target genes activated by the pre-TCR to drive its different outcomes.
Collapse
Affiliation(s)
- Cristina López-Rodríguez
- Immunology Unit, Department of Experimental and Health Sciences and Barcelona Biomedical Research Park, Universitat Pompeu Fabra, C/Doctor Aiguader Nº88, 08003, Barcelona, Barcelona, Spain,
| | | | | |
Collapse
|
143
|
Whetstone RD, Gold B. T-cells enhance stem cell mutagenesis in the mouse colon. Mutat Res 2015; 774:1-5. [PMID: 25770826 DOI: 10.1016/j.mrfmmm.2015.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 06/04/2023]
Abstract
A role of inflammation in the etiology of cancer is attributed to the production of reactive oxygen/nitrogen species that can damage DNA. To test this hypothesis, we determined the mutation frequency (MF) in colonic stem cells in C57Bl/6 mice exposed to azoxymethane (AOM), dextran sulfate sodium (DSS) and a combination of AOM and DSS (AOM+DSS). AOM+DSS efficiently and rapidly produces colon tumors in B6 mice. AOM produces promutagenic O(6)-methylguanine lesions in DNA but does not induce colon tumors in C57Bl/6 mice as a single agent. DSS produces inflammation in the colon but does not produce tumors except upon multiple cycles of treatment in some DNA repair deficient mouse models. In addition, using TCRβ null mice we tested whether α/β T cells have any effect on the colonic stem cell MF in mice treated with AOM, DSS and AOM+DSS. The TCRβ(-/-) mice are devoid of the critical receptor required for normal cytolytic and regulatory α/β T-cell functions. The MF in the untreated and DSS treated WT and TCRβ(-/-) mice was the same (<10(-5)) indicating that DSS and subsequent inflammation does not generate stem cell mutations in mice that are WT for DNA repair. AOM yielded mutant crypts in WT and TCRβ(-/-) mice with MF's of ∼4×10(-4) and 2×10(-4), respectively, which represents a statistically significant decrease in the MF in the immune compromised mice. The combined treatment of AOM+DSS afforded fully mutated crypts in both strains with a statistically significant lower MF in the TCRβ(-/-) mice. In addition, the MF in both strains of mice after the combination of AOM+DSS is lower than observed with AOM alone indicating that DSS inflammation destroyed pre-existing AOM mutated crypts. Using the MF in WT mice, the efficiency for the conversion of promutagenic O(6)-methylguanine lesions into a stem cell mutations was calculated to be ∼0.4%.
Collapse
Affiliation(s)
- Ryan D Whetstone
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Barry Gold
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|
144
|
Human congenital T-cell receptor disorders. LYMPHOSIGN JOURNAL-THE JOURNAL OF INHERITED IMMUNE DISORDERS 2015. [DOI: 10.14785/lpsn-2014-0012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Immunodeficiencies of most T-cell receptor (TCR) components (TCRID) have been reported in almost 40 patients worldwide who have also, at times, shown signs of autoimmunity. We updated their clinical, immunological, and molecular features with an emphasis on practical diagnosis, as the range of the disorder grows in complexity with new partial defects. Cellular and animal models are also reviewed and in some cases reveal their limitations for predicting TCRID immunopathology.
Collapse
|
145
|
Yilmaz B, Portugal S, Tran TM, Gozzelino R, Ramos S, Gomes J, Regalado A, Cowan PJ, d'Apice AJF, Chong AS, Doumbo OK, Traore B, Crompton PD, Silveira H, Soares MP. Gut microbiota elicits a protective immune response against malaria transmission. Cell 2015; 159:1277-89. [PMID: 25480293 PMCID: PMC4261137 DOI: 10.1016/j.cell.2014.10.053] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/26/2014] [Accepted: 09/30/2014] [Indexed: 11/29/2022]
Abstract
Glycosylation processes are under high natural selection pressure, presumably because these can modulate resistance to infection. Here, we asked whether inactivation of the UDP-galactose:β-galactoside-α1-3-galactosyltransferase (α1,3GT) gene, which ablated the expression of the Galα1-3Galβ1-4GlcNAc-R (α-gal) glycan and allowed for the production of anti-α-gal antibodies (Abs) in humans, confers protection against Plasmodium spp. infection, the causative agent of malaria and a major driving force in human evolution. We demonstrate that both Plasmodium spp. and the human gut pathobiont E. coli O86:B7 express α-gal and that anti-α-gal Abs are associated with protection against malaria transmission in humans as well as in α1,3GT-deficient mice, which produce protective anti-α-gal Abs when colonized by E. coli O86:B7. Anti-α-gal Abs target Plasmodium sporozoites for complement-mediated cytotoxicity in the skin, immediately after inoculation by Anopheles mosquitoes. Vaccination against α-gal confers sterile protection against malaria in mice, suggesting that a similar approach may reduce malaria transmission in humans.
Collapse
Affiliation(s)
- Bahtiyar Yilmaz
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal
| | - Silvia Portugal
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Twinbrook II, Room 125, 12441 Parklawn Drive, Rockville, MD 20852-8180, USA
| | - Tuan M Tran
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Twinbrook II, Room 125, 12441 Parklawn Drive, Rockville, MD 20852-8180, USA
| | - Raffaella Gozzelino
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal
| | - Susana Ramos
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal
| | - Joana Gomes
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal; Centro de Malaria e Outras Doenças Tropicais, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
| | - Ana Regalado
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal
| | - Peter J Cowan
- Immunology Research Centre, St. Vincent's Hospital, Fitzroy, Melbourne, VIC 3065, Australia; Department of Medicine, University of Melbourne, Parkville, VIC 2900, Australia
| | - Anthony J F d'Apice
- Immunology Research Centre, St. Vincent's Hospital, Fitzroy, Melbourne, VIC 3065, Australia; Department of Medicine, University of Melbourne, Parkville, VIC 2900, Australia
| | - Anita S Chong
- Section of Transplantation, Department of Surgery, University of Chicago, 5841 South Maryland Avenue, Chicago, IL 60637, USA
| | - Ogobara K Doumbo
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, 1805 Bamako, Mali
| | - Boubacar Traore
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, 1805 Bamako, Mali
| | - Peter D Crompton
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Twinbrook II, Room 125, 12441 Parklawn Drive, Rockville, MD 20852-8180, USA
| | - Henrique Silveira
- Centro de Malaria e Outras Doenças Tropicais, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira, 100, 1349-008 Lisboa, Portugal
| | - Miguel P Soares
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, 2780-156 Oeiras, Portugal.
| |
Collapse
|
146
|
Buckley MW, Trampont PC, Arandjelovic S, Fond AM, Juncadella IJ, Ravichandran KS. ShcA regulates late stages of T cell development and peripheral CD4+ T cell numbers. THE JOURNAL OF IMMUNOLOGY 2015; 194:1665-76. [PMID: 25595778 DOI: 10.4049/jimmunol.1401728] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cell development in the thymus is a highly regulated process that critically depends upon productive signaling via the preTCR at the β-selection stage, as well as via the TCR for selection from the CD4(+)CD8(+) double-positive stage to the CD4 or CD8 single-positive stage. ShcA is an adapter protein expressed in thymocytes, and it is required for productive signaling through the preTCR, with impaired signaling via ShcA leading to a developmental block at the β-selection checkpoint. However, the role of ShcA in subsequent stages of T cell development has not been addressed. In this study, we generated transgenic mice (CD4-Cre/ShcFFF mice) that specifically express a phosphorylation-defective dominant-negative ShcA mutant (ShcFFF) in late T cell development. Thymocytes in CD4-Cre/ShcFFF mice progressed normally through the β-selection checkpoint, but displayed a significant reduction in the numbers of single-positive CD4(+) and CD8(+) thymocytes. Furthermore, CD4-Cre/ShcFFF mice, when bred with transgenic TCR mouse strains, had impaired signaling through the transgenic TCRs. Consistent with defective progression to the single-positive stage, CD4-Cre/ShcFFF mice also had significant peripheral lymphopenia. Moreover, these CD4-Cre/ShcFFF mice develop attenuated disease in CD4(+) T cell-dependent experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. Collectively, these data identify an important role for the adapter protein ShcA in later stages of thymic T cell development and in peripheral T cell-dependent events.
Collapse
Affiliation(s)
- Monica W Buckley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908; and Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908
| | - Paul C Trampont
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908; and Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908
| | - Sanja Arandjelovic
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908; and Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908
| | - Aaron M Fond
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908; and Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908
| | - Ignacio J Juncadella
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908; and Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908; Carter Immunology Center, University of Virginia, Charlottesville, VA 22908; and Center for Cell Clearance, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
147
|
Lucca LE, Desbois S, Ramadan A, Ben-Nun A, Eisenstein M, Carrié N, Guéry JC, Sette A, Nguyen P, Geiger TL, Mars LT, Liblau RS. Bispecificity for myelin and neuronal self-antigens is a common feature of CD4 T cells in C57BL/6 mice. THE JOURNAL OF IMMUNOLOGY 2014; 193:3267-77. [PMID: 25135834 DOI: 10.4049/jimmunol.1400523] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The recognition of multiple ligands by a single TCR is an intrinsic feature of T cell biology, with important consequences for physiological and pathological processes. Polyspecific T cells targeting distinct self-antigens have been identified in healthy individuals as well as in the context of autoimmunity. We have previously shown that the 2D2 TCR recognizes the myelin oligodendrocyte glycoprotein epitope (MOG)35-55 as well as an epitope within the axonal protein neurofilament medium (NF-M15-35) in H-2(b) mice. In this study, we assess whether this cross-reactivity is a common feature of the MOG35-55-specific T cell response. To this end, we analyzed the CD4 T cell response of MOG35-55-immunized C57BL/6 mice for cross-reactivity with NF-M15-35. Using Ag recall responses, we established that an important proportion of MOG35-55-specific CD4 T cells also responded to NF-M15-35 in all mice tested. To study the clonality of this response, we analyzed 22 MOG35-55-specific T cell hybridomas expressing distinct TCR. Seven hybridomas were found to cross-react with NF-M15-35. Using an alanine scan of NF-M18-30 and an in silico predictive model, we dissected the molecular basis of cross-reactivity between MOG35-55 and NF-M15-35. We established that NF-M F24, R26, and V27 proved important TCR contacts. Strikingly, the identified TCR contacts are conserved within MOG38-50. Our data indicate that due to linear sequence homology, part of the MOG35-55-specific T cell repertoire of all C57BL/6 mice also recognizes NF-M15-35, with potential implications for CNS autoimmunity.
Collapse
Affiliation(s)
- Liliana E Lucca
- INSERM, U1043, Toulouse F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France; Centre de Physiopathologie Toulouse-Purpan, Université Toulouse 3, Toulouse F-31300, France
| | - Sabine Desbois
- INSERM, U1043, Toulouse F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France; Centre de Physiopathologie Toulouse-Purpan, Université Toulouse 3, Toulouse F-31300, France
| | - Abdulraouf Ramadan
- INSERM, U1043, Toulouse F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France; Centre de Physiopathologie Toulouse-Purpan, Université Toulouse 3, Toulouse F-31300, France
| | - Avraham Ben-Nun
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel; Department of Chemical Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Miriam Eisenstein
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel; Department of Chemical Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nadège Carrié
- INSERM, U1043, Toulouse F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France; Centre de Physiopathologie Toulouse-Purpan, Université Toulouse 3, Toulouse F-31300, France
| | - Jean-Charles Guéry
- INSERM, U1043, Toulouse F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France; Centre de Physiopathologie Toulouse-Purpan, Université Toulouse 3, Toulouse F-31300, France
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, San Diego, CA 92109
| | - Phuong Nguyen
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105; and
| | - Terrence L Geiger
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105; and
| | - Lennart T Mars
- INSERM, U1043, Toulouse F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France; Centre de Physiopathologie Toulouse-Purpan, Université Toulouse 3, Toulouse F-31300, France
| | - Roland S Liblau
- INSERM, U1043, Toulouse F-31300, France; Centre National de la Recherche Scientifique, U5282, Toulouse F-31300, France; Centre de Physiopathologie Toulouse-Purpan, Université Toulouse 3, Toulouse F-31300, France; Département d'Immunologie, Centre Hospitalier Universitaire Toulouse, Hôpital Purpan, Toulouse F-31300, France
| |
Collapse
|
148
|
Niebling J, E Rünker A, Schallenberg S, Kretschmer K, Kempermann G. Myelin-specific T helper 17 cells promote adult hippocampal neurogenesis through indirect mechanisms. F1000Res 2014; 3:169. [PMID: 25383186 DOI: 10.12688/f1000research.4439.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/08/2014] [Indexed: 01/05/2023] Open
Abstract
CD4 + T cells provide a neuro-immunological link in the regulation of adult hippocampal neurogenesis, but the exact mechanisms underlying enhanced neural precursor cell proliferation and the relative contribution of different T helper (Th) cell subsets have remained unclear. Here, we explored the pro-proliferative potential of interleukin 17-producing T helper (Th17) cells, a developmentally and functionally distinct Th cell subset that is a key mediator of autoimmune neurodegeneration. We found that base-line proliferation of hippocampal precursor cells in a T cell-deficient mouse model of impaired hippocampal neurogenesis can be restored upon adoptive transfer with homogeneous Th17 populations enriched for myelin-reactive T cell receptors (TCR). In these experiments, enhanced proliferation was independent of direct interactions of infiltrating Th17 cells with precursor cells or neighboring cells in the hippocampal neurogenic niche. Complementary studies in immunocompetent mice identified several receptors for Th17 cell-derived cytokines with mRNA expression in hippocampal precursor cells and dentate gyrus tissue, suggesting that Th17 cell activity in peripheral lymphoid tissues might promote hippocampal neurogenesis through secreted cytokines.
Collapse
Affiliation(s)
- Johannes Niebling
- Molecular and Cellular Immunology/Immune Regulation, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany.,Genomics of Regeneration, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Annette E Rünker
- Genomics of Regeneration, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany.,German Center for Neurodegenerative Diseases (DZNE) Dresden, Arnoldstraße 18b, 01307 Dresden, Germany
| | - Sonja Schallenberg
- Molecular and Cellular Immunology/Immune Regulation, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Karsten Kretschmer
- Molecular and Cellular Immunology/Immune Regulation, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Gerd Kempermann
- Genomics of Regeneration, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany.,German Center for Neurodegenerative Diseases (DZNE) Dresden, Arnoldstraße 18b, 01307 Dresden, Germany
| |
Collapse
|
149
|
Niebling J, E Rünker A, Schallenberg S, Kretschmer K, Kempermann G. Myelin-specific T helper 17 cells promote adult hippocampal neurogenesis through indirect mechanisms. F1000Res 2014; 3:169. [PMID: 25383186 PMCID: PMC4215755 DOI: 10.12688/f1000research.4439.2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2017] [Indexed: 12/31/2022] Open
Abstract
CD4
+ T cells provide a neuro-immunological link in the regulation of adult hippocampal neurogenesis, but the exact mechanisms underlying enhanced neural precursor cell proliferation and the relative contribution of different T helper (Th) cell subsets have remained unclear. Here, we explored the pro-proliferative potential of interleukin 17-producing T helper (Th17) cells, a developmentally and functionally distinct Th cell subset that is a key mediator of autoimmune neurodegeneration. We found that base-line proliferation of hippocampal precursor cells in a T cell-deficient mouse model of impaired hippocampal neurogenesis can be restored upon adoptive transfer with homogeneous Th17 populations enriched for myelin-reactive T cell receptors (TCR). In these experiments, enhanced proliferation was independent of direct interactions of infiltrating Th17 cells with precursor cells or neighboring cells in the hippocampal neurogenic niche. Complementary studies in immunocompetent mice identified several receptors for Th17 cell-derived cytokines with mRNA expression in hippocampal precursor cells and dentate gyrus tissue, suggesting that Th17 cell activity in peripheral lymphoid tissues might promote hippocampal neurogenesis through secreted cytokines.
Collapse
Affiliation(s)
- Johannes Niebling
- Molecular and Cellular Immunology/Immune Regulation, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany.,Genomics of Regeneration, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Annette E Rünker
- Genomics of Regeneration, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany.,German Center for Neurodegenerative Diseases (DZNE) Dresden, Arnoldstraße 18b, 01307 Dresden, Germany
| | - Sonja Schallenberg
- Molecular and Cellular Immunology/Immune Regulation, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Karsten Kretschmer
- Molecular and Cellular Immunology/Immune Regulation, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Gerd Kempermann
- Genomics of Regeneration, CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany.,German Center for Neurodegenerative Diseases (DZNE) Dresden, Arnoldstraße 18b, 01307 Dresden, Germany
| |
Collapse
|
150
|
c-Myc-induced transcription factor AP4 is required for host protection mediated by CD8+ T cells. Nat Immunol 2014; 15:884-93. [PMID: 25029552 PMCID: PMC4139462 DOI: 10.1038/ni.2943] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 06/17/2014] [Indexed: 12/12/2022]
Abstract
Although c-Myc is essential to establish a metabolically active and proliferative state in T cells after priming, its expression is transient. It remains unknown how T cell activation is maintained after c-Myc down-regulation. Here, we identify AP4 as the transcription factor that is induced by c-Myc and sustains activation of antigen-specific CD8+ T cells. Despite normal priming, AP4-deficient CD8+ T cells fail to continue transcription of a broad range of c-Myc-dependent targets. Mice lacking AP4 specifically in CD8+ T cells showed enhanced susceptibility to West Nile virus infection. Genome-wide analysis suggests that many activation-induced metabolic genes are shared targets of c-Myc and AP4. Thus, AP4 maintains c-Myc-initiated cellular activation programs in CD8+ T cells to control microbial infections.
Collapse
|