1
|
Hosang L, Löhndorf A, Dohle W, Rosche A, Marry S, Diercks BP, Müller-Kirschbaum LC, Flügel LT, Potter BVL, Odoardi F, Guse AH, Flügel A. 2-Methoxyestradiol-3,17-O,O-bis-sulfamate inhibits store-operated Ca 2+ entry in T lymphocytes and prevents experimental autoimmune encephalomyelitis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119485. [PMID: 37150482 DOI: 10.1016/j.bbamcr.2023.119485] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/09/2023]
Abstract
Ca2+ signaling is one of the essential signaling systems for T lymphocyte activation, the latter being an essential step in the pathogenesis of autoimmune diseases such as multiple sclerosis (MS). Store-operated Ca2+ entry (SOCE) ensures long lasting Ca2+ signaling and is of utmost importance for major downstream T lymphocyte activation steps, e.g. nuclear localization of the transcription factor 'nuclear factor of activated T cells' (NFAT). 2-Methoxyestradiol (2ME2), an endogenous metabolite of estradiol (E2), blocks nuclear translocation of NFAT. The likely underlying mechanism is inhibition of SOCE, as shown for its synthetic sulfamate ester analogue 2-ethyl-3-sulfamoyloxy-17β-cyanomethylestra-1,3,5(10)-triene (STX564). Here, we demonstrate that another synthetic bis-sulfamoylated 2ME2 derivative, 2-methoxyestradiol-3,17-O,O-bis-sulfamate (2-MeOE2bisMATE, STX140), an orally bioavailable, multi-targeting anticancer agent and potent steroid sulfatase (STS) inhibitor, antagonized SOCE in T lymphocytes. Downstream events, e.g. secretion of the pro-inflammatory cytokines interferon-γ and interleukin-17, were decreased by STX140 in in vitro experiments. Remarkably, STX140 dosed in vivo completely blocked the clinical disease in both active and transfer experimental autoimmune encephalomyelitis (EAE) in Lewis rats, a T cell-mediated animal model for MS, at a dose of 10 mg/kg/day i.p., whereas neither 2ME2 nor Irosustat, a pure STS inhibitor, showed any effect. The STS inhibitory activity of STX140 is therefore not responsible for its activity in this model. Taken together, inhibition of SOCE by STX140 resulting in full antagonism of clinical symptoms in EAE in the Lewis rat, paired with the known excellent bioavailability and pharmaceutical profile of this drug, open potentially new therapeutic avenues for the treatment of MS.
Collapse
Affiliation(s)
- Leon Hosang
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Anke Löhndorf
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Wolfgang Dohle
- Drug Discovery & Medicinal Chemistry, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Anette Rosche
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Stephen Marry
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Lukas C Müller-Kirschbaum
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Lioba T Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany; Department of Neurology, University Medical Center Göttingen, Robert-Koch-Straße 40, D-37075 Göttingen, Germany
| | - Barry V L Potter
- Drug Discovery & Medicinal Chemistry, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Francesca Odoardi
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany.
| |
Collapse
|
2
|
Li J, Lu L, Binder K, Xiong J, Ye L, Cheng YH, Majri-Morrison S, Lu W, Lee JW, Zhang Z, Wu YZ, Zheng L, Lenardo MJ. Mechanisms of antigen-induced reversal of CNS inflammation in experimental demyelinating disease. SCIENCE ADVANCES 2023; 9:eabo2810. [PMID: 36857453 PMCID: PMC9977187 DOI: 10.1126/sciadv.abo2810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 12/27/2022] [Indexed: 06/18/2023]
Abstract
Autoimmune central nervous system (CNS) demyelinating diseases are a major public health burden and poorly controlled by current immunosuppressants. More precise immunotherapies with higher efficacy and fewer side effects are sought. We investigated the effectiveness and mechanism of an injectable myelin-based antigenic polyprotein MMPt (myelin oligodendrocyte glycoprotein, myelin basic protein and proteolipid protein, truncated). We find that it suppresses mouse experimental autoimmune encephalomyelitis without major side effects. MMPt induces rapid apoptosis of the encephalitogenic T cells and suppresses inflammation in the affected CNS. Intravital microscopy shows that MMPt is taken up by perivascular F4/80+ cells but not conventional antigen-presenting dendritic cells, B cells, or microglia. MMPt-stimulated F4/80+ cells induce reactive T cell immobilization and apoptosis in situ, resulting in reduced infiltration of inflammatory cells and chemokine production. Our study reveals alternative mechanisms that explain how cognate antigen suppresses CNS inflammation and may be applicable for effectively and safely treating demyelinating diseases.
Collapse
Affiliation(s)
- Jian Li
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lisen Lu
- MoE Key Laboratory for Biomedical Photonics, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Kyle Binder
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jian Xiong
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan H. Cheng
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sonia Majri-Morrison
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wei Lu
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jae W. Lee
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zhihong Zhang
- MoE Key Laboratory for Biomedical Photonics, Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yu-zhang Wu
- Institute of Immunology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lixin Zheng
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael J. Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- NIAID Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3
|
Zheng P, Wei X, Cao X, Ma P, Dong R, Tang H, Meng X, Liu X, Zhang C, Zhang S, Ming L. Antigen clearance at the peak of the primary immune response induces experimental autoimmune encephalomyelitis. Eur J Immunol 2023; 53:e2250122. [PMID: 36597350 DOI: 10.1002/eji.202250122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/02/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023]
Abstract
Autoimmune demyelinating diseases can be induced by an immune response against myelin peptides; however, the exact mechanism underlying the development of such diseases remains unclear. In experimental autoimmune encephalomyelitis, we found that the clearance of exogenous myelin antigen at the peak of the primary immune response is key to the pathogenesis of the disease. The generation of effector T cells requires continuous antigen stimulation, whereas redundant antigen traps and exhausts effector T cells in the periphery, which induces resistance to the disease. Moreover, insufficient antigenic stimulation fails to induce disease efficiently owing to insufficient numbers of effector T cells. When myelin antigen is entirely cleared, the number of effector T cells reaches a peak, which facilitates infiltration of more effector T cells into the central nervous system. The peripheral antigen clearance initiates the first wave of effector T cell entry into the central nervous system and induces chronic inflammation. The inflamed central nervous system recruits the second wave of effector T cells that worsen inflammation, resulting in loss of self-tolerance. These findings provide new insights into the mechanism underlying the development of autoimmune demyelinating diseases, which may potentially impact future treatments.
Collapse
Affiliation(s)
- Peiguo Zheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Xufeng Wei
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Xuezhen Cao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Panhong Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Rui Dong
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Hongwei Tang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Xianchun Meng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Xinjing Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Cai Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| | - Shuijun Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery & Digestive Organ Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Liang Ming
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, P. R. China
| |
Collapse
|
4
|
Distinct roles of the meningeal layers in CNS autoimmunity. Nat Neurosci 2022; 25:887-899. [PMID: 35773544 DOI: 10.1038/s41593-022-01108-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/23/2022] [Indexed: 12/27/2022]
Abstract
The meninges, comprising the leptomeninges (pia and arachnoid layers) and the pachymeninx (dura layer), participate in central nervous system (CNS) autoimmunity, but their relative contributions remain unclear. Here we report on findings in animal models of CNS autoimmunity and in patients with multiple sclerosis, where, in acute and chronic disease, the leptomeninges were highly inflamed and showed structural changes, while the dura mater was only marginally affected. Although dural vessels were leakier than leptomeningeal vessels, effector T cells adhered more weakly to the dural endothelium. Furthermore, local antigen-presenting cells presented myelin and neuronal autoantigens less efficiently, and the activation of autoreactive T cells was lower in dural than leptomeningeal layers, preventing local inflammatory processes. Direct antigen application was required to evoke a local inflammatory response in the dura. Together, our data demonstrate an uneven involvement of the meningeal layers in CNS autoimmunity, in which effector T cell trafficking and activation are functionally confined to the leptomeninges, while the dura remains largely excluded from CNS autoimmune processes.
Collapse
|
5
|
Hosang L, Canals RC, van der Flier FJ, Hollensteiner J, Daniel R, Flügel A, Odoardi F. The lung microbiome regulates brain autoimmunity. Nature 2022; 603:138-144. [PMID: 35197636 DOI: 10.1038/s41586-022-04427-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/17/2022] [Indexed: 12/13/2022]
Abstract
Lung infections and smoking are risk factors for multiple sclerosis, a T-cell-mediated autoimmune disease of the central nervous system1. In addition, the lung serves as a niche for the disease-inducing T cells for long-term survival and for maturation into migration-competent effector T cells2. Why the lung tissue in particular has such an important role in an autoimmune disease of the brain is not yet known. Here we detected a tight interconnection between the lung microbiota and the immune reactivity of the brain. A dysregulation in the lung microbiome significantly influenced the susceptibility of rats to developing autoimmune disease of the central nervous system. Shifting the microbiota towards lipopolysaccharide-enriched phyla by local treatment with neomycin induced a type-I-interferon-primed state in brain-resident microglial cells. Their responsiveness towards autoimmune-dominated stimulation by type II interferons was impaired, which led to decreased proinflammatory response, immune cell recruitment and clinical signs. Suppressing lipopolysaccharide-producing lung phyla with polymyxin B led to disease aggravation, whereas addition of lipopolysaccharide-enriched phyla or lipopolysaccharide recapitulated the neomycin effect. Our data demonstrate the existence of a lung-brain axis in which the pulmonary microbiome regulates the immune reactivity of the central nervous tissue and thereby influences its susceptibility to autoimmune disease development.
Collapse
Affiliation(s)
- Leon Hosang
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Roger Cugota Canals
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Felicia Joy van der Flier
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | | | - Rolf Daniel
- Department of Genomic and Applied Microbiology, University of Göttingen, Göttingen, Germany
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany.
| | - Francesca Odoardi
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany. .,Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
6
|
Löhndorf A, Hosang L, Dohle W, Odoardi F, Waschkowski SA, Rosche A, Bauche A, Winzer R, Tolosa E, Windhorst S, Marry S, Flügel A, Potter BVL, Diercks BP, Guse AH. 2-Methoxyestradiol and its derivatives inhibit store-operated Ca 2+ entry in T cells: Identification of a new and potent inhibitor. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118988. [PMID: 33581218 PMCID: PMC8062851 DOI: 10.1016/j.bbamcr.2021.118988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 12/15/2022]
Abstract
T cell activation starts with formation of second messengers that release Ca2+ from the endoplasmic reticulum (ER) and thereby activate store-operated Ca2+ entry (SOCE), one of the essential signals for T cell activation. Recently, the steroidal 2-methoxyestradiol was shown to inhibit nuclear translocation of the nuclear factor of activated T cells (NFAT). We therefore investigated 2-methoxyestradiol for inhibition of Ca2+ entry in T cells, screened a library of 2-methoxyestradiol analogues, and characterized the derivative 2-ethyl-3-sulfamoyloxy-17β-cyanomethylestra-1,3,5(10)-triene (STX564) as a novel, potent and specific SOCE inhibitor. STX564 inhibits Ca2+ entry via SOCE without affecting other ion channels and pumps involved in Ca2+ signaling in T cells. Downstream effects such as cytokine expression and cell proliferation were also inhibited by both 2-methoxyestradiol and STX564, which has potential as a new chemical biology tool.
Collapse
Affiliation(s)
- Anke Löhndorf
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Leon Hosang
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Wolfgang Dohle
- Drug Discovery & Medicinal Chemistry, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Francesca Odoardi
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Sissy-Alina Waschkowski
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Anette Rosche
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Andreas Bauche
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Riekje Winzer
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Sabine Windhorst
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Stephen Marry
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Barry V L Potter
- Drug Discovery & Medicinal Chemistry, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Björn-Philipp Diercks
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Andreas H Guse
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.
| |
Collapse
|
7
|
Visualizing context-dependent calcium signaling in encephalitogenic T cells in vivo by two-photon microscopy. Proc Natl Acad Sci U S A 2017; 114:E6381-E6389. [PMID: 28716943 DOI: 10.1073/pnas.1701806114] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In experimental autoimmune encephalitis (EAE), autoimmune T cells are activated in the periphery before they home to the CNS. On their way, the T cells pass through a series of different cellular milieus where they receive signals that instruct them to invade their target tissues. These signals involve interaction with the surrounding stroma cells, in the presence or absence of autoantigens. To portray the serial signaling events, we studied a T-cell-mediated model of EAE combining in vivo two-photon microscopy with two different activation reporters, the FRET-based calcium biosensor Twitch1 and fluorescent NFAT. In vitro activated T cells first settle in secondary (2°) lymphatic tissues (e.g., the spleen) where, in the absence of autoantigen, they establish transient contacts with stroma cells as indicated by sporadic short-lived calcium spikes. The T cells then exit the spleen for the CNS where they first roll and crawl along the luminal surface of leptomeningeal vessels without showing calcium activity. Having crossed the blood-brain barrier, the T cells scan the leptomeningeal space for autoantigen-presenting cells (APCs). Sustained contacts result in long-lasting calcium activity and NFAT translocation, a measure of full T-cell activation. This process is sensitive to anti-MHC class II antibodies. Importantly, the capacity to activate T cells is not a general property of all leptomeningeal phagocytes, but varies between individual APCs. Our results identify distinct checkpoints of T-cell activation, controlling the capacity of myelin-specific T cells to invade and attack the CNS. These processes may be valuable therapeutic targets.
Collapse
|
8
|
Späth T, Popp M, Pérez León C, Marz M, Hoffmann-Vogel R. Near-equilibrium measurement of quantum size effects using Kelvin probe force microscopy. NANOSCALE 2017; 9:7868-7874. [PMID: 28555693 DOI: 10.1039/c7nr01874f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In nano-structures such as thin films electron confinement results in the quantization of energy levels in the direction perpendicular to the film. The discretization of the energy levels leads to the oscillatory dependence of many properties on the film thickness due to quantum size effects. Pb on Si(111) is a specially interesting system because a particular relationship between the Pb atomic layer thickness and its Fermi wavelength leads to a periodicity of the oscillation of two atomic layers. Here, we demonstrate how the combination of scanning force microscopy (SFM) and Kelvin probe force microscopy (KPFM) provides a reliable method to monitor the quantum oscillations in the work function of Pb ultra-thin film nano-structures on Si(111). Unlike other techniques, with SFM/KPFM we directly address single Pb islands, determine their height while suppressing the influence of electrostatic forces, and, in addition, simultaneously evaluate their local work function by measurements close to equilibrium, without current-dependent and non-equilibrium effects. Our results evidence even-odd oscillations in the work function as a function of the film thickness that decay linearly with the film thickness, proving that this method provides direct and precise information on the quantum states.
Collapse
Affiliation(s)
- Thomas Späth
- Physikalisches Institut, Karlsruhe Institute of Technology (KIT), Wolfgang-Gaede-Str. 1, 76128 Karlsruhe, Germany.
| | | | | | | | | |
Collapse
|
9
|
Rossi B, Constantin G. Live Imaging of Immune Responses in Experimental Models of Multiple Sclerosis. Front Immunol 2016; 7:506. [PMID: 27917173 PMCID: PMC5116921 DOI: 10.3389/fimmu.2016.00506] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/01/2016] [Indexed: 12/31/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is the most common animal model of multiple sclerosis (MS), a chronic inflammatory autoimmune disease of the central nervous system (CNS) characterized by multifocal perivascular infiltrates that predominantly comprise lymphocytes and macrophages. During EAE, autoreactive T cells first become active in the secondary lymphoid organs upon contact with antigen-presenting cells (APCs), and then gain access to CNS parenchyma, through a compromised blood–brain barrier, subsequently inducing inflammation and demyelination. Two-photon laser scanning microscopy (TPLSM) is an ideal tool for intravital imaging because of its low phototoxicity, deep tissue penetration, and high resolution. In the last decade, TPLSM has been used to visualize the behavior of T cells and their contact with APCs in the lymph nodes (LNs) and target tissues in several models of autoimmune diseases. The leptomeninges and cerebrospinal fluid represent particularly important points for T cell entry into the CNS and reactivation following contact with local APCs during the preclinical phase of EAE. In this review, we highlight recent findings concerning the pathogenesis of EAE and MS, emphasizing the use of TPLSM to characterize T cell activation in the LNs and CNS, as well as the mechanisms of tolerance induction. Furthermore, we discuss how advanced imaging unveils disease mechanisms and helps to identify novel therapeutic strategies to treat CNS autoimmunity and inflammation.
Collapse
Affiliation(s)
- Barbara Rossi
- Section of General Pathology, Department of Medicine, University of Verona , Verona , Italy
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona , Verona , Italy
| |
Collapse
|
10
|
Kawakami N. In vivo imaging in autoimmune diseases in the central nervous system. Allergol Int 2016; 65:235-42. [PMID: 26935215 DOI: 10.1016/j.alit.2016.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 11/18/2022] Open
Abstract
Intravital imaging is becoming more popular and is being used to visualize cellular motility and functions. In contrast to in vitro analysis, which resembles in vivo analysis, intravital imaging can be used to observe and analyze cells directly in vivo. In this review, I will summarize recent imaging studies of autoreactive T cell infiltration into the central nervous system (CNS) and provide technical background. During their in vivo journey, autoreactive T cells interact with many different cells. At first, autoreactive T cells interact with endothelial cells in the airways of the lung or with splenocytes, where they acquire a migratory phenotype to infiltrate into the CNS. After arriving at the CNS, they interact with endothelial cells of the leptomeningeal vessels or the choroid plexus before passing through the blood-brain barrier. CNS-infiltrating T cells become activated by recognizing endogenous autoantigens presented by local antigen-presenting cells (APCs). This activation was visualized in vivo by using protein-based sensors. One such sensor detects changes in intracellular calcium concentration as an early marker of T cell activation. Another sensor detects translocation of Nuclear factor of activated T-cells (NFAT) from cytosol to nucleus as a definitive sign of T cell activation. Importantly, intravital imaging is not just used to visualize cellular behavior. Together with precise analysis, intravital imaging deepens our knowledge of cellular functions in living organs and also provides a platform for developing therapeutic treatments.
Collapse
Affiliation(s)
- Naoto Kawakami
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians Universitaet Muenchen, Munich, Germany; Neuroimmunology Group, Max-Planck Institute of Neurobiology, Martinsried, Germany.
| |
Collapse
|
11
|
Antigen-oriented T cell migration contributes to myelin peptide induced-EAE and immune tolerance. Clin Immunol 2016; 169:36-46. [PMID: 27327113 DOI: 10.1016/j.clim.2016.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 05/05/2016] [Accepted: 06/07/2016] [Indexed: 02/08/2023]
Abstract
Treatment with soluble myelin peptide can efficiently and specifically induce tolerance to demyelination autoimmune diseases including multiple sclerosis, however the mechanism underlying this therapeutic effect remains to be elucidated. In actively induced mouse model of experimental autoimmune encephalomyelitis (EAE) we analyzed T cell and innate immune cell responses in the central nervous system (CNS) and spleen after intraperitoneal (i.p.) infusion of myelin oligodendrocyte glycoprotein (MOG). We found that i.p. MOG infusion blocked effector T cell recruitment to the CNS and protected mice from EAE and lymphoid organ atrophy. Innate immune CD11b(+) cells preferentially recruited MOG-specific effector T cells, particularly when activated to become competent antigen presenting cells (APCs). During EAE development, mature APCs were enriched in the CNS rather than in the spleen, attracting effector T cells to the CNS. Increased myelin antigen exposure induced CNS-APC maturation, recruiting additional effector T cells to the CNS, causing symptoms of disease. MOG triggered functional maturation of splenic APCs. MOG presenting APCs interacted with MOG-specific T cells in the spleen, aggregating to cluster around CD11b(+) cells, and were trapped in the periphery. This process was MHC II dependent as an MHC II directed antibody blocked CD4(+) T cell cluster formation. These findings highlight the role of myelin peptide-loaded APCs in myelin peptide-induced EAE and immune tolerance.
Collapse
|
12
|
Schläger C, Körner H, Krueger M, Vidoli S, Haberl M, Mielke D, Brylla E, Issekutz T, Cabañas C, Nelson PJ, Ziemssen T, Rohde V, Bechmann I, Lodygin D, Odoardi F, Flügel A. Effector T-cell trafficking between the leptomeninges and the cerebrospinal fluid. Nature 2016; 530:349-53. [PMID: 26863192 DOI: 10.1038/nature16939] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/17/2015] [Indexed: 12/18/2022]
Abstract
In multiple sclerosis, brain-reactive T cells invade the central nervous system (CNS) and induce a self-destructive inflammatory process. T-cell infiltrates are not only found within the parenchyma and the meninges, but also in the cerebrospinal fluid (CSF) that bathes the entire CNS tissue. How the T cells reach the CSF, their functionality, and whether they traffic between the CSF and other CNS compartments remains hypothetical. Here we show that effector T cells enter the CSF from the leptomeninges during Lewis rat experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. While moving through the three-dimensional leptomeningeal network of collagen fibres in a random Brownian walk, T cells were flushed from the surface by the flow of the CSF. The detached cells displayed significantly lower activation levels compared to T cells from the leptomeninges and CNS parenchyma. However, they did not represent a specialized non-pathogenic cellular sub-fraction, as their gene expression profile strongly resembled that of tissue-derived T cells and they fully retained their encephalitogenic potential. T-cell detachment from the leptomeninges was counteracted by integrins VLA-4 and LFA-1 binding to their respective ligands produced by resident macrophages. Chemokine signalling via CCR5/CXCR3 and antigenic stimulation of T cells in contact with the leptomeningeal macrophages enforced their adhesiveness. T cells floating in the CSF were able to reattach to the leptomeninges through steps reminiscent of vascular adhesion in CNS blood vessels, and invade the parenchyma. The molecular/cellular conditions for T-cell reattachment were the same as the requirements for detachment from the leptomeningeal milieu. Our data indicate that the leptomeninges represent a checkpoint at which activated T cells are licensed to enter the CNS parenchyma and non-activated T cells are preferentially released into the CSF, from where they can reach areas of antigen availability and tissue damage.
Collapse
Affiliation(s)
- Christian Schläger
- Institute of Neuroimmunology, Institute for Multiple Sclerosis Research, University Medical Centre Göttingen, 37073 Göttingen, Germany
| | - Henrike Körner
- Institute of Neuroimmunology, Institute for Multiple Sclerosis Research, University Medical Centre Göttingen, 37073 Göttingen, Germany
| | - Martin Krueger
- Institute of Anatomy, University of Leipzig, 04103 Leipzig, Germany
| | - Stefano Vidoli
- Department of Structural and Geotechnical Engineering, University of Rome La Sapienza, 00185 Rome, Italy
| | - Michael Haberl
- Institute of Neuroimmunology, Institute for Multiple Sclerosis Research, University Medical Centre Göttingen, 37073 Göttingen, Germany
| | - Dorothee Mielke
- Department Neurosurgery, University Medical Centre Göttingen, 37075 Göttingen, Germany
| | - Elke Brylla
- Institute of Anatomy, University of Leipzig, 04103 Leipzig, Germany
| | - Thomas Issekutz
- Division of Immunology, Department of Pediatrics Dalhousie University, Halifax B3H 4R2, Canada
| | - Carlos Cabañas
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa, 28049 Madrid, Spain
| | - Peter J Nelson
- Medical Clinic and Policlinic IV, Ludwig-Maximilians-University of Munich, 80336 Munich, Germany
| | - Tjalf Ziemssen
- Department of Neurology, University Hospital, 01307 Dresden, Germany
| | - Veit Rohde
- Department Neurosurgery, University Medical Centre Göttingen, 37075 Göttingen, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, 04103 Leipzig, Germany
| | - Dmitri Lodygin
- Institute of Neuroimmunology, Institute for Multiple Sclerosis Research, University Medical Centre Göttingen, 37073 Göttingen, Germany
| | - Francesca Odoardi
- Institute of Neuroimmunology, Institute for Multiple Sclerosis Research, University Medical Centre Göttingen, 37073 Göttingen, Germany
| | - Alexander Flügel
- Institute of Neuroimmunology, Institute for Multiple Sclerosis Research, University Medical Centre Göttingen, 37073 Göttingen, Germany.,Max-Planck-Institute for Experimental Medicine, 37075 Göttingen, Germany
| |
Collapse
|
13
|
Flügel A, Schläger C, Lühder F, Odoardi F. Autoimmune disease in the brain--how to spot the culprits and how to keep them in check. J Neurol Sci 2014; 311 Suppl 1:S3-11. [PMID: 22206764 DOI: 10.1016/s0022-510x(11)70002-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Current concepts attribute an early and central role for auto-aggressive, myelin-specific T-lymphocytes in the pathogenesis of multiple sclerosis. This view emerged from immunological and pathological findings in experimental autoimmune encephalitis, an animal model characterised by pathological lesions closely resembling the ones found in multiple sclerosis. Furthermore, therapeutic strategies targeting the functions of these encephalitogenic T cells which attenuate their pathogenicity such as glatiramer acetate or anti-VLA4 antibody treatments represent proven approaches in multiple sclerosis. Nonetheless, all therapies evaluated to date either insufficiently dampen down inflammation or completely block immune processes. For this reason, there is a need to identify new therapeutic targets. We have employed live intravital two-photon microscopy to learn more about the behaviour of T cells during the preclinical phase of EAE, when T cells acquire the properties required to invade their target organ. Furthermore, we were able to identify an unexpected locomotive behaviour of T cells at the blood-brain barrier, which occurs immediately before diapedesis and the induction of paralytic disease. Such studies might open new avenues for the treatment of CNS autoimmune diseases. Multiple sclerosis is considered to be an autoimmune disease in which self-reactive T cells enter the central nervous system (CNS) and create an inflammatory milieu that destroys myelin and neurons. Immunomodulatory strategies for the treatment of multiple sclerosis target this process by attempting to inactivate these auto-aggressive T cells. However, so far, these strategies have failed to extinguish disease activity completely. For this reason, there is a need to understand in more detail the mechanisms by which T cells become encephalitogenic, how they enter the nervous system, and what the signals are that guide them along this path. If these processes could be better understood, it may be possible to design more effective and specific therapies for multiple sclerosis. This article will give a brief overview about our recent findings obtained using intravital imaging of autoaggressive effector T cells in an experimental model of multiple sclerosis. This new technological approach might help to fill some gaps in the understanding of autoimmune pathogenesis of multiple sclerosis.
Collapse
Affiliation(s)
- Alexander Flügel
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Centre Göttingen, Göttingen, Germany
| | | | | | | |
Collapse
|
14
|
Jacobelli J, Lindsay RS, Friedman RS. Peripheral tolerance and autoimmunity: lessons from in vivo imaging. Immunol Res 2013; 55:146-54. [PMID: 22956468 DOI: 10.1007/s12026-012-8358-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Multi-photon microscopy has taken hold as a widely used technique in immunology, allowing for imaging of the kinetics of immune cell motility and cell-cell interactions, but what have we learned from this technique about the processes involved in peripheral tolerance and autoimmunity? Various studies have now looked at the dynamics of several mechanisms of peripheral T cell tolerance and efforts to examine the dynamics of the autoimmune response at the disease site are also under way. Here, we will discuss the findings of studies that use multi-photon microscopy to examine the dynamics of T cell tolerance in the lymph nodes and of the autoimmune processes involved in models of type 1 diabetes and multiple sclerosis. An emerging theme from these studies is that short T cell-antigen presenting cell interactions can lead to tolerance, and that autoreactive T cell restimulation at the disease site can play an important role in autoimmune disease exacerbation.
Collapse
Affiliation(s)
- Jordan Jacobelli
- Integrated Department of Immunology, National Jewish Health and University of Colorado School of Medicine, Denver, CO 80206, USA.
| | | | | |
Collapse
|
15
|
Dynamic imaging reveals promiscuous crosspresentation of blood-borne antigens to naive CD8+ T cells in the bone marrow. Blood 2013; 122:193-208. [PMID: 23637125 DOI: 10.1182/blood-2012-01-401265] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The bone marrow (BM) hosts memory lymphocytes and supports secondary immune responses against blood-borne antigens, but it is unsettled whether primary responses occur there and which cells present the antigen. We used 2-photon microscopy in the BM of live mice to study these questions. Naïve CD8(+) T cells crawled rapidly at steady state but arrested immediately upon sensing antigenic peptides. Following infusion of soluble protein, various cell types were imaged ingesting the antigen, while antigen-specific T cells decelerated, clustered, upregulated CD69, and were observed dividing in situ to yield effector cells. Unlike in the spleen, T-cell responses persisted when BM-resident dendritic cells (DCs) were ablated but failed when all phagocytic cells were depleted. Potential antigen-presenting cells included monocytes and macrophages but not B cells. Collectively, our results suggest that the BM supports crosspresentation of blood-borne antigens similar to the spleen; uniquely, alongside DCs, other myeloid cells participate in crosspresentation.
Collapse
|
16
|
Lodygin D, Odoardi F, Schläger C, Körner H, Kitz A, Nosov M, van den Brandt J, Reichardt HM, Haberl M, Flügel A. A combination of fluorescent NFAT and H2B sensors uncovers dynamics of T cell activation in real time during CNS autoimmunity. Nat Med 2013; 19:784-90. [PMID: 23624600 DOI: 10.1038/nm.3182] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 10/16/2012] [Indexed: 01/21/2023]
Abstract
Multiple sclerosis is an autoimmune disease of the central nervous system (CNS) that is initiated when self-reactive T cells enter the brain and become locally activated after encountering their specific nervous antigens. When and where the disease-relevant antigen encounters occur is unclear. Here we combined fluorescently labeled nuclear factor of activated T cells (NFAT) with histone protein H2B to create a broadly applicable molecular sensor for intravital imaging of T cell activation. In experimental autoimmune encephalomyelitis, an animal model for multiple sclerosis, we report that effector T cells entering the CNS become activated after short contacts with leptomeningeal phagocytes. During established disease, the activation process is extended to the depth of the CNS parenchyma, where the cells form contacts with microglia and recruited phagocytes. We show that it is the activation processes during the preclinical phase rather than during established disease that are essential for the intensity and duration of the disease bout.
Collapse
Affiliation(s)
- Dmitri Lodygin
- Institute for Multiple Sclerosis Research and Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Center Göttingen, Göttingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Pesic M, Bartholomäus I, Kyratsous NI, Heissmeyer V, Wekerle H, Kawakami N. 2-photon imaging of phagocyte-mediated T cell activation in the CNS. J Clin Invest 2013; 123:1192-201. [PMID: 23454769 DOI: 10.1172/jci67233] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/06/2012] [Indexed: 12/31/2022] Open
Abstract
Autoreactive T cells can infiltrate the CNS to cause disorders such as multiple sclerosis. In order to visualize T cell activation in the CNS, we introduced a truncated fluorescent derivative of nuclear factor of activated T cells (NFAT) as a real-time T cell activation indicator. In experimental autoimmune encephalomyelitis, a rat model of multiple sclerosis, we tracked T cells interacting with structures of the vascular blood-brain barrier (BBB). 2-photon imaging documented the cytoplasmic-nuclear translocation of fluorescent NFAT, indicative of calcium-dependent activation of the T cells in the perivascular space, but not within the vascular lumen. The activation was related to contacts with the local antigen-presenting phagocytes and was noted only in T cells with a high pathogenic potential. T cell activation implied the presentation of an autoantigen, as the weakly pathogenic T cells, which remained silent in the untreated hosts, were activated upon instillation of exogenous autoantigen. Activation did not cogently signal long-lasting arrest, as individual T cells were able to sequentially contact fresh APCs. We propose that the presentation of local autoantigen by BBB-associated APCs provides stimuli that guide autoimmune T cells to the CNS destination, enabling them to attack the target tissue.
Collapse
Affiliation(s)
- Marija Pesic
- Max Planck Institute of Neurobiology, Department of Neuroimmunology, Martinsried, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
The migration of leukocytes from the bloodstream into the central nervous system (CNS) is a key event in the pathogenesis of inflammatory neurological diseases and typically involves the movement of cells through the endothelium of post-capillary venules, which contains intercellular tight junctions. Leukocyte trafficking has predominantly been studied in animal models of multiple sclerosis, stroke and infection. However, recent evidence suggests that immune cells and inflammation mechanisms play an unexpected role in other neurological diseases, such as epilepsy and Parkinson's disease. Imaging leukocyte trafficking in the CNS can be achieved by epifluorescence intravital microscopy (IVM) and multiphoton microscopy. Epifluorescence IVM is ideal for the investigation of leukocyte-endothelial interactions, particularly tethering and rolling, signal transduction pathways controlling integrin activation, slow rolling, arrest and adhesion strengthening in CNS vessels. Multiphoton microscopy is more suitable for the investigation of intraluminal crawling, transmigration and motility inside CNS parenchyma. The mechanisms of leukocyte trafficking in the CNS are not well understood but the use of in vivo imaging techniques to unravel the underlying regulatory pathways will provide insight into the mechanisms of brain damage and may contribute to the development of novel therapeutic strategies. In this review, we discuss recent work in this field, highlighting the development and use of in vivo imaging to investigate leukocyte recruitment in the CNS.
Collapse
|
19
|
Mueller SN. Effector T-cell responses in non-lymphoid tissues: insights from in vivo imaging. Immunol Cell Biol 2013; 91:290-6. [PMID: 23295362 DOI: 10.1038/icb.2012.75] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
T-cell responses are initiated within secondary lymphoid organs, and effector T-cells are released into the circulation where they home to inflamed tissues and mediate protective immune responses. Within non-lymphoid tissues, the types of cellular interactions and the dynamics that lead to clearance of infections are still relatively poorly understood. Here I review how imaging of effector T-cells within tissues has contributed to our understanding of immune responses, and examine some of the remaining questions that may benefit from in vivo imaging to reveal the intricacies of how immune cells function. A detailed understanding of the dynamics of T-cell responses within non-lymphoid tissues is important for the rational design of targeted therapies that influence key steps in disease progression.
Collapse
Affiliation(s)
- Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
20
|
Plasmacytoid Dendritic Cells Play a Key Role in Promoting Atherosclerosis in Apolipoprotein E–Deficient Mice. Arterioscler Thromb Vasc Biol 2012; 32:2569-79. [DOI: 10.1161/atvbaha.112.251314] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Objective—
Clinical studies have identified that reduced numbers of circulating plasmacytoid dendritic cells (pDCs) act as a predictor of cardiovascular events in coronary artery disease and that pDCs are detectable in the shoulder region of human atherosclerotic plaques, where rupture is most likely to occur. Results from animal models are controversial, with pDCs seen to inhibit or promote lesion development depending on the experimental settings. Here, we investigated the role of pDCs in atherosclerosis in apolipoprotein E−deficient mice.
Methods and Results—
We demonstrated that the aorta and spleen of both apolipoprotein E−deficient and C57BL/6 mice displayed similar numbers of pDCs, with similar activation status. In contrast, assessment of antigen uptake/presentation using the Eα/Y-Ae system revealed that aortic pDCs in apolipoprotein E−deficient
-
mice were capable of presenting in vivo systemically administered antigen. Continuous treatment of apolipoprotein E−deficient mice with anti−mouse plasmacytoid dendritic cell antigen 1 (mPDCA-1) antibody caused specific depletion of pDCs in the aorta and spleen and significantly reduced atherosclerosis formation in the aortic sinus (by 46%;
P
<0.001). Depletion of pDCs also reduced macrophages (by 34%;
P
<0.05) and increased collagen content (by 41%;
P
<0.05) in aortic plaques, implying a more stable plaque phenotype. Additionally, pDC depletion reduced splenic T-cell activation and inhibited interleukin-12, chemokine (C-X-C motif) ligand 1, monokine induced by interferon-γ, interferon γ−induced protein 10, and vascular endothelium growth factor serum levels.
Conclusion—
These results identify a critical role for pDCs in atherosclerosis and suggest a potential role for pDC targeting in the control of the pathology.
Collapse
|
21
|
Kawakami N, Bartholomäus I, Pesic M, Mues M. An autoimmunity odyssey: how autoreactive T cells infiltrate into the CNS. Immunol Rev 2012; 248:140-55. [PMID: 22725959 DOI: 10.1111/j.1600-065x.2012.01133.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a widely used animal model of multiple sclerosis (MS), a human autoimmune disease. To explore how EAE and ultimately MS is induced, autoantigen-specific T cells were established, were labeled with fluorescent protein by retroviral gene transfer, and were tracked in vivo after adoptive transfer. Intravital imaging with two-photon microscopy was used to record the entire entry process of autoreactive T cells into the CNS: a small number of T cells first appear in the CNS leptomeninges before onset of EAE, and crawl on the intraluminal surface of blood vessels, which is integrin α4 and αL dependent. After extravasation, the T cells continue into the perivascular space, meeting local antigen-presenting cells (APCs), which present endogenous antigens. This interaction activates the T cells and guides them to penetrate the CNS parenchyma. As the local APCs in the CNS are not saturated with endogenous antigens, exogenous antigens stimulate the autoreactive T cells more strongly and, as a result, exacerbate the clinical outcome. Currently, we are attempting to visualize T-cell activation in vivo in both rat T-cell-mediated EAE and mouse spontaneous EAE models.
Collapse
Affiliation(s)
- Naoto Kawakami
- Institute of Clinical Neuroimmunology, Ludwig Maximilians University, München, Germany.
| | | | | | | |
Collapse
|
22
|
T cells become licensed in the lung to enter the central nervous system. Nature 2012; 488:675-9. [PMID: 22914092 DOI: 10.1038/nature11337] [Citation(s) in RCA: 312] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Accepted: 06/19/2012] [Indexed: 02/07/2023]
Abstract
The blood–brain barrier (BBB) and the environment of the central nervous system (CNS) guard the nervous tissue from peripheral immune cells. In the autoimmune disease multiple sclerosis, myelin-reactive T-cell blasts are thought to transgress the BBB and create a pro-inflammatory environment in the CNS, thereby making possible a second autoimmune attack that starts from the leptomeningeal vessels and progresses into the parenchyma. Using a Lewis rat model of experimental autoimmune encephalomyelitis, we show here that contrary to the expectations of this concept, T-cell blasts do not efficiently enter the CNS and are not required to prepare the BBB for immune-cell recruitment. Instead, intravenously transferred T-cell blasts gain the capacity to enter the CNS after residing transiently within the lung tissues. Inside the lung tissues, they move along and within the airways to bronchus-associated lymphoid tissues and lung-draining mediastinal lymph nodes before they enter the blood circulation from where they reach the CNS. Effector T cells transferred directly into the airways showed a similar migratory pattern and retained their full pathogenicity. On their way the T cells fundamentally reprogrammed their gene-expression profile, characterized by downregulation of their activation program and upregulation of cellular locomotion molecules together with chemokine and adhesion receptors. The adhesion receptors include ninjurin 1, which participates in T-cell intravascular crawling on cerebral blood vessels. We detected that the lung constitutes a niche not only for activated T cells but also for resting myelin-reactive memory T cells. After local stimulation in the lung, these cells strongly proliferate and, after assuming migratory properties, enter the CNS and induce paralytic disease. The lung could therefore contribute to the activation of potentially autoaggressive T cells and their transition to a migratory mode as a prerequisite to entering their target tissues and inducing autoimmune disease.
Collapse
|
23
|
High-resolution, noninvasive longitudinal live imaging of immune responses. Proc Natl Acad Sci U S A 2011; 108:12863-8. [PMID: 21768391 DOI: 10.1073/pnas.1105002108] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Intravital imaging emerged as an indispensible tool in biological research, and a variety of imaging techniques have been developed to noninvasively monitor tissues in vivo. However, most of the current techniques lack the resolution to study events at the single-cell level. Although intravital multiphoton microscopy has addressed this limitation, the need for repeated noninvasive access to the same tissue in longitudinal in vivo studies remains largely unmet. We now report on a previously unexplored approach to study immune responses after transplantation of pancreatic islets into the anterior chamber of the mouse eye. This approach enabled (i) longitudinal, noninvasive imaging of transplanted tissues in vivo; (ii) in vivo cytolabeling to assess cellular phenotype and viability in situ; (iii) local intervention by topical application or intraocular injection; and (iv) real-time tracking of infiltrating immune cells in the target tissue.
Collapse
|
24
|
Hugues S. Dynamics of dendritic cell-T cell interactions: a role in T cell outcome. Semin Immunopathol 2010; 32:227-38. [PMID: 20607241 DOI: 10.1007/s00281-010-0211-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 05/28/2010] [Indexed: 01/19/2023]
Abstract
Antigen-specific dendritic cells (DC)-T cell encounters occur in lymph nodes (LNs) and are essential for the induction of both priming and tolerance. In both cases, T cells are rapidly activated and proliferate. However, the subsequent outcome of T cell activation depends on the modulation of different DC- and T cell-intrinsic signals. Recent advances in two-photon (2P) microscopy have furthered our understanding regarding the complex choreography of DCs and T cells in intact LNs, and established differences in the dynamics of DC-T cell contacts during priming and tolerance induction. The mechanisms that favour DC-T cell encounters, as well as the contribution of the frequency and the duration of such encounters in dictating the T cell response, are discussed in this review.
Collapse
Affiliation(s)
- Stéphanie Hugues
- Department of Pathology, University of Geneva Medical School, Geneva, Switzerland.
| |
Collapse
|
25
|
Cordiglieri C, Odoardi F, Zhang B, Nebel M, Kawakami N, Klinkert WEF, Lodygin D, Lühder F, Breunig E, Schild D, Ulaganathan VK, Dornmair K, Dammermann W, Potter BVL, Guse AH, Flügel A. Nicotinic acid adenine dinucleotide phosphate-mediated calcium signalling in effector T cells regulates autoimmunity of the central nervous system. ACTA ACUST UNITED AC 2010; 133:1930-43. [PMID: 20519328 PMCID: PMC2892943 DOI: 10.1093/brain/awq135] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nicotinic acid adenine dinucleotide phosphate represents a newly identified second messenger in T cells involved in antigen receptor-mediated calcium signalling. Its function in vivo is, however, unknown due to the lack of biocompatible inhibitors. Using a recently developed inhibitor, we explored the role of nicotinic acid adenine dinucleotide phosphate in autoreactive effector T cells during experimental autoimmune encephalomyelitis, the animal model for multiple sclerosis. We provide in vitro and in vivo evidence that calcium signalling controlled by nicotinic acid adenine dinucleotide phosphate is relevant for the pathogenic potential of autoimmune effector T cells. Live two photon imaging and molecular analyses revealed that nicotinic acid adenine dinucleotide phosphate signalling regulates T cell motility and re-activation upon arrival in the nervous tissues. Treatment with the nicotinic acid adenine dinucleotide phosphate inhibitor significantly reduced both the number of stable arrests of effector T cells and their invasive capacity. The levels of pro-inflammatory cytokines interferon-gamma and interleukin-17 were strongly diminished. Consecutively, the clinical symptoms of experimental autoimmune encephalomyelitis were ameliorated. In vitro, antigen-triggered T cell proliferation and cytokine production were evenly suppressed. These inhibitory effects were reversible: after wash-out of the nicotinic acid adenine dinucleotide phosphate antagonist, the effector T cells fully regained their functions. The nicotinic acid derivative BZ194 induced this transient state of non-responsiveness specifically in post-activated effector T cells. Naïve and long-lived memory T cells, which express lower levels of the putative nicotinic acid adenine dinucleotide phosphate receptor, type 1 ryanodine receptor, were not targeted. T cell priming and recall responses in vivo were not reduced. These data indicate that the nicotinic acid adenine dinucleotide phosphate/calcium signalling pathway is essential for the recruitment and the activation of autoaggressive effector T cells within their target organ. Interference with this signalling pathway suppresses the formation of autoimmune inflammatory lesions and thus might qualify as a novel strategy for the treatment of T cell mediated autoimmune diseases.
Collapse
Affiliation(s)
- Chiara Cordiglieri
- Department of Neuroimmunology, Max-Planck-Institute for Neurobiology, Martinsried, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Filipe-Santos O, Pescher P, Breart B, Lippuner C, Aebischer T, Glaichenhaus N, Späth GF, Bousso P. A dynamic map of antigen recognition by CD4 T cells at the site of Leishmania major infection. Cell Host Microbe 2009; 6:23-33. [PMID: 19616763 DOI: 10.1016/j.chom.2009.04.014] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 03/13/2009] [Accepted: 05/01/2009] [Indexed: 11/16/2022]
Abstract
CD4 T helper cells play a central role in the control of infection by intracellular parasites. How efficiently pathogen-specific CD4 T cells detect infected cells in vivo is unclear. Here, we employed intravital two-photon imaging to examine the behavior of pathogen-specific CD4 T cells at the site of Leishmania major infection. While activated CD4 T cells enter the inflamed tissue irrespective of their antigen specificity, pathogen-specific T cells preferentially decelerated and accumulated in infected regions of the dermis. Antigen recognition by CD4 T cells was heterogeneous, involving both stable and dynamic contacts with infected phagocytes. However, not all infected cells induced arrest or deceleration of pathogen-specific T cells, and dense clusters of infected cells were poorly accessible to migrating T cells. Thus, disparities in the dynamics of T cell contacts with infected cells and local variation in T cell access to infected cells are important elements of the host-pathogen interplay.
Collapse
Affiliation(s)
- Orchidée Filipe-Santos
- Institut Pasteur, G5 Dynamiques des Réponses Immunes, Inserm U668, Equipe Avenir, Paris F-75724, France
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Bartholomäus I, Kawakami N, Odoardi F, Schläger C, Miljkovic D, Ellwart JW, Klinkert WEF, Flügel-Koch C, Issekutz TB, Wekerle H, Flügel A. Effector T cell interactions with meningeal vascular structures in nascent autoimmune CNS lesions. Nature 2009; 462:94-8. [PMID: 19829296 DOI: 10.1038/nature08478] [Citation(s) in RCA: 507] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 09/01/2009] [Indexed: 01/18/2023]
Abstract
The tissues of the central nervous system are effectively shielded from the blood circulation by specialized vessels that are impermeable not only to cells, but also to most macromolecules circulating in the blood. Despite this seemingly absolute seclusion, central nervous system tissues are subject to immune surveillance and are vulnerable to autoimmune attacks. Using intravital two-photon imaging in a Lewis rat model of experimental autoimmune encephalomyelitis, here we present in real-time the interactive processes between effector T cells and cerebral structures from their first arrival to manifest autoimmune disease. We observed that incoming effector T cells successively scanned three planes. The T cells got arrested to leptomeningeal vessels and immediately monitored the luminal surface, crawling preferentially against the blood flow. After diapedesis, the cells continued their scan on the abluminal vascular surface and the underlying leptomeningeal (pial) membrane. There, the T cells encountered phagocytes that effectively present antigens, foreign as well as myelin proteins. These contacts stimulated the effector T cells to produce pro-inflammatory mediators, and provided a trigger to tissue invasion and the formation of inflammatory infiltrations.
Collapse
|
28
|
Konjufca V, Miller MJ. Two-photon microscopy of host-pathogen interactions: acquiring a dynamic picture of infection in vivo. Cell Microbiol 2009; 11:551-9. [PMID: 19170686 DOI: 10.1111/j.1462-5822.2009.01289.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Two-photon (2P) microscopy has become increasingly popular among immunologists for analysing single-cell dynamics in tissues. Researchers are now taking 2P microscopy beyond the study of model antigen systems (e.g. ovalbumin immunization) and are applying the technique to examine infection in vivo. With the appropriate fluorescent probes, 2P imaging can provide high-resolution spatio-temporal information regarding cell behaviour, monitor cell functions and assess various outcomes of infection, such as host cell apoptosis or pathogen proliferation. Imaging of transgenic and knockout mice can be used to probe molecular mechanisms governing the host response to infection. From the microbe side, imaging genetically engineered mutant strains of a pathogen can test the roles of specific virulence factors in pathogenesis. Here, we discuss recent work that has applied 2P microscopy to study models of infection and highlight the tremendous potential that this approach has for investigating host-pathogen interactions.
Collapse
Affiliation(s)
- Vjollca Konjufca
- Washington University School of Medicine, Department of Pathology and Immunology, St. Louis, MO 63110-1093, USA
| | | |
Collapse
|
29
|
T-cell activation by dendritic cells in the lymph node: lessons from the movies. Nat Rev Immunol 2009; 8:675-84. [PMID: 19172690 DOI: 10.1038/nri2379] [Citation(s) in RCA: 246] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Interactions between T cells and dendritic cells (DCs) in the lymph nodes are crucial for initiating cell-mediated adaptive immune responses. With the help of two-photon imaging, the complexity of these cellular contacts in vivo has recently been captured in time-lapse movies in several immunological contexts. Well beyond the satisfaction of seeing a T-cell response as it happens, these experiments provide fundamental insights into the regulation and the biological meaning of T-cell-DC contact dynamics. This Review focuses on how this emerging field is changing our perception of T-cell activation by DCs.
Collapse
|
30
|
Kim HJ, Jung CG, Jensen MA, Dukala D, Soliven B. Targeting of myelin protein zero in a spontaneous autoimmune polyneuropathy. THE JOURNAL OF IMMUNOLOGY 2009; 181:8753-60. [PMID: 19050296 DOI: 10.4049/jimmunol.181.12.8753] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Elimination of the costimulatory molecule B7-2 prevents autoimmune diabetes in NOD mice, but leads to the development of a spontaneous autoimmune polyneuropathy (SAP), which resembles the human disease chronic inflammatory demyelinating polyneuropathy (CIDP). In this study, we examined the immunopathogenic mechanisms in this model, including identification of SAP Ags. We found that B7-2-deficient NOD mice exhibit changes in cytokine and chemokine gene expression in spleens over time. There was an increase in IL-17 and a decrease in IL-10 transcript levels at 4 mo (preclinical phase), whereas IFN-gamma expression peaked at 8 mo (clinical phase). There was also an increase in transcript levels of Th1 cytokines, CXCL10, and RANTES in sciatic nerves of mice that developed SAP. Splenocytes from SAP mice exhibited proliferative and Th1 cytokine responses to myelin P0 (180-199), but not to other P0 peptides or P2 (53-78). Adoptive transfer of P0-reactive T cells generated from SAP mice induced neuropathy in four of six NOD.SCID mice. Data from i.v. tolerance studies indicate that myelin P0 is one of the autoantigens targeted by T cells in SAP in this model. The expression of P0 by peri-islet Schwann cells provides a potential mechanism linking islet autoimmunity and inflammatory neuropathy.
Collapse
Affiliation(s)
- Hye-Jung Kim
- Department of Neurology, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
31
|
Wilson EH, Harris TH, Mrass P, John B, Tait ED, Wu GF, Pepper M, Wherry EJ, Dzierzinski F, Roos D, Haydon PG, Laufer TM, Weninger W, Hunter CA. Behavior of parasite-specific effector CD8+ T cells in the brain and visualization of a kinesis-associated system of reticular fibers. Immunity 2009; 30:300-11. [PMID: 19167248 DOI: 10.1016/j.immuni.2008.12.013] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 11/19/2008] [Accepted: 12/11/2008] [Indexed: 12/23/2022]
Abstract
To understand lymphocyte behavior in the brain, we used two-photon microscopy to visualize effector CD8(+) T cells during toxoplasmic encephalitis. These cells displayed multiple behaviors with two distinct populations of cells apparent: one with a constrained pattern of migration and one with a highly migratory subset. The proportion of these populations varied over time associated with changes in antigen availability as well as T cell expression of the inhibitory receptor PD1. Unexpectedly, the movement of infiltrating cells was closely associated with an infection-induced reticular system of fibers. This observation suggests that, whereas in other tissues pre-existing scaffolds exist that guide lymphocyte migration, in the brain specialized structures are induced by inflammation that guide migration of T cells in this immune-privileged environment.
Collapse
Affiliation(s)
- Emma H Wilson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Listeria monocytogenes infection in mice is a highly prolific model of bacterial infection. Several in vivo imaging approaches have been used to study host cell dynamics in response to infection, including bioluminescence imaging, confocal microscopy and two-photon microscopy, The application of in vivo imaging to study transgenic mouse models is providing unprecedented opportunities to test specific molecular mechanistic theories about how the host immune response unfolds. In complementary studies, in vivo imaging can be performed using genetically engineered bacterial mutants to assess the impact of specific virulence factors in host cell invasion and pathogenesis. The purpose of this chapter is to provide a general rationale for why in vivo imaging is important, provide an overview of various techniques highlighting the strengths and weaknesses of each, and provide examples of how various imaging techniques have been used to study Listeria infection. Lastly, our goal is to make the reader aware of the tremendous potential these approaches hold for studying host-pathogen interactions.
Collapse
|
33
|
Aoshi T, Zinselmeyer BH, Konjufca V, Lynch JN, Zhang X, Koide Y, Miller MJ. Bacterial entry to the splenic white pulp initiates antigen presentation to CD8+ T cells. Immunity 2008; 29:476-86. [PMID: 18760639 DOI: 10.1016/j.immuni.2008.06.013] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Revised: 05/20/2008] [Accepted: 06/26/2008] [Indexed: 11/21/2022]
Abstract
The spleen plays an important role in host-protective responses to bacteria. However, the cellular dynamics that lead to pathogen-specific immunity remain poorly understood. Here we examined Listeria monocytogenes (Lm) infection in the mouse spleen via in situ fluorescence microscopy. We found that the redistribution of Lm from the marginal zone (MZ) to the periarteriolar lymphoid sheath (PALS) was inhibited by pertussis toxin and required the presence of CD11c(+) cells. As early as 9 hr after infection, we detected infected dendritic cells in the peripheral regions of the PALS and clustering of Lm-specific T cells by two-photon microscopy. Pertussis toxin inhibited both Lm entry into the PALS and antigen presentation to CD8(+) T cells. Our study suggests that splenic dendritic cells rapidly deliver intracellular bacteria to the T cell areas of the white pulp to initiate CD8(+) T cell responses.
Collapse
Affiliation(s)
- Taiki Aoshi
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Wegmann KW, Wagner CR, Whitham RH, Hinrichs DJ. Synthetic Peptide Dendrimers Block the Development and Expression of Experimental Allergic Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2008; 181:3301-9. [DOI: 10.4049/jimmunol.181.5.3301] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
35
|
Cahalan MD, Parker I. Choreography of cell motility and interaction dynamics imaged by two-photon microscopy in lymphoid organs. Annu Rev Immunol 2008; 26:585-626. [PMID: 18173372 DOI: 10.1146/annurev.immunol.24.021605.090620] [Citation(s) in RCA: 237] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The immune system is the most diffuse cellular system in the body. Accordingly, long-range migration of cells and short-range communication by local chemical signaling and by cell-cell contacts are vital to the control of an immune response. Cellular homing and migration within lymphoid organs, antigen recognition, and cell signaling and activation are clearly vital during an immune response, but these events had not been directly observed in vivo until recently. Introduced to the field of immunology in 2002, two-photon microscopy is the method of choice for visualizing living cells deep within native tissue environments, and it is now revealing an elegant cellular choreography that underlies the adaptive immune response to antigen challenge. We review cellular dynamics and molecular factors that contribute to basal motility of lymphocytes in the lymph node and cellular interactions leading to antigen capture and recognition, T cell activation, B cell activation, cytolytic effector function, and antibody production.
Collapse
Affiliation(s)
- Michael D Cahalan
- Department of Physiology and Biophysics, University of California, Irvine, California 92697, USA.
| | | |
Collapse
|
36
|
Müller N, van den Brandt J, Odoardi F, Tischner D, Herath J, Flügel A, Reichardt HM. A CD28 superagonistic antibody elicits 2 functionally distinct waves of T cell activation in rats. J Clin Invest 2008; 118:1405-16. [PMID: 18357346 DOI: 10.1172/jci32698] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Accepted: 01/30/2008] [Indexed: 11/17/2022] Open
Abstract
Administration of the CD28 superagonistic antibody JJ316 is an efficient means to treat autoimmune diseases in rats, but the humanized antibody TGN1412 caused devastating side effects in healthy volunteers during a clinical trial. Here we show that JJ316 treatment of rats induced a dramatic redistribution of T lymphocytes from the periphery to the secondary lymphoid organs, resulting in severe T lymphopenia. Live imaging of secondary lymphoid organs revealed that JJ316 administration almost instantaneously (<2 minutes) arrested T cells in situ. This reduction in T cell motility was accompanied by profound cytoskeletal rearrangements and increased cell size. In addition, surface expression of lymphocyte function-associated antigen-1 was enhanced, endothelial differentiation sphingolipid G protein-coupled receptor 1 and L selectin levels were downregulated, and the cells lost their responsiveness to sphingosine 1-phosphate-directed migration. These proadhesive alterations were accompanied by signs of strong activation, including upregulation of CD25, CD69, CD134, and proinflammatory mediators. However, this did not lead to a cytokine storm similar to the clinical trial. While most of the early changes disappeared within 48 hours, we observed that CD4+CD25+FoxP3+ regulatory T cells experienced a second phase of activation, which resulted in massive cell enlargement, extensive polarization, and increased motility. These data suggest that CD28 superagonists elicit 2 qualitatively distinct waves of activation.
Collapse
Affiliation(s)
- Nora Müller
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
37
|
Ng LG, Mrass P, Kinjyo I, Reiner SL, Weninger W. Two-photon imaging of effector T-cell behavior: lessons from a tumor model. Immunol Rev 2008; 221:147-62. [PMID: 18275480 DOI: 10.1111/j.1600-065x.2008.00596.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Recent advances in two-photon microscopy have provided a new way of visualizing the behavior of fluorescently tagged cells within their natural microenvironment. This technology has allowed for generating a detailed picture of the cellular interaction dynamics operant in the activation of T cells and B cells during primary immune responses within secondary lymphoid organs. In contrast, relatively little is known about the migratory and interactive behavior of effector T cells within peripheral organs. We have recently developed a two-photon microscopy model that enables tracking of cytotoxic T cells within tumors. We have demonstrated that tumor-infiltrating T lymphocytes (TILs) follow random migratory paths and that their migratory properties depend on signals from the T-cell receptor. We further showed that TILs underwent short- and long-term interactions with tumor cells as well as macrophages. Recently, we succeeded in dynamic imaging of the distribution of fluorescently tagged molecules within TILs at subcellular resolution, which will be instrumental for defining the composition of the lytic synapse as well as the targeted release of cytotoxic granules by these cells. The purpose of this review is to put our findings into the context of the current literature and to point out the molecular cues mediating effector T-cell function as candidates for future investigation.
Collapse
|
38
|
|
39
|
Blood-borne soluble protein antigen intensifies T cell activation in autoimmune CNS lesions and exacerbates clinical disease. Proc Natl Acad Sci U S A 2007; 104:18625-30. [PMID: 18000062 DOI: 10.1073/pnas.0705033104] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We explored the effect of i.v. soluble antigen on autoaggressive, myelin basic protein-specific effector T cells within their target organ during acute experimental autoimmune encephalomyelitis (EAE). Intravital two-photon imaging revealed that i.v. autoantigen reached the CNS and was taken up and processed by antigen-presenting cells within 30 min after injection. The exogenous autoantigen dramatically changed the motility and function of autoreactive effector T cells within the EAE lesions: T cells that had been cruising through the tissue slowed down and became tethered to local antigen-presenting cells within 1 h. One hour later, the effector T cells massively produced proinflammatory cytokines and up-regulated membranous activation markers. This strong activation of the T cells boosted CNS inflammation and aggravated clinical disease. Postactivated effector and resting memory T cells specific for a non-CNS antigen (ovalbumin) were recruited to EAE lesions and moved there without contacting antigen-presenting cells. These cells were similarly arrested and activated after i.v. infusion of ovalbumin, and they also exacerbated clinical disease. Our data are relevant for autoantigen-based therapies of autoimmune disorders. Further, the study indicates how brain unrelated antigens (microbial components) leaking into the chronically inflamed CNS through the bloodstream might trigger relapses in multiple sclerosis.
Collapse
|
40
|
Flügel A, Odoardi F, Nosov M, Kawakami N. Autoaggressive effector T cells in the course of experimental autoimmune encephalomyelitis visualized in the light of two-photon microscopy. J Neuroimmunol 2007; 191:86-97. [PMID: 17976745 DOI: 10.1016/j.jneuroim.2007.09.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Accepted: 09/12/2007] [Indexed: 10/22/2022]
Abstract
Two photon microscopy (TPM) recently emerged as optical tool for the visualization of immune processes hundreds of micrometers deep in living tissue and organs. Here we summarize recent work on exploiting this technology to study brain antigen specific T cells. These cells are the cause of Experimental Autoimmune Encephalomyelitis (EAE) an autoimmune disease model of Multiple Sclerosis. TPM studies elucidated the dynamics of the autoaggressive effector T cells in peripheral immune milieus during preclinical EAE, where the cells become reprogrammed to enter their target organ. These studies revealed an unexpectedly lively locomotion behavior of the cells interrupted only by short-lasting contacts with the local immune stroma. Live T cell behavior was furthermore studied within the acutely inflamed CNS. Two distinct migratory patterns of the T cells were found: the majority of cells (60-70%) moved fast and seemingly unhindered through the compact CNS parenchyma. The motility of the other cell fraction was highly confined. The cells swung around a fixed cell pole forming long-lasting contacts to putative local antigen presenting cells.
Collapse
|