1
|
Tritz ZP, Orozco RC, Malo CS, Ayasoufi K, Fain CE, Khadka RH, Goddery EN, Yokanovich LT, Settell ML, Hansen MJ, Jin F, Pavelko KD, Pease LR, Johnson AJ. Conditional Silencing of H-2D b Class I Molecule Expression Modulates the Protective and Pathogenic Kinetics of Virus-Antigen-Specific CD8 T Cell Responses during Theiler's Virus Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:1228-1238. [PMID: 32737149 DOI: 10.4049/jimmunol.2000340] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
Theiler's murine encephalomyelitis virus (TMEV) infection of the CNS is cleared in C57BL/6 mice by a CD8 T cell response restricted by the MHC class I molecule H-2Db The identity and function of the APC(s) involved in the priming of this T cell response is (are) poorly defined. To address this gap in knowledge, we developed an H-2Db LoxP-transgenic mouse system using otherwise MHC class I-deficient C57BL/6 mice, thereby conditionally ablating MHC class I-restricted Ag presentation in targeted APC subpopulations. We observed that CD11c+ APCs are critical for early priming of CD8 T cells against the immunodominant TMEV peptide VP2121-130 Loss of H-2Db on CD11c+ APCs mitigates the CD8 T cell response, preventing early viral clearance and immunopathology associated with CD8 T cell activity in the CNS. In contrast, animals with H-2Db-deficient LysM+ APCs retained early priming of Db:VP2121-130 epitope-specific CD8 T cells, although a modest reduction in immune cell entry into the CNS was observed. This work establishes a model enabling the critical dissection of H-2Db-restricted Ag presentation to CD8 T cells, revealing cell-specific and temporal features involved in the generation of CD8 T cell responses. Employing this novel system, we establish CD11c+ cells as pivotal to the establishment of acute antiviral CD8 T cell responses against the TMEV immunodominant epitope VP2121-130, with functional implications both for T cell-mediated viral control and immunopathology.
Collapse
Affiliation(s)
- Zachariah P Tritz
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Robin C Orozco
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Courtney S Malo
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | | | - Cori E Fain
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Roman H Khadka
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Emma N Goddery
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Lila T Yokanovich
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Megan L Settell
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905
| | | | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN 55905
| | | | - Larry R Pease
- Mayo Clinic Department of Immunology, Rochester, MN 55905.,Mayo Clinic Department of Biochemistry, Rochester, MN 55905
| | - Aaron J Johnson
- Mayo Clinic Department of Immunology, Rochester, MN 55905; .,Mayo Clinic Department of Molecular Medicine, Rochester, MN 55905; and.,Mayo Clinic Department of Neurology, Rochester, MN 55905
| |
Collapse
|
2
|
Gentile A, De Vito F, Fresegna D, Rizzo FR, Bullitta S, Guadalupi L, Vanni V, Buttari F, Stampanoni Bassi M, Leuti A, Chiurchiù V, Marfia GA, Mandolesi G, Centonze D, Musella A. Peripheral T cells from multiple sclerosis patients trigger synaptotoxic alterations in central neurons. Neuropathol Appl Neurobiol 2019; 46:160-170. [DOI: 10.1111/nan.12569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 05/22/2019] [Indexed: 01/03/2023]
Affiliation(s)
- A. Gentile
- Synaptic Immunopathology Lab Department of Systems Medicine University of Rome Tor Vergata Rome Italy
- Synaptic Immunopathology Lab IRCCS San Raffaele Rome Italy
| | - F. De Vito
- Unit of Neurology IRCCS Neuromed Pozzilli Italy
| | - D. Fresegna
- Synaptic Immunopathology Lab IRCCS San Raffaele Rome Italy
| | - F. R. Rizzo
- Synaptic Immunopathology Lab Department of Systems Medicine University of Rome Tor Vergata Rome Italy
| | - S. Bullitta
- Synaptic Immunopathology Lab Department of Systems Medicine University of Rome Tor Vergata Rome Italy
- Synaptic Immunopathology Lab IRCCS San Raffaele Rome Italy
| | - L. Guadalupi
- Synaptic Immunopathology Lab Department of Systems Medicine University of Rome Tor Vergata Rome Italy
- Synaptic Immunopathology Lab IRCCS San Raffaele Rome Italy
| | - V. Vanni
- Synaptic Immunopathology Lab IRCCS San Raffaele Rome Italy
| | - F. Buttari
- Unit of Neurology IRCCS Neuromed Pozzilli Italy
| | | | - A. Leuti
- Department of Medicine Campus Bio‐Medico University of Rome Rome Italy
- European Center for Brain Research (CERC)/IRCCS Fondazione Santa Lucia Rome Italy
| | - V. Chiurchiù
- Department of Medicine Campus Bio‐Medico University of Rome Rome Italy
- European Center for Brain Research (CERC)/IRCCS Fondazione Santa Lucia Rome Italy
| | - G. A. Marfia
- Multiple Sclerosis Research Unit Department of Systems Medicine Tor Vergata University Rome Italy
| | - G. Mandolesi
- Synaptic Immunopathology Lab IRCCS San Raffaele Pisana and University San Raffaele Rome Italy
| | - D. Centonze
- Synaptic Immunopathology Lab Department of Systems Medicine University of Rome Tor Vergata Rome Italy
- Unit of Neurology IRCCS Neuromed Pozzilli Italy
| | - A. Musella
- Synaptic Immunopathology Lab IRCCS San Raffaele Pisana and University San Raffaele Rome Italy
| |
Collapse
|
3
|
Peitzsch C, Kurth I, Ebert N, Dubrovska A, Baumann M. Cancer stem cells in radiation response: current views and future perspectives in radiation oncology. Int J Radiat Biol 2019; 95:900-911. [PMID: 30897014 DOI: 10.1080/09553002.2019.1589023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Purpose: Despite technological improvement and advances in biology-driven patient stratification, many patients still fail radiotherapy resulting in loco-regional and distant recurrence. Tumor heterogeneity remains a key challenge to effective cancer treatment, and reliable stratification of cancer patients for prediction of outcomes is highly important. Intratumoral heterogeneity is manifested at the different levels, including different tumorigenic properties of cancer cells. Since John Dick et al. isolated leukemia initiating cells in 1990, the populations of tumor initiating or cancer stem cells (CSCs) were identified and characterized also for a broad spectrum of solid tumor types. The properties of CSCs are of considerable clinical relevance: CSCs have self-renewal and tumor initiating potential, and the metastases are initiated by the CSC clones with the ability to disseminate from the primary tumor site. Conclusion: Evidence from both, experimental and clinical studies demonstrates that the probability of achieving local tumor control by radiation therapy depends on the complete eradication of CSC populations. The number, properties and molecular signature of CSCs are highly predictive for clinical outcome of radiotherapy, whereas targeted therapies against CSCs combined with conventional treatment are expected to provide an improved clinical response and prevent tumor relapse. In this review, we discuss the modern methods to study CSCs in radiation biology, the role of CSCs in personalized cancer therapy as well as future directions for CSC research in translational radiooncology.
Collapse
Affiliation(s)
- Claudia Peitzsch
- a OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany.,b National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz-Zentrum Dresden - Rossendorf (HZDR) , Dresden , Germany.,c German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Ina Kurth
- d German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Nadja Ebert
- d German Cancer Research Center (DKFZ) , Heidelberg , Germany.,f Department of Radiotherapy and Radiation Oncology , Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany
| | - Anna Dubrovska
- a OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany.,c German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ) , Heidelberg , Germany.,e Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay , Dresden , Germany
| | - Michael Baumann
- d German Cancer Research Center (DKFZ) , Heidelberg , Germany.,f Department of Radiotherapy and Radiation Oncology , Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany
| |
Collapse
|
4
|
Panja D, Vedeler CA, Schubert M. Paraneoplastic cerebellar degeneration: Yo antibody alters mitochondrial calcium buffering capacity. Neuropathol Appl Neurobiol 2018; 45:141-156. [PMID: 29679372 PMCID: PMC7379599 DOI: 10.1111/nan.12492] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/02/2018] [Indexed: 12/16/2022]
Abstract
Aim Neurodegeneration is associated with dysfunction of calcium buffering capacity and thereby sustained cellular and mitochondrial calcium overload. Paraneoplastic cerebellar degeneration (PCD), characterized by progressive Purkinje neurone degeneration following paraneoplastic Yo antibody internalization and binding to cerebellar degeneration‐related protein CDR2 and CDR2L, has been linked to intracellular calcium homeostasis imbalance due to calbindin D28k malfunction. Therefore, we hypothesized that Yo antibody internalization affects not only calbindin calcium binding capacity, but also calcium‐sensitive mitochondrial‐associated signalling, causing mitochondrial calcium overload and thereby Purkinje neurone death. Methods Immunohistochemically, we evaluated cerebellar organotypic slice cultures of rat brains after inducing PCD through the application of Yo antibody‐positive PCD patient sera or purified antibodies against CDR2 and CDR2L how pharmacologically biased mitochondrial signalling affected PCD pathology. Results We found that Yo antibody internalization into Purkinje neurons caused depletion of Purkinje neurone calbindin‐immunoreactivity, cannabinoid 1 receptor over‐activation and alterations in the actions of the mitochondria permeability transition pore (MPTP), voltage‐dependent anion channels, reactive oxygen species (ROS) and Na+/Ca2+ exchangers (NCX). The pathological mechanisms caused by Yo antibody binding to CDR2 or CDR2L differed between the two targets. Yo‐CDR2 binding did not alter the mitochondrial calcium retention capacity, cyclophilin D‐independent opening of MPTP or activity of NCX. Conclusion These findings suggest that minimizing intracellular calcium overload toxicity either directly with cyclosporin‐A or indirectly with cannabidiol or the ROS scavenger butylated hydroxytoluene promotes mitochondrial calcium homeostasis and may therefore be used as future neuroprotective therapy for PCD patients.
Collapse
Affiliation(s)
- D Panja
- Department of Clinical Medicine (K1), University of Bergen, Bergen, Norway
| | - C A Vedeler
- Department of Clinical Medicine (K1), University of Bergen, Bergen, Norway.,Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - M Schubert
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
5
|
Dombrowski Y, O'Hagan T, Dittmer M, Penalva R, Mayoral SR, Bankhead P, Fleville S, Eleftheriadis G, Zhao C, Naughton M, Hassan R, Moffat J, Falconer J, Boyd A, Hamilton P, Allen IV, Kissenpfennig A, Moynagh PN, Evergren E, Perbal B, Williams AC, Ingram RJ, Chan JR, Franklin RJM, Fitzgerald DC. Regulatory T cells promote myelin regeneration in the central nervous system. Nat Neurosci 2017; 20:674-680. [PMID: 28288125 PMCID: PMC5409501 DOI: 10.1038/nn.4528] [Citation(s) in RCA: 348] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 02/03/2017] [Indexed: 02/08/2023]
Abstract
Regeneration of CNS myelin involves differentiation of oligodendrocytes from oligodendrocyte progenitor cells. In multiple sclerosis, remyelination can fail despite abundant oligodendrocyte progenitor cells, suggesting impairment of oligodendrocyte differentiation. T cells infiltrate the CNS in multiple sclerosis, yet little is known about T cell functions in remyelination. We report that regulatory T cells (Treg) promote oligodendrocyte differentiation and (re)myelination. Treg-deficient mice exhibited substantially impaired remyelination and oligodendrocyte differentiation, which was rescued by adoptive transfer of Treg. In brain slice cultures, Treg accelerated developmental myelination and remyelination, even in the absence of overt inflammation. Treg directly promoted oligodendrocyte progenitor cell differentiation and myelination in vitro. We identified CCN3 as a Treg-derived mediator of oligodendrocyte differentiation and myelination in vitro. These findings reveal a new regenerative function of Treg in the CNS, distinct from immunomodulation. Although the cells were originally named 'Treg' to reflect immunoregulatory roles, this also captures emerging, regenerative Treg functions.
Collapse
Affiliation(s)
- Yvonne Dombrowski
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Thomas O'Hagan
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Marie Dittmer
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Rosana Penalva
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Sonia R Mayoral
- Department of Neurology and Program in Neurosciences, University of California, San Francisco, California, USA
| | - Peter Bankhead
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Samara Fleville
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - George Eleftheriadis
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Clifford Allbutt Building, Cambridge Biomedical Campus, University of Cambridge, UK
| | - Michelle Naughton
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Rachel Hassan
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Jill Moffat
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - John Falconer
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Amanda Boyd
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Peter Hamilton
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Ingrid V Allen
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Adrien Kissenpfennig
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Paul N Moynagh
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK.,Institute of Immunology, Department of Biology, National University of Ireland Maynooth, Ireland
| | - Emma Evergren
- Centre for Cancer Research and Cell Biology, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Bernard Perbal
- Université Côte d'Azur, CNRS, GREDEG, Nice, France.,International CCN Society, Paris, France
| | - Anna C Williams
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Rebecca J Ingram
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Jonah R Chan
- Department of Neurology and Program in Neurosciences, University of California, San Francisco, California, USA
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Clifford Allbutt Building, Cambridge Biomedical Campus, University of Cambridge, UK
| | - Denise C Fitzgerald
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| |
Collapse
|
6
|
Madill M, Fitzgerald D, O'Connell KE, Dev KK, Shen S, FitzGerald U. In vitro and ex vivo models of multiple sclerosis. Drug Discov Today 2016; 21:1504-1511. [PMID: 27265771 DOI: 10.1016/j.drudis.2016.05.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/20/2016] [Accepted: 05/27/2016] [Indexed: 01/25/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disorder of the central nervous system (CNS). Current therapies suppress a misdirected myelin-destructive immune response. To combat the progressive, neurodestructive phase of MS, the therapeutic research focus is currently on compounds that might boost the endogenous potential of the brain to remyelinate axons, thereby achieving lesion repair. Here, we describe the testing of fingolimod on cultures of oligodendrocytes (OLs) and organotypic brain slices. We detail the protocols, pros, and cons of these in vitro and ex vivo approaches, along with the potential benefit of exploiting skin-punch biopsies from patients with MS, before concluding with a summary of future developments.
Collapse
Affiliation(s)
- Martin Madill
- Regenerative Medicine Institute (REMEDI), School of Medicine and School of Natural Sciences, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Denise Fitzgerald
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Kara E O'Connell
- Drug Development, School of Medicine, Trinity College Dublin, Ireland
| | - Kumlesh K Dev
- Drug Development, School of Medicine, Trinity College Dublin, Ireland
| | - Sanbing Shen
- Regenerative Medicine Institute (REMEDI), School of Medicine and School of Natural Sciences, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Una FitzGerald
- Galway Neuroscience Centre, School of Natural Sciences, National University of Ireland, Galway, Ireland.
| |
Collapse
|
7
|
O'Sullivan C, Schubart A, Mir AK, Dev KK. The dual S1PR1/S1PR5 drug BAF312 (Siponimod) attenuates demyelination in organotypic slice cultures. J Neuroinflammation 2016; 13:31. [PMID: 26856814 PMCID: PMC4746808 DOI: 10.1186/s12974-016-0494-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 01/24/2016] [Indexed: 02/07/2023] Open
Abstract
Background BAF312 (Siponimod) is a dual agonist at the sphingosine-1 phosphate receptors, S1PR1 and S1PR5. This drug is currently undergoing clinical trials for the treatment of secondary progressive multiple sclerosis (MS). Here, we investigated the effects of BAF312 on isolated astrocyte and microglia cultures as well as in slice culture models of demyelination. Methods Mouse and human astrocytes were treated with S1PR modulators and changes in the levels of pERK, pAkt, and calcium signalling as well as S1PR1 internalization and cytokine levels was investigated using Western blotting, immunochemistry, ELISA and confocal microscopy. Organotypic slice cultures were prepared from the cerebellum of 10-day-old mice and treated with lysophosphatidylcholine (LPC), psychosine and/or S1PR modulators, and changes in myelination states were measured by fluorescence of myelin basic protein and neurofilament H. Results BAF312 treatment of human and mouse astrocytes activated pERK, pAKT and Ca2+ signalling as well as inducing S1PR1 internalization. Notably, activation of S1PR1 increased pERK and pAKT in mouse astrocytes while both S1PR1 and S1PR3 equally increased pERK and pAKT in human astrocytes, suggesting that the coupling of S1PR1 and S1PR3 to pERK and pAKT differ in mouse and human astrocytes. We also observed that BAF312 moderately attenuated lipopolysaccharide (LPS)- or TNFα/IL17-induced levels of IL6 in both astrocyte and microglia cell cultures. In organotypic slice cultures, BAF312 reduced LPC-induced levels of IL6 and attenuated LPC-mediated demyelination. We have shown previously that the toxic lipid metabolite psychosine induces demyelination in organotypic slice cultures, without altering the levels of cytokines, such as IL6. Importantly, psychosine-induced demyelination was also attenuated by BAF312. Conclusions Overall, this study suggests that BAF312 can modulate glial cell function and attenuate demyelination, highlighting this drug as a further potential therapy in demyelinating disorders, beyond MS.
Collapse
Affiliation(s)
| | - Anna Schubart
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Anis K Mir
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Kumlesh K Dev
- Drug Development, School of Medicine, Trinity College, Dublin, Ireland.
| |
Collapse
|
8
|
Göbel K, Bittner S, Cerina M, Herrmann AM, Wiendl H, Meuth SG. An ex vivo model of an oligodendrocyte-directed T-cell attack in acute brain slices. J Vis Exp 2015. [PMID: 25741800 DOI: 10.3791/52205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Death of oligodendrocytes accompanied by destruction of neurons and axons are typical histopathological findings in cortical and subcortical grey matter lesions in inflammatory demyelinating disorders like multiple sclerosis (MS). In these disorders, mainly CD8+ T-cells of putative specificity for myelin- and oligodendrocyte-related antigens are found, so that neuronal apoptosis in grey matter lesions may be a collateral effect of these cells. Different types of animal models are established to study the underlying mechanisms of the mentioned pathophysiological processes. However, although they mimic some aspects of MS, it is impossible to dissect the exact mechanism and time course of ''collateral'' neuronal cell death. To address this course, here we show a protocol to study the mechanisms and time response of neuronal damage following an oligodendrocyte-directed CD8+ T cell attack. To target only the myelin sheath and the oligodendrocytes, in vitro activated oligodendrocyte-specific CD8+ T-cells are transferred into acutely isolated brain slices. After a defined incubation period, myelin and neuronal damage can be analysed in different regions of interest. Potential applications and limitations of this model will be discussed.
Collapse
Affiliation(s)
| | - Stefan Bittner
- Department of Neurology, University of Münster; Germany and Interdisciplinary Center for Clinical Research (IZKF) Münster
| | - Manuela Cerina
- Institute of Physiology I - Neuropathophysiology I, University of Münster
| | | | | | - Sven G Meuth
- Department of Neurology, University of Münster; Institute of Physiology I - Neuropathophysiology I, University of Münster
| |
Collapse
|
9
|
Paraneoplastic CDR2 and CDR2L antibodies affect Purkinje cell calcium homeostasis. Acta Neuropathol 2014; 128:835-52. [PMID: 25341622 PMCID: PMC4231287 DOI: 10.1007/s00401-014-1351-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/29/2014] [Accepted: 09/29/2014] [Indexed: 12/20/2022]
Abstract
Paraneoplastic cerebellar degeneration (PCD) is characterized by loss of Purkinje cells (PCs) associated with progressive pancerebellar dysfunction in the presence of onconeural Yo antibodies. These antibodies recognize the cerebellar degeneration-related antigens CDR2 and CDR2L. Response to PCD therapy is disappointing due to limited understanding of the neuropathological mechanisms. Here, we report the pathological role of CDR antibodies on the calcium homeostasis in PCs. We developed an antibody-mediated PCD model based on co-incubation of cerebellar organotypic slice culture with human patient serum or rabbit CDR2 and CDR2L antibodies. The CDR antibody-induced pathology was investigated by high-resolution multiphoton imaging and biochemical analysis. Both human and rabbit CDR antibodies were rapidly internalized by PCs and led to reduced immunoreactivity of calbindin D28K (CB) and L7/Pcp-2 as well as reduced dendritic arborizations in the remaining PCs. Washout of the CDR antibodies partially recovered CB immunoreactivity, suggesting a transient structural change in CB calcium-binding site. We discovered that CDR2 and CB co-immunoprecipitate. Furthermore, the expression levels of voltage-gated calcium channel Cav2.1, protein kinase C gamma and calcium-dependent protease, calpain-2, were increased after CDR antibody internalization. Inhibition of these signaling pathways prevented or attenuated CDR antibody-induced CB and L7/Pcp-2 immunoreactivity loss, morphological changes and increased protein expression. These results signify that CDR antibody internalization causes dysregulation of cell calcium homeostasis. Hence, drugs that modulate these events may represent novel neuroprotective therapies that limit the damaging effects of CDR antibodies and prevent PC neurodegeneration.
Collapse
|
10
|
Fingolimod attenuates splenocyte-induced demyelination in cerebellar slice cultures. PLoS One 2014; 9:e99444. [PMID: 24911000 PMCID: PMC4049809 DOI: 10.1371/journal.pone.0099444] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 05/15/2014] [Indexed: 12/02/2022] Open
Abstract
The family of sphingosine-1-phosphate receptors (S1PRs) is G-protein-coupled, comprised of subtypes S1PR1-S1PR5 and activated by the endogenous ligand S1P. The phosphorylated version of Fingolimod (pFTY720), an oral therapy for multiple sclerosis (MS), induces S1PR1 internalisation in T cells, subsequent insensitivity to S1P gradients and sequestering of these cells within lymphoid organs, thus limiting immune response. S1PRs are also expressed in neuronal and glial cells where pFTY720 is suggested to directly protect against lysolecithin-induced deficits in myelination state in organotypic cerebellar slices. Of note, the effect of pFTY720 on immune cells already migrated into the CNS, prior to treatment, has not been well established. We have previously found that organotypic slice cultures do contain immune cells, which, in principle, could also be regulated by pFTY720 to maintain levels of myelin. Here, a mouse organotypic cerebellar slice and splenocyte co-culture model was thus used to investigate the effects of pFTY720 on splenocyte-induced demyelination. Spleen cells isolated from myelin oligodendrocyte glycoprotein immunised mice (MOG-splenocytes) or from 2D2 transgenic mice (2D2-splenocytes) both induced demyelination when co-cultured with mouse organotypic cerebellar slices, to a similar extent as lysolecithin. As expected, in vivo treatment of MOG-immunised mice with FTY720 inhibited demyelination induced by MOG-splenocytes. Importantly, in vitro treatment of MOG- and 2D2-splenocytes with pFTY720 also attenuated demyelination caused by these cells. In addition, while in vitro treatment of 2D2-splenocytes with pFTY720 did not alter cell phenotype, pFTY720 inhibited the release of the pro-inflammatory cytokines such as interferon gamma (IFNγ) and interleukin 6 (IL6) from these cells. This work suggests that treatment of splenocytes by pFTY720 attenuates demyelination and reduces pro-inflammatory cytokine release, which likely contributes to enhanced myelination state induced by pFTY720 in organotypic cerebellar slices.
Collapse
|
11
|
Sheridan GK, Dev KK. S1P1 receptor subtype inhibits demyelination and regulates chemokine release in cerebellar slice cultures. Glia 2011; 60:382-92. [DOI: 10.1002/glia.22272] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Accepted: 11/01/2011] [Indexed: 12/14/2022]
|
12
|
Kang SS, Herz J, Kim JV, Nayak D, Stewart-Hutchinson P, Dustin ML, McGavern DB. Migration of cytotoxic lymphocytes in cell cycle permits local MHC I-dependent control of division at sites of viral infection. ACTA ACUST UNITED AC 2011; 208:747-59. [PMID: 21464219 PMCID: PMC3135345 DOI: 10.1084/jem.20101295] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Virus-specific cytotoxic CD8+ T cells are in cell cycle as they transit from lymphoid tissues to sites of infection. After virus infection, cytotoxic T lymphocytes (CTLs) divide rapidly to eradicate the pathogen and prevent the establishment of persistence. The magnitude of an antiviral CTL response is thought to be controlled by the initiation of a cell cycle program within lymphoid tissues. However, it is presently not known whether this division program proceeds during migration or is influenced locally at sites of viral infection. We demonstrate that antiviral CTLs remain in cell cycle while transiting to infected tissues. Up to one third of virus-specific CTLs within blood were found to be in cell cycle after infection with lymphocytic choriomeningitis virus or vesicular stomatitis virus. Using two-photon microscopy, we found that effector CTL divided rapidly upon arrest in the virus-infected central nervous system as well as in meningeal blood vessels. We also observed that MHC I–dependent interactions, but not costimulation, influenced the division program by advancing effector CTL through stages of the cell cycle. These results demonstrate that CTLs are poised to divide in transit and that their numbers can be influenced locally at the site of infection through interactions with cells displaying cognate antigen.
Collapse
Affiliation(s)
- Silvia S Kang
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Zozulya AL, Ortler S, Fabry Z, Sandor M, Wiendl H. The level of B7 homologue 1 expression on brain DC is decisive for CD8 Treg cell recruitment into the CNS during EAE. Eur J Immunol 2009; 39:1536-43. [PMID: 19424967 PMCID: PMC2889907 DOI: 10.1002/eji.200839165] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
DC in the CNS have emerged as the major rate-limiting factor for immune invasion and subsequent neuroinflammation during EAE. The mechanism of how this is regulated by brain-localized DC remains unknown. Here, we describe the ability of brain-localized DC expressing B7-H1 molecules to recruit CD8(+) T cells to the site of inflammation. Using intracerebral microinjections of B7-homologue 1-deficient DC, we demonstrate a substantial brain infiltration of CD8(+) T cells displaying a regulatory phenotype (CD122(+)) and function, resulting in a decrease of EAE peak clinical values. The recruitment of regulatory-type CD8(+) T cells into the CNS and the role of brain DC expressing B7-homologue 1 molecules in this process open up the possibility of DC-targeted therapeutic manipulation of neuroinflammatory diseases.
Collapse
MESH Headings
- Animals
- B7-1 Antigen/metabolism
- B7-H1 Antigen
- Brain/cytology
- Brain/immunology
- Brain/pathology
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Movement/immunology
- Cell Proliferation
- Central Nervous System/immunology
- Central Nervous System/pathology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/transplantation
- Encephalomyelitis, Autoimmune, Experimental/diagnosis
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Female
- Glycoproteins/administration & dosage
- Glycoproteins/immunology
- Immune Tolerance/physiology
- Interleukin-2 Receptor beta Subunit/metabolism
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Myelin-Oligodendrocyte Glycoprotein
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Peptides/metabolism
- Receptors, CCR6/metabolism
- Spleen/cytology
- Spleen/immunology
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Vaccination/methods
Collapse
Affiliation(s)
- Alla L. Zozulya
- University of Wuerzburg, Department of Neurology, Wuerzburg, Germany
| | - Sonja Ortler
- University of Wuerzburg, Department of Neurology, Wuerzburg, Germany
| | - Zsuzsanna Fabry
- University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, WI, USA
| | - Matyas Sandor
- University of Wisconsin-Madison, Department of Pathology and Laboratory Medicine, WI, USA
| | - Heinz Wiendl
- University of Wuerzburg, Department of Neurology, Wuerzburg, Germany
| |
Collapse
|
14
|
Kang SS, McGavern DB. Inflammation on the mind: visualizing immunity in the central nervous system. Curr Top Microbiol Immunol 2009; 334:227-63. [PMID: 19521688 PMCID: PMC4988846 DOI: 10.1007/978-3-540-93864-4_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The central nervous system (CNS) is a remarkably complex structure that utilizes electrochemical signaling to coordinate activities throughout the entire body. Because the nervous system contains nonreplicative cells, it is postulated that, through evolutionary pressures, this compartment has acquired specialized mechanisms to limit damage. One potential source of damage comes from our immune system, which has the capacity to survey the CNS and periphery for the presence of foreign material. The immune system is equipped with numerous effector mechanisms and can greatly alter the homeostasis and function of the CNS. Degeneration, autoimmunity, and pathogen infection can all result in acute, and sometimes chronic, inflammation within the CNS. Understanding the specialized functionality of innate and adaptive immune cells within the CNS is critical to the design of more efficacious treatments to mitigate CNS inflammatory conditions. Much of our knowledge of CNS-immune interactions stems from seminal studies that have used static and dynamic imaging approaches to visualize inflammatory cells responding to different CNS conditions. This review will focus on how imaging techniques have elevated our understanding of CNS inflammation as well as the exciting prospects that lie ahead as we begin to pursue investigation of the inflamed CNS in real time.
Collapse
Affiliation(s)
- Silvia S. Kang
- National Institutes of Neurological Disorders and Stroke, National Institutes of Health (NIH), 10 Center Drive, Bldg 10, Rm 7C213, Bethesda, MD 20892, USA
| | - Dorian B. McGavern
- National Institutes of Neurological Disorders and Stroke, National Institutes of Health (NIH), 10 Center Drive, Bldg 10, Rm 7C213, Bethesda, MD 20892, USA
| |
Collapse
|
15
|
Abstract
T-cell mediated immunotherapy is a conceptually attractive treatment option to envisage for glioma, since T lymphocytes can actively seek out neoplastic cells in the brain, and they have the potential to safely and specifically eliminate tumor. Some antigenic targets on glioma cells are already defined, and we can be optimistic that more will be discovered from progress in T-cell epitope identification and gene expression profiling of brain tumors. In parallel, advances in immunology (regional immunology, neuroimmunology, tumor immunology) now equip us to build upon the results from current immunotherapy trials in which the safety and feasibility of brain tumor immunotherapy have already been confirmed. We can now look to the next phase of immunotherapy, in which we must harness the most promising basic science advances and existing clinical expertise, and apply these to randomized clinical trials to determine the real clinical impact and applicability of these approaches for treating patients with currently incurable malignant brain tumors.
Collapse
Affiliation(s)
- Erwin G. Meir
- School of Medicine, Emory University, Clifton Road 1365C, Atlanta, 30322 U.S.A
| |
Collapse
|
16
|
Fabry Z, Schreiber HA, Harris MG, Sandor M. Sensing the microenvironment of the central nervous system: immune cells in the central nervous system and their pharmacological manipulation. Curr Opin Pharmacol 2008; 8:496-507. [PMID: 18691672 PMCID: PMC2614337 DOI: 10.1016/j.coph.2008.07.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Revised: 07/09/2008] [Accepted: 07/11/2008] [Indexed: 12/16/2022]
Abstract
Immune responses are highly regulated in all organs and severely restricted in certain tissues within the central nervous system (CNS). This phenomenon, called 'immune privilege', has been linked to the existence of multiple anatomical and physiological protective mechanisms. The finely balanced anti-inflammatory microenvironment within the CNS contributes to the immune privilege status of this tissue. The regulation of this compartment changes under pathological conditions when pro-inflammatory mediators might dominate. The past few years brought a wealth of novel information fostering our understanding of how CNS resident cells regulate the functions of immune cells, particularly helper T lymphocytes (Ths) and dendritic cells (DCs). These two cell types play a crucial role in the initiation and maintenance of neuroinflammatory diseases. The change from anti-inflammatory to pro-inflammatory microenvironment in the inflamed CNS affects Th and DC accumulation and function in the nervous tissue. A new era of DC-targeted therapies has begun, with the possibility of designing novel immunomodulatory therapies to intervene with neuroinflammation in a wide range of neurological diseases.
Collapse
Affiliation(s)
- Zsuzsanna Fabry
- School of Medicine and Public Health, Department of Pathology and Laboratory Medicine, 6130 MSC University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA.
| | | | | | | |
Collapse
|