51
|
Altmann C, Hardt S, Fischer C, Heidler J, Lim HY, Häussler A, Albuquerque B, Zimmer B, Möser C, Behrends C, Koentgen F, Wittig I, Schmidt MH, Clement AM, Deller T, Tegeder I. Progranulin overexpression in sensory neurons attenuates neuropathic pain in mice: Role of autophagy. Neurobiol Dis 2016; 96:294-311. [DOI: 10.1016/j.nbd.2016.09.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 08/26/2016] [Accepted: 09/06/2016] [Indexed: 12/14/2022] Open
|
52
|
Altmann C, Vasic V, Hardt S, Heidler J, Häussler A, Wittig I, Schmidt MHH, Tegeder I. Progranulin promotes peripheral nerve regeneration and reinnervation: role of notch signaling. Mol Neurodegener 2016; 11:69. [PMID: 27770818 PMCID: PMC5075406 DOI: 10.1186/s13024-016-0132-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 09/28/2016] [Indexed: 01/11/2023] Open
Abstract
Background Peripheral nerve injury is a frequent cause of lasting motor deficits and chronic pain. Although peripheral nerves are capable of regrowth they often fail to re-innervate target tissues. Results Using newly generated transgenic mice with inducible neuronal progranulin overexpression we show that progranulin accelerates axonal regrowth, restoration of neuromuscular synapses and recovery of sensory and motor functions after injury of the sciatic nerve. Oppositely, progranulin deficient mice have long-lasting deficits in motor function tests after nerve injury due to enhanced losses of motor neurons and stronger microglia activation in the ventral horn of the spinal cord. Deep proteome and gene ontology (GO) enrichment analysis revealed that the proteins upregulated in progranulin overexpressing mice were involved in ‘regulation of transcription’ and ‘response to insulin’ (GO terms). Transcription factor prediction pointed to activation of Notch signaling and indeed, co-immunoprecipitation studies revealed that progranulin bound to the extracellular domain of Notch receptors, and this was functionally associated with higher expression of Notch target genes in the dorsal root ganglia of transgenic mice with neuronal progranulin overexpression. Functionally, these transgenic mice recovered normal gait and running, which was not achieved by controls and was stronger impaired in progranulin deficient mice. Conclusion We infer that progranulin activates Notch signaling pathways, enhancing thereby the regenerative capacity of partially injured neurons, which leads to improved motor function recovery. Graphical abstract ![]()
Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0132-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christine Altmann
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Verica Vasic
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stefanie Hardt
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Juliana Heidler
- Functional Proteomics, SFB815 Core Unit, Goethe-University, Frankfurt, Germany
| | - Annett Häussler
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Ilka Wittig
- Functional Proteomics, SFB815 Core Unit, Goethe-University, Frankfurt, Germany
| | - Mirko H H Schmidt
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany.
| |
Collapse
|
53
|
Essential protective role of tumor necrosis factor receptor 2 in neurodegeneration. Proc Natl Acad Sci U S A 2016; 113:12304-12309. [PMID: 27791020 DOI: 10.1073/pnas.1605195113] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite the recognized role of tumor necrosis factor (TNF) in inflammation and neuronal degeneration, anti-TNF therapeutics failed to treat neurodegenerative diseases. Animal disease models had revealed the antithetic effects of the two TNF receptors (TNFR) in the central nervous system, whereby TNFR1 has been associated with inflammatory degeneration and TNFR2 with neuroprotection. We here show the therapeutic potential of selective inhibition of TNFR1 and activation of TNFR2 by ATROSAB, a TNFR1-selective antagonistic antibody, and EHD2-scTNFR2, an agonistic TNFR2-selective TNF, respectively, in a mouse model of NMDA-induced acute neurodegeneration. Coadministration of either ATROSAB or EHD2-scTNFR2 into the magnocellular nucleus basalis significantly protected cholinergic neurons and their cortical projections against cell death, and reverted the neurodegeneration-associated memory impairment in a passive avoidance paradigm. Simultaneous blocking of TNFR1 and TNFR2 signaling, however, abrogated the therapeutic effect. Our results uncover an essential role of TNFR2 in neuroprotection. Accordingly, the therapeutic activity of ATROSAB is mediated by shifting the balance of the antithetic activity of endogenous TNF toward TNFR2, which appears essential for neuroprotection. Our data also explain earlier results showing that complete blocking of TNF activity by anti-TNF drugs was detrimental rather than protective and argue for the use of next-generation TNFR-selective TNF therapeutics as an effective approach in treating neurodegenerative diseases.
Collapse
|
54
|
Voisinne G, García-Blesa A, Chaoui K, Fiore F, Bergot E, Girard L, Malissen M, Burlet-Schiltz O, Gonzalez de Peredo A, Malissen B, Roncagalli R. Co-recruitment analysis of the CBL and CBLB signalosomes in primary T cells identifies CD5 as a key regulator of TCR-induced ubiquitylation. Mol Syst Biol 2016; 12:876. [PMID: 27474268 PMCID: PMC4965873 DOI: 10.15252/msb.20166837] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
T-cell receptor (TCR) signaling is essential for the function of T cells and negatively regulated by the E3 ubiquitin-protein ligases CBL and CBLB Here, we combined mouse genetics and affinity purification coupled to quantitative mass spectrometry to monitor the dynamics of the CBL and CBLB signaling complexes that assemble in normal T cells over 600 seconds of TCR stimulation. We identify most previously known CBL and CBLB interacting partners, as well as a majority of proteins that have not yet been implicated in those signaling complexes. We exploit correlations in protein association with CBL and CBLB as a function of time of TCR stimulation for predicting the occurrence of direct physical association between them. By combining co-recruitment analysis with biochemical analysis, we demonstrated that the CD5 transmembrane receptor constitutes a key scaffold for CBL- and CBLB-mediated ubiquitylation following TCR engagement. Our results offer an integrated view of the CBL and CBLB signaling complexes induced by TCR stimulation and provide a molecular basis for their negative regulatory function in normal T cells.
Collapse
Affiliation(s)
- Guillaume Voisinne
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - Antonio García-Blesa
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - Karima Chaoui
- Institut de Pharmacologie et de Biologie Structurale, Département Biologie Structural Biophysique, Protéomique Génopole Toulouse Midi Pyrénées, CNRS UMR 5089, Toulouse Cedex, France
| | - Frédéric Fiore
- Centre d'Immunophénomique, Aix Marseille Université UM2, Inserm US012, CNRS UMS3367, Marseille, France
| | - Elise Bergot
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| | - Laura Girard
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France Centre d'Immunophénomique, Aix Marseille Université UM2, Inserm US012, CNRS UMS3367, Marseille, France
| | - Marie Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France Centre d'Immunophénomique, Aix Marseille Université UM2, Inserm US012, CNRS UMS3367, Marseille, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale, Département Biologie Structural Biophysique, Protéomique Génopole Toulouse Midi Pyrénées, CNRS UMR 5089, Toulouse Cedex, France
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale, Département Biologie Structural Biophysique, Protéomique Génopole Toulouse Midi Pyrénées, CNRS UMR 5089, Toulouse Cedex, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France Centre d'Immunophénomique, Aix Marseille Université UM2, Inserm US012, CNRS UMS3367, Marseille, France
| | - Romain Roncagalli
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Inserm, CNRS, Marseille, France
| |
Collapse
|
55
|
Abstract
The mouse has joined the fruit fly, bread mold, and cyanobacteria as a tractable genetic system for studying mechanisms of circadian rhythms. The circadian rhythms of “knock-out” mice for specific clock genes, however, have demonstrated significant variability between laboratories. In this brief review, the authors discuss possible sources of this variability, focusing particularly on questions of modifier loci of circadian rhythms that vary between inbred mouse strains. They conclude with a short list of recommendations for researchers working on circadian rhythms in mixed-strain mice.
Collapse
Affiliation(s)
- Russell N Van Gelder
- Department of Ophthalmology and Visual Sciences, Washington University Medical School, St. Louis, MO 63110, USA.
| | | |
Collapse
|
56
|
Vlantis K, Polykratis A, Welz PS, van Loo G, Pasparakis M, Wullaert A. TLR-independent anti-inflammatory function of intestinal epithelial TRAF6 signalling prevents DSS-induced colitis in mice. Gut 2016; 65:935-43. [PMID: 25761602 PMCID: PMC4893119 DOI: 10.1136/gutjnl-2014-308323] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 02/17/2015] [Indexed: 01/31/2023]
Abstract
OBJECTIVE The gut microbiota modulates host susceptibility to intestinal inflammation, but the cell types and the signalling pathways orchestrating this bacterial regulation of intestinal homeostasis remain poorly understood. Here, we investigated the function of intestinal epithelial toll-like receptor (TLR) responses in the dextran sodium sulfate (DSS)-induced mouse model of colitis. DESIGN We applied an in vivo genetic approach allowing intestinal epithelial cell (IEC)-specific deletion of the critical TLR signalling adaptors, MyD88 and/or TIR-domain-containing adapter-inducing interferon-β (TRIF), as well as the downstream ubiquitin ligase TRAF6 in order to reveal the IEC-intrinsic function of these TLR signalling molecules during DSS colitis. RESULTS Mice lacking TRAF6 in IECs showed exacerbated DSS-induced inflammatory responses that ensued in the development of chronic colon inflammation. Antibiotic pretreatment abolished the increased DSS susceptibility of these mice, showing that epithelial TRAF6 signalling pathways prevent the gut microbiota from driving excessive colitis. However, in contrast to epithelial TRAF6 deletion, blocking epithelial TLR signalling by simultaneous deletion of MyD88 and TRIF specifically in IECs did not affect DSS-induced colitis severity. This in vivo functional comparison between TRAF6 and MyD88/TRIF deletion in IECs shows that the colitis-protecting effects of epithelial TRAF6 signalling are not triggered by TLRs. CONCLUSIONS Intestinal epithelial TRAF6-dependent but MyD88/TRIF-independent and, thus, TLR-independent signalling pathways are critical for preventing propagation of DSS-induced colon inflammation by the gut microbiota. Moreover, our experiments using mice with dual MyD88/TRIF deletion in IECs unequivocally show that the gut microbiota trigger non-epithelial TLRs rather than epithelial TLRs to restrict DSS colitis severity.
Collapse
Affiliation(s)
- Katerina Vlantis
- Institute for Genetics, University of Cologne, Cologne, Germany,Centre for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Apostolos Polykratis
- Institute for Genetics, University of Cologne, Cologne, Germany,Centre for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Patrick-Simon Welz
- Institute for Genetics, University of Cologne, Cologne, Germany,Centre for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany,Institute for Research in Biomedicine (IRB), Barcelona, Spain
| | - Geert van Loo
- Inflammation Research Center, VIB, Ghent, Belgium,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, Cologne, Germany,Centre for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Andy Wullaert
- Institute for Genetics, University of Cologne, Cologne, Germany,Centre for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany,Department of Medical Protein Research, VIB, Ghent, Belgium,Department of Biochemistry, Ghent University, Ghent, Belgium
| |
Collapse
|
57
|
Koentgen F, Lin J, Katidou M, Chang I, Khan M, Watts J, Mombaerts P. Exclusive transmission of the embryonic stem cell-derived genome through the mouse germline. Genesis 2016; 54:326-33. [PMID: 27012318 PMCID: PMC5084746 DOI: 10.1002/dvg.22938] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 03/15/2016] [Accepted: 03/22/2016] [Indexed: 12/27/2022]
Abstract
Gene targeting in embryonic stem (ES) cells remains best practice for introducing complex mutations into the mouse germline. One aspect in this multistep process that has not been streamlined with regard to the logistics and ethics of mouse breeding is the efficiency of germline transmission: the transmission of the ES cell‐derived genome through the germline of chimeras to their offspring. A method whereby male chimeras transmit exclusively the genome of the injected ES cells to their offspring has been developed. The new technology, referred to as goGermline, entails injecting ES cells into blastocysts produced by superovulated homozygous Tsc22d3 floxed females mated with homozygous ROSA26‐Cre males. This cross produces males that are sterile due to a complete cell‐autonomous defect in spermatogenesis. The resulting male chimeras can be sterile but when fertile, they transmit the ES cell‐derived genome to 100% of their offspring. The method was validated extensively and in two laboratories for gene‐targeted ES clones that were derived from the commonly used parental ES cell lines Bruce4, E14, and JM8A3. The complete elimination of the collateral birth of undesired, non‐ES cell‐derived offspring in goGermline technology fulfills the reduction imperative of the 3R principle of humane experimental technique with animals. genesis 54:326–333, 2016. © 2016 The Authors. Genesis Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Frank Koentgen
- Ozgene Pty Ltd, Bentley, Western Australia, 6983, Australia
| | - Jiangwei Lin
- Max Planck Research Unit for Neurogenetics, Max-von-Laue-Strasse 4, Frankfurt, 60438, Germany
| | - Markella Katidou
- Max Planck Research Unit for Neurogenetics, Max-von-Laue-Strasse 4, Frankfurt, 60438, Germany
| | - Isabelle Chang
- Max Planck Research Unit for Neurogenetics, Max-von-Laue-Strasse 4, Frankfurt, 60438, Germany
| | - Mona Khan
- Max Planck Research Unit for Neurogenetics, Max-von-Laue-Strasse 4, Frankfurt, 60438, Germany
| | - Jacqui Watts
- Ozgene Pty Ltd, Bentley, Western Australia, 6983, Australia
| | - Peter Mombaerts
- Max Planck Research Unit for Neurogenetics, Max-von-Laue-Strasse 4, Frankfurt, 60438, Germany
| |
Collapse
|
58
|
Chao BN, Baldwin WH, Healey JF, Parker ET, Shafer-Weaver K, Cox C, Jiang P, Kanellopoulou C, Lollar P, Meeks SL, Lenardo MJ. Characterization of a genetically engineered mouse model of hemophilia A with complete deletion of the F8 gene. J Thromb Haemost 2016; 14:346-55. [PMID: 26588198 PMCID: PMC4755856 DOI: 10.1111/jth.13202] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Indexed: 12/27/2022]
Abstract
UNLABELLED ESSENTIALS: Anti-factor VIII (FVIII) inhibitory antibody formation is a severe complication in hemophilia A therapy. We genetically engineered and characterized a mouse model with complete deletion of the F8 coding region. F8(TKO) mice exhibit severe hemophilia, express no detectable F8 mRNA, and produce FVIII inhibitors. The defined background and lack of FVIII in F8(TKO) mice will aid in studying FVIII inhibitor formation. BACKGROUND The most important complication in hemophilia A treatment is the development of inhibitory anti-Factor VIII (FVIII) antibodies in patients after FVIII therapy. Patients with severe hemophilia who express no endogenous FVIII (i.e. cross-reacting material, CRM) have the greatest incidence of inhibitor formation. However, current mouse models of severe hemophilia A produce low levels of truncated FVIII. The lack of a corresponding mouse model hampers the study of inhibitor formation in the complete absence of FVIII protein. OBJECTIVES We aimed to generate and characterize a novel mouse model of severe hemophilia A (designated the F8(TKO) strain) lacking the complete coding sequence of F8 and any FVIII CRM. METHODS Mice were created on a C57BL/6 background using Cre-Lox recombination and characterized using in vivo bleeding assays, measurement of FVIII activity by coagulation and chromogenic assays, and anti-FVIII antibody production using ELISA. RESULTS All F8 exonic coding regions were deleted from the genome and no F8 mRNA was detected in F8(TKO) mice. The bleeding phenotype of F8(TKO) mice was comparable to E16 mice by measurements of factor activity and tail snip assay. Similar levels of anti-FVIII antibody titers after recombinant FVIII injections were observed between F8(TKO) and E16 mice. CONCLUSIONS We describe a new C57BL/6 mouse model for severe hemophilia A patients lacking CRM. These mice can be directly bred to the many C57BL/6 strains of genetically engineered mice, which is valuable for studying the impact of a wide variety of genes on FVIII inhibitor formation on a defined genetic background.
Collapse
Affiliation(s)
- Brittany N. Chao
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD
| | - Wallace H. Baldwin
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta; the Department of Pediatrics, Emory University, Atlanta, GA
| | - John F. Healey
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta; the Department of Pediatrics, Emory University, Atlanta, GA
| | - Ernest T. Parker
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta; the Department of Pediatrics, Emory University, Atlanta, GA
| | - Kimberly Shafer-Weaver
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD
| | - Courtney Cox
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta; the Department of Pediatrics, Emory University, Atlanta, GA
| | - Ping Jiang
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD
| | - Chrysi Kanellopoulou
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD
| | - Pete Lollar
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta; the Department of Pediatrics, Emory University, Atlanta, GA
| | - Shannon L. Meeks
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta; the Department of Pediatrics, Emory University, Atlanta, GA
| | - Michael J. Lenardo
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD
| |
Collapse
|
59
|
Moulin M, Voss AK, Thomas T, Wong WWL, Cook WD, Koentgen F, Vince J, Silke J, Vaux DL. Response to Heard et al. EMBO J 2015; 34:2396-7. [PMID: 26427759 DOI: 10.15252/embj.201592761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
| | - Anne K Voss
- The Walter and Eliza Hall Institute, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Tim Thomas
- The Walter and Eliza Hall Institute, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Wendy Wei-Lynn Wong
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Wendy D Cook
- La Trobe Institute for Molecular Sciences, La Trobe University, Bundoora, Vic., Australia
| | | | - James Vince
- The Walter and Eliza Hall Institute, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - John Silke
- The Walter and Eliza Hall Institute, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - David L Vaux
- The Walter and Eliza Hall Institute, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
60
|
Duraes FV, Lippens C, Steinbach K, Dubrot J, Brighouse D, Bendriss-Vermare N, Issazadeh-Navikas S, Merkler D, Hugues S. pDC therapy induces recovery from EAE by recruiting endogenous pDC to sites of CNS inflammation. J Autoimmun 2015; 67:8-18. [PMID: 26341385 PMCID: PMC4758828 DOI: 10.1016/j.jaut.2015.08.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 11/25/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) exhibit both innate and adaptive functions. In particular they are the main source of type I IFNs and directly impact T cell responses through antigen presentation. We have previously demonstrated that during experimental autoimmune encephalomyelitis (EAE) initiation, myelin-antigen presentation by pDCs is associated with suppressive Treg development and results in attenuated EAE. Here, we show that pDCs transferred during acute disease phase confer recovery from EAE. Clinical improvement is associated with migration of injected pDCs into inflamed CNS and is dependent on the subsequent and selective chemerin-mediated recruitment of endogenous pDCs to the CNS. The protective effect requires pDC pre-loading with myelin antigen, and is associated with the modulation of CNS-infiltrating pDC phenotype and inhibition of CNS encephalitogenic T cells. This study may pave the way for novel pDC-based cell therapies in autoimmune diseases, aiming at specifically modulating pathogenic cells that induce and sustain autoimmune inflammation. pDC therapy ameliorates established EAE. CNS inflammation is locally modulated after pDC transfer. Upon pDC transfer, resting endogenous pDCs are selectively recruited to the CNS via chemerin/CMKLR1 axis. Therapeutic pDC injection promotes a tolerogenic environment and inhibits encephalitogenic T cells in the CNS.
Collapse
Affiliation(s)
- Fernanda V Duraes
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Carla Lippens
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Karin Steinbach
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Juan Dubrot
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Dale Brighouse
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Nathalie Bendriss-Vermare
- Université Lyon 1, INSERM U1052, CNRS, UMR5286, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, LabEx DEVweCAN, Lyon, France
| | | | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; Department of Pathology and Immunology, Division of Clinical Pathology, University & University Hospital of Geneva, Switzerland
| | - Stephanie Hugues
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland.
| |
Collapse
|
61
|
Vulin A, Wein N, Simmons TR, Rutherford AM, Findlay AR, Yurkoski JA, Kaminoh Y, Flanigan KM. The first exon duplication mouse model of Duchenne muscular dystrophy: A tool for therapeutic development. Neuromuscul Disord 2015; 25:827-34. [PMID: 26365037 DOI: 10.1016/j.nmd.2015.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 12/27/2022]
Abstract
Exon duplication mutations account for up to 11% of all cases of Duchenne muscular dystrophy (DMD), and a duplication of exon 2 is the most common duplication in patients. For use as a platform for testing of duplication-specific therapies, we developed a mouse model that carries a Dmd exon 2 duplication. By using homologous recombination we duplicated exon 2 within intron 2 at a location consistent with a human duplication hotspot. mRNA analysis confirms the inclusion of a duplicated exon 2 in mouse muscle. Dystrophin expression is essentially absent by immunofluorescent and immunoblot analysis, although some muscle specimens show very low-level trace dystrophin expression. Phenotypically, the mouse shows similarities to mdx, the standard laboratory model of DMD. In skeletal muscle, areas of necrosis and phagocytosis are seen at 3 weeks, with central nucleation prominent by four weeks, recapitulating the "crisis" period in mdx. Marked diaphragm fibrosis is noted by 6 months, and remains unchanged at 12 months. Our results show that the Dup2 mouse is both pathologically (in degree and distribution) and physiologically similar to mdx. As it recapitulates the most common single exon duplication found in DMD patients, this new model will be a useful tool to assess the potential of duplicated exon skipping.
Collapse
Affiliation(s)
- Adeline Vulin
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Nicolas Wein
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Tabatha R Simmons
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Andrea M Rutherford
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Andrew R Findlay
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Jacqueline A Yurkoski
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Yuuki Kaminoh
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Kevin M Flanigan
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA; Department of Neurology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
62
|
Mosaic Deficiency in Mitochondrial Oxidative Metabolism Promotes Cardiac Arrhythmia during Aging. Cell Metab 2015; 21:667-77. [PMID: 25955204 DOI: 10.1016/j.cmet.2015.04.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 02/04/2015] [Accepted: 03/31/2015] [Indexed: 11/21/2022]
Abstract
Aging is a progressive decline of body function, during which many tissues accumulate few cells with high levels of deleted mitochondrial DNA (mtDNA), leading to a defect of mitochondrial functions. Whether this mosaic mitochondrial deficiency contributes to organ dysfunction is unknown. To investigate this, we generated mice with an accelerated accumulation of mtDNA deletions in the myocardium, by expressing a dominant-negative mutant mitochondrial helicase. These animals accumulated few randomly distributed cardiomyocytes with compromised mitochondrial function, which led to spontaneous ventricular premature contractions and AV blocks at 18 months. These symptoms were not caused by a general mitochondrial dysfunction in the entire myocardium, and were not observed in mice at 12 months with significantly lower numbers of dysfunctional cells. Therefore, our results suggest that the disposition to arrhythmia typically found in the aged human heart might be due to the random accumulation of mtDNA deletions and the subsequent mosaic respiratory chain deficiency.
Collapse
|
63
|
Lu S, Seravalli J, Harrison-Findik D. Inductively coupled mass spectrometry analysis of biometals in conditional Hamp1 and Hamp1 and Hamp2 transgenic mouse models. Transgenic Res 2015; 24:765-73. [PMID: 25904410 DOI: 10.1007/s11248-015-9879-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 04/14/2015] [Indexed: 12/12/2022]
Abstract
Hepcidin, a circulatory antimicrobial peptide, is involved in iron homeostasis, inflammation, infection and metabolic signals. Humans express one hepcidin gene, HAMP but mice express two hepcidin genes, Hamp1 and Hamp2. Consecutive gene targeting events were performed to produce transgenic mice expressing conditional alleles of either Hamp1 or both Hamp1 and Hamp2 (Hamp1/2). The deletion of Hamp1 alleles elevated Hamp2 expression, particularly in males, which was reduced by endotoxin treatment. The tissue levels of iron and other biometals were quantified by inductively coupled mass spectrometry. The ubiquitous or liver-specific deletion of Hamp1 alleles yielded similar quantitative changes in iron levels in the liver, duodenum, spleen, kidney, heart and brain. The introduction of Hamp2 null allele did not exacerbate the iron-related phenotype of Hamp1 null allele. Besides iron, Hamp1 null allele significantly elevated the levels of selenium in the liver, manganese in the liver and duodenum, and copper in the brain. Mice with conditional Hamp alleles will be useful to determine the tissue-specific regulation and functions of Hamp1 and Hamp2 in biometal homeostasis and other biological processes.
Collapse
Affiliation(s)
- S Lu
- Department of Internal Medicine, University of Nebraska Medical Center, 95820 UNMC, DRC I, Omaha, NE, 68198-5820, USA
| | | | | |
Collapse
|
64
|
Karsten CM, Laumonnier Y, Eurich B, Ender F, Bröker K, Roy S, Czabanska A, Vollbrandt T, Figge J, Köhl J. Monitoring and cell-specific deletion of C5aR1 using a novel floxed GFP-C5aR1 reporter knock-in mouse. THE JOURNAL OF IMMUNOLOGY 2015; 194:1841-55. [PMID: 25589074 DOI: 10.4049/jimmunol.1401401] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Many of the biological properties of C5a are mediated through activation of its receptor (C5aR1), the expression of which has been demonstrated convincingly on myeloid cells, such as neutrophils, monocytes, and macrophages. In contrast, conflicting results exist regarding C5aR1 expression in dendritic cells (DCs) and lymphoid lineage cells. In this article, we report the generation of a floxed GFP-C5aR1 reporter knock-in mouse. Using this mouse strain, we confirmed strong C5aR1 expression in neutrophils from bone marrow, blood, lung, and spleen, as well as in peritoneal macrophages. Further, we show C5aR1 expression in lung eosinophils, lung- and lamina propria-resident and alveolar macrophages, bone marrow-derived DCs, and lung-resident CD11b(+) and monocyte-derived DCs, whereas intestinal and pulmonary CD103(+) DCs stained negative. Also, some splenic NKT cells expressed GFP, whereas naive NK cells and B2 cells lacked GFP expression. Finally, we did not observe any C5aR1 expression in naive or activated CD4(+) Th cells in vitro or in vivo. Mating the floxed GFP-C5aR1 mouse strain with LysMCre mice, we were able to specifically delete C5aR1 in neutrophils and macrophages, whereas C5aR1 expression was retained in DCs. In summary, our findings suggest that C5aR1 expression in mice is largely restricted to cells of the myeloid lineage. The novel floxed C5aR1 reporter knock-in mouse will prove useful to track C5aR1 expression in experimental models of acute and chronic inflammation and to conditionally delete C5aR1 in immune cells.
Collapse
Affiliation(s)
- Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Benjamin Eurich
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Katharina Bröker
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Sreeja Roy
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Anna Czabanska
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | | | - Julia Figge
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| |
Collapse
|
65
|
Paster W, Bruger AM, Katsch K, Grégoire C, Roncagalli R, Fu G, Gascoigne NRJ, Nika K, Cohnen A, Feller SM, Simister PC, Molder KC, Cordoba SP, Dushek O, Malissen B, Acuto O. A THEMIS:SHP1 complex promotes T-cell survival. EMBO J 2014; 34:393-409. [PMID: 25535246 PMCID: PMC4339124 DOI: 10.15252/embj.201387725] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
THEMIS is critical for conventional T-cell development, but its precise molecular function remains elusive. Here, we show that THEMIS constitutively associates with the phosphatases SHP1 and SHP2. This complex requires the adapter GRB2, which bridges SHP to THEMIS in a Tyr-phosphorylation-independent fashion. Rather, SHP1 and THEMIS engage with the N-SH3 and C-SH3 domains of GRB2, respectively, a configuration that allows GRB2-SH2 to recruit the complex onto LAT. Consistent with THEMIS-mediated recruitment of SHP to the TCR signalosome, THEMIS knock-down increased TCR-induced CD3-ζ phosphorylation, Erk activation and CD69 expression, but not LCK phosphorylation. This generalized TCR signalling increase led to augmented apoptosis, a phenotype mirrored by SHP1 knock-down. Remarkably, a KI mutation of LCK Ser59, previously suggested to be key in ERK-mediated resistance towards SHP1 negative feedback, did not affect TCR signalling nor ligand discrimination in vivo. Thus, the THEMIS:SHP complex dampens early TCR signalling by a previously unknown molecular mechanism that favours T-cell survival. We discuss possible implications of this mechanism in modulating TCR output signals towards conventional T-cell development and differentiation.
Collapse
Affiliation(s)
- Wolfgang Paster
- T Cell Signalling Laboratory, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Annika M Bruger
- T Cell Signalling Laboratory, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Kristin Katsch
- T Cell Signalling Laboratory, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Claude Grégoire
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Romain Roncagalli
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Guo Fu
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| | - Nicholas R J Gascoigne
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Konstantina Nika
- T Cell Signalling Laboratory, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Andre Cohnen
- T Cell Signalling Laboratory, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Stephan M Feller
- Biological Systems Architecture Group, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK Tumor Biology Unit, Institute of Molecular Medicine, ZAMED, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Philip C Simister
- Biological Systems Architecture Group, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Kelly C Molder
- T Cell Signalling Laboratory, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Shaun-Paul Cordoba
- Molecular Immunology Group, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Omer Dushek
- Molecular Immunology Group, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France INSERM U1104, Marseille, France CNRS UMR7280, Marseille, France
| | - Oreste Acuto
- T Cell Signalling Laboratory, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
66
|
Anderson RJ, Tang CW, Daniels NJ, Compton BJ, Hayman CM, Johnston KA, Knight DA, Gasser O, Poyntz HC, Ferguson PM, Larsen DS, Ronchese F, Painter GF, Hermans IF. A self-adjuvanting vaccine induces cytotoxic T lymphocytes that suppress allergy. Nat Chem Biol 2014; 10:943-9. [DOI: 10.1038/nchembio.1640] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 08/04/2014] [Indexed: 01/12/2023]
|
67
|
Guéry L, Dubrot J, Lippens C, Brighouse D, Malinge P, Irla M, Pot C, Reith W, Waldburger JM, Hugues S. Ag-presenting CpG-activated pDCs prime Th17 cells that induce tumor regression. Cancer Res 2014; 74:6430-40. [PMID: 25252912 DOI: 10.1158/0008-5472.can-14-1149] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Plasmacytoid dendritic cells (pDC) rapidly and massively produce type I IFN and other inflammatory cytokines in response to foreign nucleic acids, thereby indirectly influencing T-cell responses. Moreover, antigen (Ag)-presenting pDCs directly regulate T-cell differentiation. Depending on the immune environment, pDCs exhibit either tolerogenic or immunogenic properties. Here, we show that CpG-activated pDCs promote efficient Th17 differentiation. Indeed, Th17 responses are defective in mice selectively lacking MHCII on pDCs upon antigenic challenge. Importantly, in those mice, the frequency of Th17 cells infiltrating solid tumors is impaired. As a result, the recruitment of infiltrating leukocytes in tumors, including tumor-specific cytotoxic T lymphocytes (CTL), is altered and results in increased tumor growth. Importantly, following immunization with tumor Ag and CpG-B, MHCII-restricted Ag presentation by pDCs promotes the differentiation of antitumor Th17 cells that induce intratumor CTL recruitment and subsequent regression of established tumors. Our results highlight a new role for Ag presenting activated pDCs in promoting the development of Th17 cells and impacting on antitumor immunity.
Collapse
Affiliation(s)
- Leslie Guéry
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Juan Dubrot
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Carla Lippens
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Dale Brighouse
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | | | - Magali Irla
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland. Centre d'immunology de Marseille Luminy, Université de la Méditerranée, Marseille, France
| | - Caroline Pot
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland. Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland
| | - Walter Reith
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Jean-Marc Waldburger
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland.
| |
Collapse
|
68
|
Vega-Ramos J, Roquilly A, Zhan Y, Young LJ, Mintern JD, Villadangos JA. Inflammation Conditions Mature Dendritic Cells To Retain the Capacity To Present New Antigens but with Altered Cytokine Secretion Function. THE JOURNAL OF IMMUNOLOGY 2014; 193:3851-9. [DOI: 10.4049/jimmunol.1303215] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
69
|
Dubrot J, Duraes FV, Potin L, Capotosti F, Brighouse D, Suter T, LeibundGut-Landmann S, Garbi N, Reith W, Swartz MA, Hugues S. Lymph node stromal cells acquire peptide-MHCII complexes from dendritic cells and induce antigen-specific CD4⁺ T cell tolerance. ACTA ACUST UNITED AC 2014; 211:1153-66. [PMID: 24842370 PMCID: PMC4042642 DOI: 10.1084/jem.20132000] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
LNSCs present peptide–MHCII complexes acquired from DCs to CD4+ T cells and induce T cell dysfunction by preventing their proliferation and survival. Dendritic cells (DCs), and more recently lymph node stromal cells (LNSCs), have been described to tolerize self-reactive CD8+ T cells in LNs. Although LNSCs express MHCII, it is unknown whether they can also impact CD4+ T cell functions. We show that the promoter IV (pIV) of class II transactivator (CIITA), the master regulator of MHCII expression, controls endogenous MHCII expression by LNSCs. Unexpectedly, LNSCs also acquire peptide–MHCII complexes from DCs and induce CD4+ T cell dysfunction by presenting transferred complexes to naive CD4+ T cells and preventing their proliferation and survival. Our data reveals a novel, alternative mechanism where LN-resident stromal cells tolerize CD4+ T cells through the presentation of self-antigens via transferred peptide–MHCII complexes of DC origin.
Collapse
Affiliation(s)
- Juan Dubrot
- Department of Pathology and Immunology, University of Geneva Medical School, 1211 Geneva, Switzerland
| | - Fernanda V Duraes
- Department of Pathology and Immunology, University of Geneva Medical School, 1211 Geneva, Switzerland
| | - Lambert Potin
- Laboratory of Lymphatic and Cancer Bioengineering, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Francesca Capotosti
- Laboratory of Lymphatic and Cancer Bioengineering, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Dale Brighouse
- Department of Pathology and Immunology, University of Geneva Medical School, 1211 Geneva, Switzerland
| | - Tobias Suter
- Department of Neurology, Section of Neuroimmunology and MS Research, University Hospital Zurich, 8091 Zurich, Switzerland
| | | | - Natalio Garbi
- Institute of Molecular Medicine and Institute of Experimental Immunology, University of Bonn, 53105 Bonn, Germany Institute of Molecular Medicine and Institute of Experimental Immunology, University of Bonn, 53105 Bonn, Germany
| | - Walter Reith
- Department of Pathology and Immunology, University of Geneva Medical School, 1211 Geneva, Switzerland
| | - Melody A Swartz
- Laboratory of Lymphatic and Cancer Bioengineering, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland ISREC, SV, EPFL, 1015 Lausanne, Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, University of Geneva Medical School, 1211 Geneva, Switzerland
| |
Collapse
|
70
|
Neunkirchner A, Schmetterer KG, Pickl WF. Lymphocyte-based model systems for allergy research: a historic overview. Int Arch Allergy Immunol 2014; 163:259-91. [PMID: 24777172 PMCID: PMC7617143 DOI: 10.1159/000360163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
During the last decades, a multitude of studies applying distinct in vitro and in vivo model systems have contributed greatly to our better understanding of the initiation and regulation of inflammatory processes leading to allergic diseases. Over the years, it has become evident that among lymphocytes, not only IgE-producing B cells and allergy-orchestrating CD4(+) helper cells but also cytotoxic CD8(+) T cells, γδ-T cells and innate lymphoid cells, as well as regulatory lymphocytes, might critically shape the immune response towards usually innocuous allergens. In this review, we provide a historic overview of pioneering work leading to the establishment of important lymphocyte-based model systems for allergy research. Moreover, we contrast the original findings with our currently more refined knowledge to appreciate the actual validity of the respective models and to reassess the conclusions obtained from them. Conflicting studies and interpretations are identified and discussed. The tables are intended to provide an easy overview of the field not only for scientists newly entering the field but also for the broader readership interested in updating their knowledge. Along those lines, herein we discuss in vitro and in vivo approaches to the investigation of lymphocyte effector cell activation, polarization and regulation, and describe depletion and adoptive transfer models along with gene knockout and transgenic (tg) methodologies. In addition, novel attempts to establish humanized T cell antigen receptor tg mouse models for allergy research are described and discussed.
Collapse
Affiliation(s)
- Alina Neunkirchner
- Christian Doppler Laboratory for Immunomodulation, Medical University of Vienna, Vienna, Austria
| | | | | |
Collapse
|
71
|
Zevnik B, Uyttersprot NC, Perez AV, Bothe GWM, Kern H, Kauselmann G. C57BL/6N albino/agouti mutant mice as embryo donors for efficient germline transmission of C57BL/6 ES cells. PLoS One 2014; 9:e90570. [PMID: 24599260 PMCID: PMC3944090 DOI: 10.1371/journal.pone.0090570] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 02/01/2014] [Indexed: 12/18/2022] Open
Abstract
We generated C57BL/6NTac mice carrying a tyrosinase loss-of function mutation and a reversion of the nonagouti locus to agouti. This strain has a high superovulation response, allows visual detection of chimeric coat color contribution of C57BL/6 ES-cells and provides a simplified breeding format that generates black G1 offspring of pure inbred C57BL/6 background in one step, providing the ideal host for genetically manipulated C57BL/6 ES cells.
Collapse
Affiliation(s)
- Branko Zevnik
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
- * E-mail:
| | | | - Ana V. Perez
- Taconic, Hudson, New York, United States of America
| | | | | | | |
Collapse
|
72
|
Roncagalli R, Hauri S, Fiore F, Liang Y, Chen Z, Sansoni A, Kanduri K, Joly R, Malzac A, Lähdesmäki H, Lahesmaa R, Yamasaki S, Saito T, Malissen M, Aebersold R, Gstaiger M, Malissen B. Quantitative proteomics analysis of signalosome dynamics in primary T cells identifies the surface receptor CD6 as a Lat adaptor-independent TCR signaling hub. Nat Immunol 2014; 15:384-392. [PMID: 24584089 DOI: 10.1038/ni.2843] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 02/03/2014] [Indexed: 02/08/2023]
Abstract
T cell antigen receptor (TCR)-mediated activation of T cells requires the interaction of dozens of proteins. Here we used quantitative mass spectrometry and activated primary CD4(+) T cells from mice in which a tag for affinity purification was knocked into several genes to determine the composition and dynamics of multiprotein complexes that formed around the kinase Zap70 and the adaptors Lat and SLP-76. Most of the 112 high-confidence time-resolved protein interactions we observed were previously unknown. The surface receptor CD6 was able to initiate its own signaling pathway by recruiting SLP-76 and the guanine nucleotide-exchange factor Vav1 regardless of the presence of Lat. Our findings provide a more complete model of TCR signaling in which CD6 constitutes a signaling hub that contributes to the diversification of TCR signaling.
Collapse
Affiliation(s)
- Romain Roncagalli
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France
| | - Simon Hauri
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.,Competence Center for Systems Physiology and Metabolic Diseases, ETH Zurich, Switzerland
| | - Fréderic Fiore
- Centre d'Immunophénomique, UM2 Aix-Marseille Université, Marseille, France.,INSERM US012, Marseille, France.,CNRS UMS3367, Marseille, France
| | - Yinming Liang
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France
| | - Zhi Chen
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, Turku, Finland
| | - Amandine Sansoni
- Centre d'Immunophénomique, UM2 Aix-Marseille Université, Marseille, France.,INSERM US012, Marseille, France.,CNRS UMS3367, Marseille, France
| | - Kartiek Kanduri
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, Turku, Finland
| | - Rachel Joly
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France
| | - Aurélie Malzac
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France
| | - Harri Lähdesmäki
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, Turku, Finland.,Department of Information and Computer Science, Aalto University, Finland
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Abo Akademi University, Turku, Finland
| | - Sho Yamasaki
- Division of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takashi Saito
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan
| | - Marie Malissen
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.,Faculty of Science, University of Zurich, Zurich, Switzerland
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.,Competence Center for Systems Physiology and Metabolic Diseases, ETH Zurich, Switzerland
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, UM2 Aix-Marseille Université, Marseille, France.,INSERM U1104, Marseille, France.,CNRS UMR7280, Marseille, France.,Centre d'Immunophénomique, UM2 Aix-Marseille Université, Marseille, France.,INSERM US012, Marseille, France.,CNRS UMS3367, Marseille, France
| |
Collapse
|
73
|
Hasan S, van der Veen DR, Winsky-Sommerer R, Hogben A, Laing EE, Koentgen F, Dijk DJ, Archer SN. A human sleep homeostasis phenotype in mice expressing a primate-specific PER3 variable-number tandem-repeat coding-region polymorphism. FASEB J 2014; 28:2441-54. [PMID: 24577121 PMCID: PMC4046067 DOI: 10.1096/fj.13-240135] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In humans, a primate-specific variable-number tandem-repeat (VNTR) polymorphism (4 or 5 repeats 54 nt in length) in the circadian gene PER3 is associated with differences in sleep timing and homeostatic responses to sleep loss. We investigated the effects of this polymorphism on circadian rhythmicity and sleep homeostasis by introducing the polymorphism into mice and assessing circadian and sleep parameters at baseline and during and after 12 h of sleep deprivation (SD). Microarray analysis was used to measure hypothalamic and cortical gene expression. Circadian behavior and sleep were normal at baseline. The response to SD of 2 electrophysiological markers of sleep homeostasis, electroencephalography (EEG) θ power during wakefulness and δ power during sleep, were greater in the Per35/5 mice. During recovery, the Per35/5 mice fully compensated for the SD-induced deficit in δ power, but the Per34/4 and wild-type mice did not. Sleep homeostasis-related transcripts (e.g., Homer1, Ptgs2, and Kcna2) were differentially expressed between the humanized mice, but circadian clock genes were not. These data are in accordance with the hypothesis derived from human data that the PER3 VNTR polymorphism modifies the sleep homeostatic response without significantly influencing circadian parameters.—Hasan, S., van der Veen, D. R., Winsky-Sommerer, R., Hogben, A., Laing, E. E., Koentgen, F., Dijk, D.-J., Archer, S. N. A human sleep homeostasis phenotype in mice expressing a primate-specific PER3 variable-number tandem-repeat coding-region polymorphism.
Collapse
Affiliation(s)
- Sibah Hasan
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK; and
| | - Daan R van der Veen
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK; and
| | | | - Alexandra Hogben
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK; and
| | - Emma E Laing
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK; and
| | | | - Derk-Jan Dijk
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK; and
| | - Simon N Archer
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK; and
| |
Collapse
|
74
|
Lehoczky JA, Thomas PE, Patrie KM, Owens KM, Villarreal LM, Galbraith K, Washburn J, Johnson CN, Gavino B, Borowsky AD, Millen KJ, Wakenight P, Law W, Van Keuren ML, Gavrilina G, Hughes ED, Saunders TL, Brihn L, Nadeau JH, Innis JW. A novel intergenic ETnII-β insertion mutation causes multiple malformations in polypodia mice. PLoS Genet 2013; 9:e1003967. [PMID: 24339789 PMCID: PMC3854779 DOI: 10.1371/journal.pgen.1003967] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 10/04/2013] [Indexed: 11/28/2022] Open
Abstract
Mouse early transposon insertions are responsible for ∼10% of spontaneous mutant phenotypes. We previously reported the phenotypes and genetic mapping of Polypodia, (Ppd), a spontaneous, X-linked dominant mutation with profound effects on body plan morphogenesis. Our new data shows that mutant mice are not born in expected Mendelian ratios secondary to loss after E9.5. In addition, we refined the Ppd genetic interval and discovered a novel ETnII-β early transposon insertion between the genes for Dusp9 and Pnck. The ETn inserted 1.6 kb downstream and antisense to Dusp9 and does not disrupt polyadenylation or splicing of either gene. Knock-in mice engineered to carry the ETn display Ppd characteristic ectopic caudal limb phenotypes, showing that the ETn insertion is the Ppd molecular lesion. Early transposons are actively expressed in the early blastocyst. To explore the consequences of the ETn on the genomic landscape at an early stage of development, we compared interval gene expression between wild-type and mutant ES cells. Mutant ES cell expression analysis revealed marked upregulation of Dusp9 mRNA and protein expression. Evaluation of the 5′ LTR CpG methylation state in adult mice revealed no correlation with the occurrence or severity of Ppd phenotypes at birth. Thus, the broad range of phenotypes observed in this mutant is secondary to a novel intergenic ETn insertion whose effects include dysregulation of nearby interval gene expression at early stages of development. Mobile genetic elements, particularly early transposons (ETn), cause malformations by inserting within genes leading to disruption of exons, splicing or polyadenylation. Few mutagenic early transposon insertions have been found outside genes and the effects of such insertions on surrounding gene regulation is poorly understood. We discovered a novel intergenic ETnII-β insertion in the mouse mutant Polypodia (Ppd). We reproduced the mutant phenotype after engineering the mutation in wild-type cells with homologous recombination, proving that this early transposon insertion is Ppd. Mutant mice are not born in expected Mendelian ratios secondary to loss after E9.5. Embryonic stem cells from mutant mice show upregulated transcription of an adjacent gene, Dusp9. Thus, at an early and critical stage of development, dysregulated gene transcription is one consequence of the insertion mutation. DNA methylation of the ETn 5′ LTR is not correlated with phenotypic outcome in mutant mice. Polypodia is an example of an intergenic mobile element insertion in mice causing dramatic morphogenetic defects and fetal death.
Collapse
Affiliation(s)
- Jessica A. Lehoczky
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Peedikayil E. Thomas
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
- Pediatrics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kevin M. Patrie
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kailey M. Owens
- Pediatrics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lisa M. Villarreal
- Pediatrics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kenneth Galbraith
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Joe Washburn
- Biomedical Research Core Facilities, DNA Sequencing Core Lab, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Craig N. Johnson
- Biomedical Research Core Facilities, DNA Sequencing Core Lab, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Bryant Gavino
- Murine Molecular Constructs Laboratory-MMCL Mouse Biology Program, University of California, Davis, California, United States of America
| | - Alexander D. Borowsky
- University of California, Davis, Center for Comparative Medicine and Comprehensive Cancer Center, Department of Pathology and Laboratory Medicine, Davis, California, United States of America
| | - Kathleen J. Millen
- Division of Genetic Medicine, Department of Pediatrics, Seattle Children's Hospital, Seattle, Washington, United States of America
| | - Paul Wakenight
- Division of Genetic Medicine, Department of Pediatrics, Seattle Children's Hospital, Seattle, Washington, United States of America
| | - William Law
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Margaret L. Van Keuren
- Transgenic Animal Model Core Lab, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Galina Gavrilina
- Transgenic Animal Model Core Lab, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Elizabeth D. Hughes
- Transgenic Animal Model Core Lab, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Thomas L. Saunders
- Transgenic Animal Model Core Lab, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lesil Brihn
- Department of Genetics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Joseph H. Nadeau
- Pacific Northwest Research Institute, Seattle, Washington, United States of America
| | - Jeffrey W. Innis
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
- Pediatrics, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
75
|
Mizuta R, Araki S, Furukawa M, Furukawa Y, Ebara S, Shiokawa D, Hayashi K, Tanuma SI, Kitamura D. DNase γ is the effector endonuclease for internucleosomal DNA fragmentation in necrosis. PLoS One 2013; 8:e80223. [PMID: 24312463 PMCID: PMC3846476 DOI: 10.1371/journal.pone.0080223] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 10/01/2013] [Indexed: 11/25/2022] Open
Abstract
Apoptosis and necrosis, two major forms of cell death, can be distinguished morphologically and biochemically. Internucleosomal DNA fragmentation (INDF) is a biochemical hallmark of apoptosis, and caspase-activated DNase (CAD), also known as DNA fragmentation factor 40 kDa (DFF40), is one of the major effector endonucleases. DNase γ, a Mg(2+)/Ca(2+)-dependent endonuclease, is also known to generate INDF but its role among other apoptosis-associated endonucleases in cell death is unclear. Here we show that (i) INDF occurs even during necrosis in cell lines, primary cells, and in tissues of mice in vivo, and (ii) DNase γ, but not CAD, is the effector endonuclease for INDF in cells undergoing necrosis. These results document a previously unappreciated role for INDF in necrosis and define its molecular basis.
Collapse
Affiliation(s)
- Ryushin Mizuta
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Shinsuke Araki
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Makoto Furukawa
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Yuki Furukawa
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Syota Ebara
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Daisuke Shiokawa
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Katsuhiko Hayashi
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Sei-ichi Tanuma
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Daisuke Kitamura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
- Department of Molecular Immunology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| |
Collapse
|
76
|
Gennequin B, Otte DM, Zimmer A. CRISPR/Cas-induced double-strand breaks boost the frequency of gene replacements for humanizing the mouse Cnr2 gene. Biochem Biophys Res Commun 2013; 441:815-9. [DOI: 10.1016/j.bbrc.2013.10.138] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 10/26/2013] [Indexed: 11/28/2022]
|
77
|
A missing PD-L1/PD-1 coinhibition regulates diabetes induction by preproinsulin-specific CD8 T-cells in an epitope-specific manner. PLoS One 2013; 8:e71746. [PMID: 23977133 PMCID: PMC3747217 DOI: 10.1371/journal.pone.0071746] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/02/2013] [Indexed: 11/19/2022] Open
Abstract
Coinhibitory PD-1/PD-L1 (B7-H1) interactions provide critical signals for the regulation of autoreactive T-cell responses. We established mouse models, expressing the costimulator molecule B7.1 (CD80) on pancreatic beta cells (RIP-B7.1 tg mice) or are deficient in coinhibitory PD-L1 or PD-1 molecules (PD-L1−/− and PD-1−/− mice), to study induction of preproinsulin (ppins)-specific CD8 T-cell responses and experimental autoimmune diabetes (EAD) by DNA-based immunization. RIP-B7.1 tg mice allowed us to identify two CD8 T-cell specificities: pCI/ppins DNA exclusively induced Kb/A12–21-specific CD8 T-cells and EAD, whereas pCI/ppinsΔA12–21 DNA (encoding ppins without the COOH-terminal A12–21 epitope) elicited Kb/B22–29-specific CD8 T-cells and EAD. Specific expression/processing of mutant ppinsΔA12–21 (but not ppins) in non-beta cells, targeted by intramuscular DNA-injection, thus facilitated induction of Kb/B22–29-specific CD8 T-cells. The A12–21 epitope binds Kb molecules with a very low avidity as compared with B22–29. Interestingly, immunization of coinhibition-deficient PD-L1−/− or PD-1−/− mice with pCI/ppins induced Kb/A12–21-monospecific CD8 T-cells and EAD but injections with pCI/ppinsΔA12–21 did neither recruit Kb/B22–29-specific CD8 T-cells into the pancreatic target tissue nor induce EAD. PpinsΔA12–21/(Kb/B22–29)-mediated EAD was efficiently restored in RIP-B7.1+/PD-L1−/− mice, differing from PD-L1−/− mice only in the tg B7.1 expression in beta cells. Alternatively, an ongoing beta cell destruction and tissue inflammation, initiated by ppins/(Kb/A12–21)-specific CD8 T-cells in pCI/ppins+pCI/ppinsΔA12–21 co-immunized PD-L1−/− mice, facilitated the expansion of ppinsΔA12–21/(Kb/B22–29)-specific CD8 T-cells. CD8 T-cells specific for the high-affinity Kb/B22–29- (but not the low-affinity Kb/A12–21)-epitope thus require stimulatory ´help from beta cells or inflamed islets to expand in PD-L1-deficient mice. The new PD-1/PD-L1 diabetes models may be valuable tools to study under well controlled experimental conditions distinct hierarchies of autoreactive CD8 T-cell responses, which trigger the initial steps of beta cell destruction or emerge during the pathogenic progression of EAD.
Collapse
|
78
|
Colley SM, Wintle L, Searles R, Russell V, Firman RC, Smith S, DeBoer K, Merriner DJ, Genevieve B, Bentel JM, Stuckey BGA, Phillips MR, Simmons LW, de Kretser DM, O'Bryan MK, Leedman PJ. Loss of the nuclear receptor corepressor SLIRP compromises male fertility. PLoS One 2013; 8:e70700. [PMID: 23976951 PMCID: PMC3744554 DOI: 10.1371/journal.pone.0070700] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 06/20/2013] [Indexed: 11/24/2022] Open
Abstract
Nuclear receptors (NRs) and their coregulators play fundamental roles in initiating and directing gene expression influencing mammalian reproduction, development and metabolism. SRA stem Loop Interacting RNA-binding Protein (SLIRP) is a Steroid receptor RNA Activator (SRA) RNA-binding protein that is a potent repressor of NR activity. SLIRP is present in complexes associated with NR target genes in the nucleus; however, it is also abundant in mitochondria where it affects mitochondrial mRNA transcription and energy turnover. In further characterisation studies, we observed SLIRP protein in the testis where its localization pattern changes from mitochondrial in diploid cells to peri-acrosomal and the tail in mature sperm. To investigate the in vivo effects of SLIRP, we generated a SLIRP knockout (KO) mouse. This animal is viable, but sub-fertile. Specifically, when homozygous KO males are crossed with wild type (WT) females the resultant average litter size is reduced by approximately one third compared with those produced by WT males and females. Further, SLIRP KO mice produced significantly fewer progressively motile sperm than WT animals. Electron microscopy identified disruption of the mid-piece/annulus junction in homozygous KO sperm and altered mitochondrial morphology. In sum, our data implicates SLIRP in regulating male fertility, wherein its loss results in asthenozoospermia associated with compromised sperm structure and mitochondrial morphology.
Collapse
Affiliation(s)
- Shane M. Colley
- Laboratory for Cancer Medicine, The University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
| | - Larissa Wintle
- Laboratory for Cancer Medicine, The University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
| | | | - Victoria Russell
- Laboratory for Cancer Medicine, The University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
| | - Renee C. Firman
- Centre for Evolutionary Biology, School of Animal Biology, The University of Western Australia, Crawley, Australia
| | - Stephanie Smith
- Male Infertility and Germ Cell Biology Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Kathleen DeBoer
- Male Infertility and Germ Cell Biology Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - D. Jo Merriner
- Male Infertility and Germ Cell Biology Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Ben Genevieve
- Keogh Institute for Medical Research, Sir Charles Gairdner Hospital, Nedlands, Australia
| | - Jacqueline M. Bentel
- Anatomical Pathology, PathWest Laboratory Medicine, Royal Perth Hospital, Perth, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Australia
| | - Bronwyn G. A. Stuckey
- Keogh Institute for Medical Research, Sir Charles Gairdner Hospital, Nedlands, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Australia
| | - Michael R. Phillips
- Laboratory for Cancer Medicine, The University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Australia
| | - Leigh W. Simmons
- Centre for Evolutionary Biology, School of Animal Biology, The University of Western Australia, Crawley, Australia
| | - David M. de Kretser
- Male Infertility and Germ Cell Biology Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Moira K. O'Bryan
- Male Infertility and Germ Cell Biology Laboratory, Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Peter J. Leedman
- Laboratory for Cancer Medicine, The University of Western Australia Centre for Medical Research, Western Australian Institute for Medical Research, Perth, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Australia
- * E-mail:
| |
Collapse
|
79
|
C-reactive protein (CRP) is essential for efficient systemic transduction of recombinant adeno-associated virus vector 1 (rAAV-1) and rAAV-6 in mice. J Virol 2013; 87:10784-91. [PMID: 23903832 DOI: 10.1128/jvi.01813-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The clinical relevance of gene therapy using the recombinant adeno-associated virus (rAAV) vectors often requires widespread distribution of the vector, and in this case, systemic delivery is the optimal route of administration. Humoral blood factors, such as antibodies or complement, are the first barriers met by the vectors administered systemically. We have found that other blood proteins, galectin 3 binding protein (G3BP) and C-reactive protein (CRP), can interact with different AAV serotypes in a species-specific manner. While interactions of rAAV vectors with G3BP, antibodies, or complement lead to a decrease in vector efficacy, systemic transduction of the CRP-deficient mouse and its respective control clearly established that binding to mouse CRP (mCRP) boosts rAAV vector 1 (rAAV-1) and rAAV-6 transduction efficiency in skeletal muscles over 10 times. Notably, the high efficacy of rAAV-6 in CRP-deficient mice can be restored by reconstitution of the CRP-deficient mouse with mCRP. Human CRP (hCRP) does not interact with either rAAV-1 or rAAV-6, and, consequently, the high efficiency of mCRP-mediated muscle transduction by these serotypes in mice cannot be translated to humans. No interaction of mCRP or hCRP was observed with rAAV-8 and rAAV-9. We show, for the first time, that serum components can significantly enhance rAAV-mediated tissue transduction in a serotype- and species-specific manner. Bioprocessing in body fluids should be considered when transfer of a preclinical proof of concept for AAV-based gene therapy to humans is planned.
Collapse
|
80
|
Suzuki J, Denning DP, Imanishi E, Horvitz HR, Nagata S. Xk-Related Protein 8 and CED-8 Promote Phosphatidylserine Exposure in Apoptotic Cells. Science 2013; 341:403-6. [DOI: 10.1126/science.1236758] [Citation(s) in RCA: 370] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
81
|
Alsharifi M, Koskinen A, Wijesundara DK, Bettadapura J, Müllbacher A. MHC class II-alpha chain knockout mice support increased viral replication that is independent of their lack of MHC class II cell surface expression and associated immune function deficiencies. PLoS One 2013; 8:e68458. [PMID: 23840854 PMCID: PMC3695910 DOI: 10.1371/journal.pone.0068458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 06/03/2013] [Indexed: 01/14/2023] Open
Abstract
MHCII molecules are heterodimeric cell surface proteins composed of an α and β chain. These molecules are almost exclusively expressed on thymic epithelium and antigen presenting cells (APCs) and play a central role in the development and function of CD4 T cells. Various MHC-II knockout mice have been generated including MHC-IIAα-/- (I-Aα-/-), MHC-IIAβ-/- (I-β-/-) and the double knockout (I-Aαxβ-/-). Here we report a very striking observation, namely that alphaviruses including the avirulent strain of Semliki Forest virus (aSFV), which causes asymptomatic infection in wild-type C57BL6/J (B6) mice, causes a very acute and lethal infection in I-Aα-/-, but not in I-β-/- or I-Aαxβ-/-, mice. This susceptibility to aSFV is associated with high virus titres in muscle, spleen, liver, and brain compared to B6 mice. In addition, I-Aα-/- mice show intact IFN-I responses in terms of IFN-I serum levels and IFN-I receptor expression and function. Radiation bone marrow chimeras of B6 mice reconstituted with I-Aα-/- bone marrow expressed B6 phenotype, whereas radiation chimeras of I-Aα-/- mice reconstituted with B6 bone marrow expressed the phenotype of high viral susceptibility. Virus replication experiments both in vivo and in vitro showed enhanced virus growth in tissues and cell cultures derived form I-Aα-/- compared to B6 mice. This enhanced virus replication is evident for other alpha-, flavi- and poxviruses and may be of great benefit to producers of viral vaccines. In conclusion, I-Aα-/- mice exhibit a striking susceptibility to virus infections independent of their defective MHC-II expression. Detailed genetic analysis will be carried out to characterise the underlining genetic defects responsible for the observed phenomenon.
Collapse
Affiliation(s)
- Mohammed Alsharifi
- Department of Immunology, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
- * E-mail: (MA); (AM)
| | - Aulikki Koskinen
- Department of Immunology, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Danushka K. Wijesundara
- Department of Immunology, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Jayaram Bettadapura
- Department of Immunology, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Arno Müllbacher
- Department of Immunology, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
- * E-mail: (MA); (AM)
| |
Collapse
|
82
|
Ngo D, Beaulieu E, Gu R, Leaney A, Santos L, Fan H, Yang Y, Kao W, Xu J, Escriou V, Loiler S, Vervoordeldonk MJ, Morand EF. Divergent Effects of Endogenous and Exogenous Glucocorticoid-Induced Leucine Zipper in Animal Models of Inflammation and Arthritis. ACTA ACUST UNITED AC 2013; 65:1203-12. [DOI: 10.1002/art.37858] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 01/03/2013] [Indexed: 12/26/2022]
|
83
|
Bolinger B, Sims S, O’Hara G, de Lara C, Tchilian E, Firner S, Engeler D, Ludewig B, Klenerman P. A new model for CD8+ T cell memory inflation based upon a recombinant adenoviral vector. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:4162-74. [PMID: 23509359 PMCID: PMC3672979 DOI: 10.4049/jimmunol.1202665] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
CD8(+) T cell memory inflation, first described in murine CMV (MCMV) infection, is characterized by the accumulation of high-frequency, functional Ag-specific CD8(+) T cell pools with an effector-memory phenotype and enrichment in peripheral organs. Although persistence of Ag is considered essential, the rules underpinning memory inflation are still unclear. The MCMV model is, however, complicated by the virus's low-level persistence and stochastic reactivation. We developed a new model of memory inflation based on a β-galactosidase (βgal)-recombinant adenovirus vector. After i.v. administration in C57BL/6 mice, we observed marked memory inflation in the βgal96 epitope, whereas a second epitope, βgal497, undergoes classical memory formation. The inflationary T cell responses show kinetics, distribution, phenotype, and functions similar to those seen in MCMV and are reproduced using alternative routes of administration. Memory inflation in this model is dependent on MHC class II. As in MCMV, only the inflating epitope showed immunoproteasome independence. These data define a new model for memory inflation, which is fully replication independent, internally controlled, and reproduces the key immunologic features of the CD8(+) T cell response. This model provides insight into the mechanisms responsible for memory inflation and, because it is based on a vaccine vector, also is relevant to novel T cell-inducing vaccines in humans.
Collapse
Affiliation(s)
- Beatrice Bolinger
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Stuart Sims
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Geraldine O’Hara
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Catherine de Lara
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Elma Tchilian
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Sonja Firner
- Institute of Immunobiology, Cantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland
| | - Daniel Engeler
- Institute of Immunobiology, Cantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Cantonal Hospital St. Gallen, CH-9007 St. Gallen, Switzerland
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| |
Collapse
|
84
|
Ngo D, Cheng Q, O′Connor AE, DeBoer KD, Lo CY, Beaulieu E, De Seram M, Hobbs RM, O′Bryan MK, Morand EF. Glucocorticoid-induced leucine zipper (GILZ) regulates testicular FOXO1 activity and spermatogonial stem cell (SSC) function. PLoS One 2013; 8:e59149. [PMID: 23516608 PMCID: PMC3597624 DOI: 10.1371/journal.pone.0059149] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 02/12/2013] [Indexed: 01/08/2023] Open
Abstract
Spermatogonia stem cell (SSC) self-renewal and differentiation are tightly regulated processes that ensure a continued production of mature sperm throughout male adulthood. In the present study, we investigated the role of glucocorticoid-induced leucine zipper (GILZ) in maintenance of the male germline and spermatogenesis. GILZ was detectable in germ cells of wild type mice on the day of birth, suggesting a role for GILZ in prospermatogonia and SSC pool formation. Gilz KO mice were generated and adult males were azoospermic and sterile. During the first wave of spermatogenesis in Gilz KO mice, spermatogenesis arrested part way through pachytene of meiosis I. Subsequent waves resulted in a progressive depletion of germ cells through apoptosis to ultimately produce a Sertoli cell-only phenotype. Further, in contrast to wild type littermates, PLZF+ cells were detected in the peri-luminal region of Gilz KO mice at day 6 post-natal, suggesting a defect in prospermatogonia migration in the absence of GILZ. At age 30 days, transient accumulation of PLZF+ cells in a subset of tubules and severely compromised spermatogenesis were observed in Gilz KO mice, consistent with defective SSC differentiation. GILZ deficiency was associated with an increase in FOXO1 transcriptional activity, which leads to activation of a selective set of FOXO1 target genes, including a pro-apoptotic protein, BIM. On the other hand, no evidence of a heightened immune response was observed. Together, these results suggest that GILZ suppresses FOXO1 nuclear translocation, promotes SSC differentiation over self-renewal, and favours germ cell survival through inhibition of BIM-dependent pro-apoptotic signals. These findings provide a mechanism for the effects of GILZ on spermatogenesis and strengthen the case for GILZ being a critical molecule in the regulation of male fertility.
Collapse
Affiliation(s)
- Devi Ngo
- Department of Medicine, Centre for Inflammatory Diseases, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| | - Qiang Cheng
- Department of Medicine, Centre for Inflammatory Diseases, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
- * E-mail: (QC); (EM)
| | - Anne E. O′Connor
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Kathleen D. DeBoer
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Camden Y. Lo
- Monash Micro Imaging, Monash University, Clayton, Victoria, Australia
| | - Elaine Beaulieu
- Department of Medicine, Centre for Inflammatory Diseases, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| | - Mia De Seram
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Robin M. Hobbs
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Moira K. O′Bryan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Eric F. Morand
- Department of Medicine, Centre for Inflammatory Diseases, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
- * E-mail: (QC); (EM)
| |
Collapse
|
85
|
Abstract
The ability to introduce DNA sequences (e.g., genes) of interest into the germline genome has rendered the mouse a powerful and indispensable experimental model in fundamental and medical research. The DNA sequences can be integrated into the genome randomly or into a specific locus by homologous recombination, in order to: (1) delete or insert mutations into genes of interest to determine their function, (2) introduce human genes into the genome of mice to generate animal models enabling study of human-specific genes and diseases, e.g., mice susceptible to infections by human-specific pathogens of interest, (3) introduce individual genes or genomes of pathogens (such as viruses) in order to examine the contributions of such genes to the pathogenesis of the parent pathogens, (4) and last but not least introduce reporter genes that allow monitoring in vivo or ex vivo the expression of genes of interest. Furthermore, the use of recombination systems, such as Cre/loxP or FRT/FLP, enables conditional induction or suppression of gene expression of interest in a restricted period of mouse's lifetime, in a particular cell type, or in a specific tissue. In this review, we will give an updated summary of the gene targeting technology and discuss some important considerations in the design of gene-targeted mice.
Collapse
Affiliation(s)
- Hicham Bouabe
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | | |
Collapse
|
86
|
Wei Y, Li L, Zhou X, Zhang QY, Dunbar A, Liu F, Kluetzman K, Yang W, Ding X. Generation and characterization of a novel Cyp2a(4/5)bgs-null mouse model. Drug Metab Dispos 2013; 41:132-40. [PMID: 23073733 PMCID: PMC3533424 DOI: 10.1124/dmd.112.048736] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 10/15/2012] [Indexed: 12/30/2022] Open
Abstract
Knockout mouse models targeting various cytochrome P450 (P450 or CYP) genes are valuable for determining P450's biologic functions, including roles in drug metabolism and chemical toxicity. In this study, a novel Cyp2a(4/5)bgs-null mouse model was generated, in which a 1.2-megabase pair genomic fragment containing nine Cyp genes in mouse chromosome 7 (including, sequentially, Cyp2a5, 2g1, 2b19, 2b23, 2a4, 2b9, 2b13, 2b10, and 2s1) are deleted, through Cre-mediated recombination in vivo. The resultant mouse strain was viable and fertile, without any developmental deficits or morphologic abnormalities. Deletion of the constitutive genes in the cluster was confirmed by polymerase chain reaction analysis of the genes and the mRNAs in tissues known to express each gene. The loss of this gene cluster led to significant decreases in microsomal activities toward testosterone hydroxylation in various tissues examined, including olfactory mucosa (OM), lung, liver, and brain. In addition, systemic clearance of pentobarbital was decreased in Cyp2a(4/5)bgs-null mice, as indicated by >60% increases in pentobarbital-induced sleeping time, compared with wild-type (WT) mice. This novel Cyp2a(4/5)bgs-null mouse model will be valuable for in vivo studies of drug metabolism and chemical toxicities in various tissues, including the liver, lung, brain, intestine, kidney, skin, and OM, where one or more of the targeted Cyp genes are known to be expressed in WT mice. The model will also be valuable for preparation of humanized mice that express human CYP2A6, CYP2A13, CYP2B6, or CYP2S1, and as a knockout mouse model for five non-P450 genes (Vmn1r184, Nalp9c, Nalp4a, Nalp9a, and Vmn1r185) that were also deleted.
Collapse
Affiliation(s)
- Yuan Wei
- Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12201-0509, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Antigen recognition by autoreactive CD4⁺ thymocytes drives homeostasis of the thymic medulla. PLoS One 2012; 7:e52591. [PMID: 23300712 PMCID: PMC3531460 DOI: 10.1371/journal.pone.0052591] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 11/16/2012] [Indexed: 01/07/2023] Open
Abstract
The thymic medulla is dedicated for purging the T-cell receptor (TCR) repertoire of self-reactive specificities. Medullary thymic epithelial cells (mTECs) play a pivotal role in this process because they express numerous peripheral tissue-restricted self-antigens. Although it is well known that medulla formation depends on the development of single-positive (SP) thymocytes, the mechanisms underlying this requirement are incompletely understood. We demonstrate here that conventional SP CD4+ thymocytes bearing autoreactive TCRs drive a homeostatic process that fine-tunes medullary plasticity in adult mice by governing the expansion and patterning of the medulla. This process exhibits strict dependence on TCR-reactivity with self-antigens expressed by mTECs, as well as engagement of the CD28-CD80/CD86 costimulatory axis. These interactions induce the expression of lymphotoxin α in autoreactive CD4+ thymocytes and RANK in mTECs. Lymphotoxin in turn drives mTEC development in synergy with RANKL and CD40L. Our results show that Ag-dependent interactions between autoreactive CD4+ thymocytes and mTECs fine-tune homeostasis of the medulla by completing the signaling axes implicated in mTEC expansion and medullary organization.
Collapse
|
88
|
Irla M, Guenot J, Sealy G, Reith W, Imhof BA, Sergé A. Three-dimensional visualization of the mouse thymus organization in health and immunodeficiency. THE JOURNAL OF IMMUNOLOGY 2012; 190:586-96. [PMID: 23248258 DOI: 10.4049/jimmunol.1200119] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lymphoid organs exhibit complex structures tightly related to their function. Surprisingly, although the thymic medulla constitutes a specialized microenvironment dedicated to the induction of T cell tolerance, its three-dimensional topology remains largely elusive because it has been studied mainly in two dimensions using thymic sections. To overcome this limitation, we have developed an automated method for full organ reconstruction in three dimensions, allowing visualization of intact mouse lymphoid organs from a collection of immunolabeled slices. We validated full organ reconstruction in three dimensions by reconstructing the well-characterized structure of skin-draining lymph nodes, before revisiting the complex and poorly described corticomedullary organization of the thymus. Wild-type thymi contain ~200 small medullae that are connected to or separated from a major medullary compartment. In contrast, thymi of immunodeficient Rag2(-/-) mice exhibit only ~20 small, unconnected medullary islets. Upon total body irradiation, medullary complexity was partially reduced and then recovered upon bone marrow transplantation. This intricate topology presents fractal properties, resulting in a considerable corticomedullary area. This feature ensures short distances between cortex and medulla, hence efficient thymocyte migration, as assessed by mathematical models. Remarkably, this junction is enriched, particularly in neonates, in medullary thymic epithelial cells expressing the autoimmune regulator. The emergence of a major medullary compartment is induced by CD4(+) thymocytes via CD80/86 and lymphotoxin-α signals. This comprehensive three-dimensional view of the medulla emphasizes a complex topology favoring efficient interactions between developing T cells and autoimmune regulator-positive medullary thymic epithelial cells, a key process for central tolerance induction.
Collapse
Affiliation(s)
- Magali Irla
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.
| | | | | | | | | | | |
Collapse
|
89
|
Hunn MK, Farrand KJ, Broadley KWR, Weinkove R, Ferguson P, Miller RJ, Field CS, Petersen T, McConnell MJ, Hermans IF. Vaccination with irradiated tumor cells pulsed with an adjuvant that stimulates NKT cells is an effective treatment for glioma. Clin Cancer Res 2012; 18:6446-59. [PMID: 23147997 DOI: 10.1158/1078-0432.ccr-12-0704] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The prognosis for patients with glioblastoma multiforme (GBM) remains extremely poor despite recent treatment advances. There is an urgent need to develop novel therapies for this disease. EXPERIMENTAL DESIGN We used the implantable GL261 murine glioma model to investigate the therapeutic potential of a vaccine consisting of intravenous injection of irradiated whole tumor cells pulsed with the immuno-adjuvant α-galactosylceramide (α-GalCer). RESULTS Vaccine treatment alone was highly effective in a prophylactic setting. In a more stringent therapeutic setting, administration of one dose of vaccine combined with depletion of regulatory T cells (Treg) resulted in 43% long-term survival and the disappearance of mass lesions detected by MRI. Mechanistically, the α-GalCer component was shown to act by stimulating "invariant" natural killer-like T cells (iNKT cells) in a CD1d-restricted manner, which in turn supported the development of a CD4(+) T-cell-mediated adaptive immune response. Pulsing α-GalCer onto tumor cells avoided the profound iNKT cell anergy induced by free α-GalCer. To investigate the potential for clinical application of this vaccine, the number and function of iNKT cells was assessed in patients with GBM and shown to be similar to age-matched healthy volunteers. Furthermore, irradiated GBM tumor cells pulsed with α-GalCer were able to stimulate iNKT cells and augment a T-cell response in vitro. CONCLUSIONS Injection of irradiated tumor cells loaded with α-GalCer is a simple procedure that could provide effective immunotherapy for patients with high-grade glioma.
Collapse
Affiliation(s)
- Martin K Hunn
- Malaghan Institute of Medical Research, University of Otago, New Zealand.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Espéli M, Clatworthy MR, Bökers S, Lawlor KE, Cutler AJ, Köntgen F, Lyons PA, Smith KGC. Analysis of a wild mouse promoter variant reveals a novel role for FcγRIIb in the control of the germinal center and autoimmunity. ACTA ACUST UNITED AC 2012; 209:2307-19. [PMID: 23109709 PMCID: PMC3501356 DOI: 10.1084/jem.20121752] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Genetic variants of the inhibitory Fc receptor FcγRIIb have been associated with systemic lupus erythematosus in humans and mice. The mechanism by which Fcgr2b variants contribute to the development of autoimmunity is unknown and was investigated by knocking in the most commonly conserved wild mouse Fcgr2b promoter haplotype, also associated with autoimmune-prone mouse strains, into the C57BL/6 background. We found that in the absence of an AP-1-binding site in its promoter, FcγRIIb failed to be up-regulated on activated and germinal center (GC) B cells. This resulted in enhanced GC responses, increased affinity maturation, and autoantibody production. Accordingly, in the absence of FcγRIIb activation-induced up-regulation, mice developed more severe collagen-induced arthritis and spontaneous glomerular immune complex deposition. Our data highlight how natural variation in Fcgr2b drives the development of autoimmune disease. They also show how the study of such variants using a knockin approach can provide insight into immune mechanisms not possible using conventional genetic manipulation, in this case demonstrating an unexpected critical role for the activation-induced up-regulation of FcγRIIb in controlling affinity maturation, autoantibody production, and autoimmunity.
Collapse
Affiliation(s)
- Marion Espéli
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 OXY, England, UK
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Kaji T, Ishige A, Hikida M, Taka J, Hijikata A, Kubo M, Nagashima T, Takahashi Y, Kurosaki T, Okada M, Ohara O, Rajewsky K, Takemori T. Distinct cellular pathways select germline-encoded and somatically mutated antibodies into immunological memory. ACTA ACUST UNITED AC 2012; 209:2079-97. [PMID: 23027924 PMCID: PMC3478929 DOI: 10.1084/jem.20120127] [Citation(s) in RCA: 221] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
B cell memory is generated along two fundamentally distinct cellular differentiation pathways. One component of memory in the antibody system is long-lived memory B cells selected for the expression of somatically mutated, high-affinity antibodies in the T cell–dependent germinal center (GC) reaction. A puzzling observation has been that the memory B cell compartment also contains cells expressing unmutated, low-affinity antibodies. Using conditional Bcl6 ablation, we demonstrate that these cells are generated through proliferative expansion early after immunization in a T cell–dependent but GC-independent manner. They soon become resting and long-lived and display a novel distinct gene expression signature which distinguishes memory B cells from other classes of B cells. GC-independent memory B cells are later joined by somatically mutated GC descendants at roughly equal proportions and these two types of memory cells efficiently generate adoptive secondary antibody responses. Deletion of T follicular helper (Tfh) cells significantly reduces the generation of mutated, but not unmutated, memory cells early on in the response. Thus, B cell memory is generated along two fundamentally distinct cellular differentiation pathways. One pathway is dedicated to the generation of high-affinity somatic antibody mutants, whereas the other preserves germ line antibody specificities and may prepare the organism for rapid responses to antigenic variants of the invading pathogen.
Collapse
Affiliation(s)
- Tomohiro Kaji
- Laboratory for Immunological Memory, RIKEN Research Center for Allergy and Immunology, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Lehmann FM, Feicht S, Helm F, Maurberger A, Ladinig C, Zimber-Strobl U, Kühn R, Mautner J, Gerbitz A, Bornkamm GW. Humanized c-Myc mouse. PLoS One 2012; 7:e42021. [PMID: 22860051 PMCID: PMC3409231 DOI: 10.1371/journal.pone.0042021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 07/02/2012] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND A given tumor is usually dependent on the oncogene that is activated in the respective tumor entity. This phenomenon called oncogene addiction provides the rationale for attempts to target oncogene products in a therapeutic manner, be it by small molecules, by small interfering RNAs (siRNA) or by antigen-specific T cells. As the proto-oncogene product is required also for the function of normal cells, this raises the question whether there is a therapeutic window between the adverse effects of specific inhibitors or T cells to normal tissue that may limit their application, and their beneficial tumor-specific therapeutic action. To address this crucial question, suitable mouse strains need to be developed, that enable expression of the human proto-oncogene not only in tumor but also in normal cells. The aim of this work is to provide such a mouse strain for the human proto-oncogene product c-MYC. PRINCIPAL FINDINGS We generated C57BL/6-derived embryonic stem cells that are transgenic for a humanized c-Myc gene and established a mouse strain (hc-Myc) that expresses human c-MYC instead of the murine ortholog. These transgenic animals harbor the humanized c-Myc gene integrated into the endogenous murine c-Myc locus. Despite the lack of the endogenous murine c-Myc gene, homozygous mice show a normal phenotype indicating that human c-MYC can replace its murine ortholog. CONCLUSIONS The newly established hc-Myc mouse strain provides a model system to study in detail the adverse effects of therapies that target the human c-MYC protein. To mimic the clinical situation, hc-Myc mice may be cross-bred to mice that develop tumors due to overexpression of human c-MYC. With these double transgenic mice it will be possible to study simultaneously the therapeutic efficiency and adverse side effects of MYC-specific therapies in the same mouse.
Collapse
Affiliation(s)
- Frank M. Lehmann
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Center Munich, Munich, Germany
| | - Samantha Feicht
- Department of Gene Vectors, Helmholtz Center Munich, Munich, Germany
| | - Florian Helm
- Department of Immunology, Charité Berlin, Berlin, Germany
| | - Anna Maurberger
- Department of Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Camilla Ladinig
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Center Munich, Munich, Germany
| | | | - Ralf Kühn
- Institute of Developmental Genetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Josef Mautner
- Department of Pediatrics, Technical University (TU) Munich and Clinical Cooperation Group Pediatric Tumor Immunology, TU Munich and Helmholtz Center Munich, Munich, Germany
| | - Armin Gerbitz
- Department of Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Georg W. Bornkamm
- Institute of Clinical Molecular Biology and Tumor Genetics, Helmholtz Center Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
93
|
Notini AJ, McClive PJ, Meachem SJ, van den Bergen JA, Western PS, Gustin SE, Harley VR, Koopman P, Sinclair AH. Redd1 is a novel marker of testis development but is not required for normal male reproduction. Sex Dev 2012; 6:223-30. [PMID: 22797524 DOI: 10.1159/000339723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2012] [Indexed: 11/19/2022] Open
Abstract
In an effort to identify novel candidate genes involved in testis determination, we previously used suppression subtraction hybridisation PCR on male and female whole embryonic (12.0-12.5 days post coitum) mouse gonads. One gene to emerge from our screen was Redd1. In the current study, we demonstrate by whole-mount in situ hybridisation that Redd1 is differentially expressed in the developing mouse gonad at the time of sex determination, with higher expression in testis than ovary. Furthermore, Redd1 expression was first detected as Sry expression peaks, immediately prior to morphological sex determination, suggesting a potential role for Redd1 during testis development. To determine the functional importance of this gene during testis development, we generated Redd1-deficient mice. Morphologically, Redd1-deficient mice were indistinguishable from control littermates and showed normal fertility. Our results show that Redd1 alone is not required for testis development or fertility in mice. The lack of a male reproductive phenotype in Redd1 mice may be due to functional compensation by the related gene Redd2.
Collapse
Affiliation(s)
- A J Notini
- Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Vic., Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Hinkle KM, Yue M, Behrouz B, Dächsel JC, Lincoln SJ, Bowles EE, Beevers JE, Dugger B, Winner B, Prots I, Kent CB, Nishioka K, Lin WL, Dickson DW, Janus CJ, Farrer MJ, Melrose HL. LRRK2 knockout mice have an intact dopaminergic system but display alterations in exploratory and motor co-ordination behaviors. Mol Neurodegener 2012; 7:25. [PMID: 22647713 PMCID: PMC3441373 DOI: 10.1186/1750-1326-7-25] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 04/27/2012] [Indexed: 11/14/2022] Open
Abstract
Mutations in the LRRK2 gene are the most common cause of genetic Parkinson’s disease. Although the mechanisms behind the pathogenic effects of LRRK2 mutations are still not clear, data emerging from in vitro and in vivo models suggests roles in regulating neuronal polarity, neurotransmission, membrane and cytoskeletal dynamics and protein degradation. We created mice lacking exon 41 that encodes the activation hinge of the kinase domain of LRRK2. We have performed a comprehensive analysis of these mice up to 20 months of age, including evaluation of dopamine storage, release, uptake and synthesis, behavioral testing, dendritic spine and proliferation/neurogenesis analysis. Our results show that the dopaminergic system was not functionally comprised in LRRK2 knockout mice. However, LRRK2 knockout mice displayed abnormal exploratory activity in the open-field test. Moreover, LRRK2 knockout mice stayed longer than their wild type littermates on the accelerated rod during rotarod testing. Finally, we confirm that loss of LRRK2 caused degeneration in the kidney, accompanied by a progressive enhancement of autophagic activity and accumulation of autofluorescent material, but without evidence of biphasic changes.
Collapse
Affiliation(s)
- Kelly M Hinkle
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Murine CD4+ T cell responses are inhibited by cytotoxic T cell-mediated killing of dendritic cells and are restored by antigen transfer. PLoS One 2012; 7:e37481. [PMID: 22649530 PMCID: PMC3359309 DOI: 10.1371/journal.pone.0037481] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 04/24/2012] [Indexed: 12/21/2022] Open
Abstract
Cytotoxic T lymphocytes (CTL) provide protection against pathogens and tumors. In addition, experiments in mouse models have shown that CTL can also kill antigen-presenting dendritic cells (DC), reducing their ability to activate primary and secondary CD8(+) T cell responses. In contrast, the effects of CTL-mediated killing on CD4(+) T cell responses have not been fully investigated. Here we use adoptive transfer of TCR transgenic T cells and DC immunization to show that specific CTL significantly inhibited CD4(+) T cell proliferation induced by DC loaded with peptide or low concentrations of protein antigen. In contrast, CTL had little effect on CD4(+) T cell proliferation induced by DC loaded with high protein concentrations or expressing antigen endogenously, even if these DC were efficiently killed and failed to accumulate in the lymph node (LN). Residual CD4(+) T cell proliferation was due to the transfer of antigen from carrier DC to host APC, and predominantly involved skin DC populations. Importantly, the proliferating CD4(+) T cells also developed into IFN-γ producing memory cells, a property normally requiring direct presentation by activated DC. Thus, CTL-mediated DC killing can inhibit CD4(+) T cell proliferation, with the extent of inhibition being determined by the form and amount of antigen used to load DC. In the presence of high antigen concentrations, antigen transfer to host DC enables the generation of CD4(+) T cell responses regardless of DC killing, and suggests mechanisms whereby CD4(+) T cell responses can be amplified.
Collapse
|
96
|
Abstract
The dramatic increase in the amount of research data being produced necessarily leads to higher demands on statistical thresholds and on experimental planning. This is to avoid positive selection of multiple tested data. Here we would like to highlight the need for including littermate controls in animal experiments, in particular when genetically modified strains are analysed for quantitative phenotypes. Thus, this suggestion will have impact on most immunological experiments performed today.
Collapse
|
97
|
Gilley J, Seereeram A, Ando K, Mosely S, Andrews S, Kerschensteiner M, Misgeld T, Brion JP, Anderton B, Hanger DP, Coleman MP. Age-dependent axonal transport and locomotor changes and tau hypophosphorylation in a "P301L" tau knockin mouse. Neurobiol Aging 2012; 33:621.e1-621.e15. [PMID: 21492964 DOI: 10.1016/j.neurobiolaging.2011.02.014] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 02/03/2011] [Accepted: 02/20/2011] [Indexed: 12/19/2022]
Abstract
Tauopathies are characterized by hyperphosphorylation of the microtubule-associated protein tau and its accumulation into fibrillar aggregates. Toxic effects of aggregated tau and/or dysfunction of soluble tau could both contribute to neural defects in these neurodegenerative diseases. We have generated a novel knockin mouse model of an inherited tauopathy, frontotemporal dementia with parkinsonism linked to tau mutations on chromosome 17 (FTDP-17T). We incorporated a single mutation, homologous to the common FTDP-17T P301L mutation, directly into the endogenous mouse gene, mimicking the human disease situation. These mice express P301L-equivalent mutant tau at normal physiological levels from the knockin allele. Importantly, in contrast to existing transgenic mouse models that overexpress human P301L mutant tau, no overt tau pathology developed during the normal lifespan of the knockin mice. In fact, overall phosphorylation of tau was reduced, perhaps due to reduced microtubule binding. However, homozygous knockin mice did display intriguing age-dependent changes in axonal transport of mitochondria, and increased spontaneous locomotor activity in old age. These could represent early consequences of the tau dysfunction that eventually precipitates pathogenesis in humans.
Collapse
|
98
|
IAPs limit activation of RIP kinases by TNF receptor 1 during development. EMBO J 2012; 31:1679-91. [PMID: 22327219 DOI: 10.1038/emboj.2012.18] [Citation(s) in RCA: 165] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 01/11/2012] [Indexed: 12/17/2022] Open
Abstract
Inhibitor of apoptosis (IAP) proteins cIAP1, cIAP2, and XIAP (X-linked IAP) regulate apoptosis and cytokine receptor signalling, but their overlapping functions make it difficult to distinguish their individual roles. To do so, we deleted the genes for IAPs separately and in combination. While lack of any one of the IAPs produced no overt phenotype in mice, deletion of cIap1 with cIap2 or Xiap resulted in mid-embryonic lethality. In contrast, Xiap(-/-)cIap2(-/-) mice were viable. The death of cIap2(-/-)cIap1(-/-) double mutants was rescued to birth by deletion of tumour necrosis factor (TNF) receptor 1, but not TNFR2 genes. Remarkably, hemizygosity for receptor-interacting protein kinase 1 (Ripk1) allowed Xiap(-/-)cIap1(-/-) double mutants to survive past birth, and prolonged cIap2(-/-)cIap1(-/-) embryonic survival. Similarly, deletion of Ripk3 was able to rescue the mid-gestation defect of cIap2(-/-)cIap1(-/-) embryos, as these embryos survived to E15.5. cIAPs are therefore required during development to limit activity of RIP kinases in the TNF receptor 1 signalling pathway.
Collapse
|
99
|
Khan M, Vaes E, Mombaerts P. Regulation of the probability of mouse odorant receptor gene choice. Cell 2012; 147:907-21. [PMID: 22078886 DOI: 10.1016/j.cell.2011.09.049] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 08/29/2011] [Accepted: 09/19/2011] [Indexed: 11/18/2022]
Abstract
Each olfactory sensory neuron (OSN) in mouse chooses one of 1,200 odorant receptor (OR) genes for expression. OR genes are chosen for expression by greatly varying numbers of OSNs. The mechanisms that regulate the probability of OR gene choice remain unclear. Here, we have applied the NanoString platform of fluorescent barcodes and digital readout to measure RNA levels of 577 OR genes in a single reaction, with probes designed against coding sequences. In an inbred mouse strain with a targeted deletion in the P element, we find that this element regulates OR gene choice differentially across its cluster of 24 OR genes. Importantly, the fold changes of NanoString counts in ΔP or ΔH mice are in very close agreement with the fold changes of cell counts, determined by in situ hybridization. Thus, the P and H elements regulate the probability of OR gene choice, not OR transcript level per OSN.
Collapse
Affiliation(s)
- Mona Khan
- Max Planck Institute of Biophysics, 60438 Frankfurt, Germany
| | | | | |
Collapse
|
100
|
Wray J, Hartmann C. WNTing embryonic stem cells. Trends Cell Biol 2011; 22:159-68. [PMID: 22196214 DOI: 10.1016/j.tcb.2011.11.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 11/08/2011] [Accepted: 11/14/2011] [Indexed: 01/02/2023]
Abstract
Embryonic stem cells (ESCs) - undifferentiated cells originating from preimplantation stage embryos - have prolonged self-renewal capacity and are pluripotent. Activation of the canonical Wnt pathway is implicated in maintenance of and exit from the pluripotent state. Recent findings demonstrate that the essential mediator of canonical Wnt signaling, β-catenin, is dispensable for ESC maintenance; however, its activation inhibits differentiation through derepression of T cell factor 3 (Tcf3)-bound genes. Wnt agonists are useful in deriving ESCs from recalcitrant mouse strains and the rat and in nuclear reprogramming of somatic stem cells. We discuss recent advances in our understanding of the role of canonical Wnt signaling in the regulation of ESC self-renewal and how its manipulation can improve pluripotent ESC derivation and maintenance.
Collapse
Affiliation(s)
- Jason Wray
- University College London, Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | | |
Collapse
|