22 results on '"Natalia Pikor"'
Search Results
2. Secondary B cell receptor diversification is necessary for T cell mediated neuro-inflammation during experimental autoimmune encephalomyelitis.
- Author
-
Georgina Galicia, Bryant Boulianne, Natalia Pikor, Alberto Martin, and Jennifer L Gommerman
- Subjects
Medicine ,Science - Abstract
Clinical studies of B cell depletion in Multiple Sclerosis (MS) have revealed that B Lymphocytes are involved in the neuro-inflammatory process, yet it remains unclear how B cells can exert pro- and anti-inflammatory functions during MS. Experimental Autoimmune Encephalomyelitis (EAE) is an animal model of MS whereby myelin-specific T cells become activated and subsequently migrate to the Central Nervous System (CNS) where they perform pro-inflammatory functions such as cytokine secretion. Typically EAE is induced by immunization of mice of a susceptible genetic background with peptide antigen emulsified in Complete Freund's Adjuvant. However, novel roles for B-lymphocytes in EAE may also be explored by immunization with full-length myelin oligodendrocyte glycoprotein (MOG) that contains the B cell conformational epitope. Here we show that full length MOG immunization promotes a chronic disease in mice that depends on antigen-driven secondary diversification of the B cell receptor.Activation-Induced Deaminase (AID) is an enzyme that is essential for antigen-driven secondary diversification of the B cell receptor. We immunized AID(-/-) mice with the extracellular domain (amino acids 1-120) of recombinant human MOG protein (rhMOG) and examined the incidence and severity of disease in AID(-/-) versus wild type mice. Corresponding with these clinical measurements, we also evaluated parameters of T cell activation in the periphery and the CNS as well as the generation of anti-MOG antibodies (Ab).AID(-/-) mice exhibit reduced severity and incidence of EAE. This suggests that the secondary diversification of the B cell receptor is required for B cells to exert their full encephalogenic potential during rhMOG-induced EAE, and possibly also during MS.
- Published
- 2013
- Full Text
- View/download PDF
3. Differentiation and activation of fibroblastic reticular cells
- Author
-
Natalia Pikor, Mechthild Lütge, and Burkhard Ludewig
- Subjects
0301 basic medicine ,Stromal cell ,Immunology ,Cell ,cell‐fate mapping ,Context (language use) ,Cell Communication ,Adaptive Immunity ,Biology ,Transcriptome ,single‐cell RNA‐sequencing ,transcriptomics ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Reticular cell ,medicine ,Animals ,Immunology and Allergy ,Invited Reviews ,Invited Review ,lymphoid tissue organizer cell ,Cell Differentiation ,Fibroblasts ,Acquired immune system ,030104 developmental biology ,medicine.anatomical_structure ,fibroblastic reticular cells ,Lymph Nodes ,Stromal Cells ,Neuroscience ,Function (biology) ,030215 immunology - Abstract
Secondary lymphoid organs (SLO) are underpinned by fibroblastic reticular cells (FRC) that form dedicated microenvironmental niches to secure induction and regulation of innate and adaptive immunity. Distinct FRC subsets are strategically positioned in SLOs to provide niche factors and govern efficient immune cell interaction. In recent years, the use of specialized mouse models in combination with single‐cell transcriptomics has facilitated the elaboration of the molecular FRC landscape at an unprecedented resolution. While single‐cell RNA‐sequencing has advanced the resolution of FRC subset characterization and function, the high dimensionality of the generated data necessitates careful analysis and validation. Here, we reviewed novel findings from high‐resolution transcriptomic analyses that refine our understanding of FRC differentiation and activation processes in the context of infection and inflammation. We further discuss concepts, strategies, and limitations for the analysis of single‐cell transcriptome data from FRCs and the wide‐ranging implications for our understanding of stromal cell biology.
- Published
- 2021
- Full Text
- View/download PDF
4. Insights into coronavirus immunity taught by the murine coronavirus
- Author
-
Sarah Grabherr, Burkhard Ludewig, and Natalia Pikor
- Subjects
0301 basic medicine ,Systemic disease ,viruses ,Host–pathogen interaction ,Immunology ,Reviews ,Virulence ,Immune responses ,Disease ,Adaptive Immunity ,Biology ,medicine.disease_cause ,Severity of Illness Index ,SARS‐CoV‐2 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Mouse hepatitis virus ,Mouse Hepatitis Virus ,Immunity ,medicine ,Animals ,Humans ,Immunology and Allergy ,Basic ,Coronavirus ,Murine hepatitis virus ,SARS-CoV-2 ,virus diseases ,respiratory system ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,medicine.disease ,Virology ,Animal models ,respiratory tract diseases ,Disease Models, Animal ,Viral Tropism ,Highlights ,Host/pathogen interaction ,030104 developmental biology ,Severe acute respiratory syndrome-related coronavirus ,Spike Glycoprotein, Coronavirus ,Middle East Respiratory Syndrome Coronavirus ,Review|Basic ,Coronavirus Infections ,030215 immunology - Abstract
Coronaviruses (CoVs) represent enveloped, ss RNA viruses with the ability to infect a range of vertebrates causing mainly lung, CNS, enteric, and hepatic disease. While the infection with human CoV is commonly associated with mild respiratory symptoms, the emergence of SARS‐CoV, MERS‐CoV, and SARS‐CoV‐2 highlights the potential for CoVs to cause severe respiratory and systemic disease. The devastating global health burden caused by SARS‐CoV‐2 has spawned countless studies seeking clinical correlates of disease severity and host susceptibility factors, revealing a complex network of antiviral immune circuits. The mouse hepatitis virus (MHV) is, like SARS‐CoV‐2, a beta‐CoV and is endemic in wild mice. Laboratory MHV strains have been extensively studied to reveal coronavirus virulence factors and elucidate host mechanisms of antiviral immunity. These are reviewed here with the aim to identify translational insights for SARS‐CoV‐2 learned from murine CoVs., Common properties of the infection etiology, host‐pathogen interactions, and immune responses shared between the mouse hepatitis virus (MHV‐A59) and SARS‐CoV‐2. A main distinguisher of MHV from other preclinical animal models of COVID‐19 is the fully adapted host replication machinery that recapitulates a multiorgan disease.
- Published
- 2021
- Full Text
- View/download PDF
5. Development and Immunological Function of Lymph Node Stromal Cells
- Author
-
Lucas Onder, Natalia Pikor, Hung-Wei Cheng, and Burkhard Ludewig
- Subjects
Stromal cell ,Immunology ,Cell ,Cell Differentiation ,Inflammation ,Adaptive Immunity ,Biology ,Lymphocyte Activation ,Cell biology ,medicine.anatomical_structure ,Immune system ,Cellular Microenvironment ,Genetic model ,medicine ,Lymph node stromal cell ,Animals ,Humans ,Immunology and Allergy ,Lymph Nodes ,Lymphocytes ,Lymph ,Stromal Cells ,medicine.symptom ,Lymph node - Abstract
Stromal cells have for a long time been viewed as structural cells that support distinct compartments within lymphoid tissues and little more. Instead, an active cross-talk between endothelial and fibroblastic stromal cells drives the maturation of lymphoid niches, a relationship that is recapitulated during lymph node organogenesis, steady-state conditions, and following inflammation. In this review, we go over recent advances in genetic models and high-resolution transcriptomic analyses that have propelled the finer resolution of the stromal cell infrastructure of lymph nodes, revealing that the distinct subsets are strategically positioned to deliver a catered mixture of niche factors to interacting immune cell populations. Moreover, we discuss how changes in the activation state of poised stromal cell–underpinned niches rather than on-demand differentiation of new stromal cell subsets govern the efficient interaction of Ag, APC, and cognate B and T lymphocytes during adaptive immune responses.
- Published
- 2021
- Full Text
- View/download PDF
6. B cell zone reticular cell microenvironments shape CXCL13 gradient formation
- Author
-
Urs Mörbe, Rita Pinter, Mariagrazia Uguccioni, Charles J.N. Lacey, Mark Coles, Marcus Thelen, Mark C. Leake, Zhaoukun Zhou, Natalia Pikor, Jason Cosgrove, Tom Cupedo, Anne Thuery, Jens V. Stein, Jon Timmis, Jovana Cupovic, Stefan Albrecht, Manfred Heller, Bénédicte Manoury, Kieran Alden, Mario Novkovic, Daniel F. Legler, Helen Miller, Burkhard Ludewig, Wojciech G. Polak, Marlene Wolf, Andrew Coatesworth, Daniel Venetz, Lucas Onder, Emily Taylor, Peter O'Toole, Simon Jarrett, Cantonal Hospital St. Gallen (KSSG), Brustzentrum Kantonsspital St. Gallen, Surgery, and Hematology
- Subjects
0301 basic medicine ,Palatine Tonsil ,Cell ,General Physics and Astronomy ,Adaptive Immunity ,[SDV.IMM.II]Life Sciences [q-bio]/Immunology/Innate immunity ,Cathepsin B ,Extracellular matrix ,Mice ,0302 clinical medicine ,Reticular cell ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,610 Medicine & health ,lcsh:Science ,Mice, Knockout ,B-Lymphocytes ,Multidisciplinary ,Chemistry ,Recombinant Proteins ,Extracellular Matrix ,medicine.anatomical_structure ,Cellular Microenvironment ,030220 oncology & carcinogenesis ,Imaging the immune system ,Lymph node ,Chemokines ,Science ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Models, Biological ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,ddc:570 ,Extracellular ,medicine ,Animals ,Humans ,Computer Simulation ,CXCL13 ,B cell ,B cells ,General Chemistry ,Chemokine CXCL13 ,030104 developmental biology ,Microscopy, Fluorescence ,Cell culture ,Biophysics ,lcsh:Q ,Stromal Cells ,Dendritic Cells, Follicular - Abstract
Through the formation of concentration gradients, morphogens drive graded responses to extracellular signals, thereby fine-tuning cell behaviors in complex tissues. Here we show that the chemokine CXCL13 forms both soluble and immobilized gradients. Specifically, CXCL13+ follicular reticular cells form a small-world network of guidance structures, with computer simulations and optimization analysis predicting that immobilized gradients created by this network promote B cell trafficking. Consistent with this prediction, imaging analysis show that CXCL13 binds to extracellular matrix components in situ, constraining its diffusion. CXCL13 solubilization requires the protease cathepsin B that cleaves CXCL13 into a stable product. Mice lacking cathepsin B display aberrant follicular architecture, a phenotype associated with effective B cell homing to but not within lymph nodes. Our data thus suggest that reticular cells of the B cell zone generate microenvironments that shape both immobilized and soluble CXCL13 gradients., Morphogens such as chemokines form gradients to direct graded responses and modulate cell behaviors. Here the authors show, using imaging and computer simulation, that the chemokine CXCL13 originated from follicular reticular cells in the lymph nodes forms both soluble and immobilized gradients to regulate B cell recruitment and migration.
- Published
- 2020
- Full Text
- View/download PDF
7. Stromal Cell Niches in the Inflamed Central Nervous System
- Author
-
Jovana Cupovic, Jennifer L. Gommerman, Natalia Pikor, Lucas Onder, and Burkhard Ludewig
- Subjects
Central Nervous System ,0301 basic medicine ,Encephalomyelitis, Autoimmune, Experimental ,Stromal cell ,Endothelium ,Immunology ,Central nervous system ,Inflammation ,Biology ,Blood–brain barrier ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cell Movement ,medicine ,Lymph node stromal cell ,Animals ,Humans ,Immunology and Allergy ,Lymphocytes ,Lymph node ,Fibroblasts ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Blood-Brain Barrier ,Lymph Nodes ,Stromal Cells ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
Inflammation in the CNS must be tightly regulated to respond efficiently to infection with neurotropic pathogens. Access of immune cells to the CNS and their positioning within the tissue are controlled by stromal cells that construct the barriers of the CNS. Although the role of the endothelium in regulating the passage of leukocytes and small molecules into the CNS has been studied extensively, the contribution of fibroblastic stromal cells as portals of entry into the CNS was only recently uncovered. We review the critical immune-stimulating role of meningeal fibroblasts in promoting recruitment and retention of lymphocytes during CNS inflammation. Activated meningeal fibroblastic stromal cells have the capacity to rapidly elaborate an immune-competent niche that sustains protective immune cells entering the CNS from the draining cervical lymph node. Such stromal cell niches can ultimately foster the establishment of tertiary lymphoid tissues during chronic neuroinflammatory conditions.
- Published
- 2017
- Full Text
- View/download PDF
8. Type I Interferon Signaling Disrupts the Hepatic Urea Cycle and Alters Systemic Metabolism to Suppress T Cell Function
- Author
-
Jörg Menche, Hatoon Baazim, Bettina Gürtl, Andreas Bergthaler, Peter Májek, Burkhard Ludewig, Maria Ozsvar-Kozma, Gernot Schabbauer, Michael Caldera, Anannya Bhattacharya, Anna Orlova, Ulrich Kalinke, Lindsay Kosack, Daniela Reil, Julia S. Brunner, Natalia Pikor, Moritz F. Schlapansky, Paul N. Cheng, Jakob-Wendelin Genger, Dijana Vitko, Michael Trauner, Alexander Lercher, Keiryn L. Bennett, Richard Moriggl, Benedikt Agerer, Kristaps Klavins, Theresa Pinter, Alexandra Popa, and TWINCORE, Zentrum für experimentelle und klinische Infektionsforschung GmbH,Feodor-Lynen Str. 7, 30625 Hannover, Germany.
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Male ,Ornithine ,immunometabolism ,Receptor, Interferon alpha-beta ,CD8-Positive T-Lymphocytes ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Interferon ,Cricetinae ,Chlorocebus aethiops ,Immunology and Allergy ,Lymphocytic choriomeningitis virus ,Urea ,hepatitis ,Mice, Knockout ,3. Good health ,Cell biology ,interferons ,Infectious Diseases ,Liver ,030220 oncology & carcinogenesis ,Urea cycle ,Interferon Type I ,Female ,medicine.symptom ,medicine.drug ,Signal Transduction ,Immunology ,Alpha interferon ,CD8 T cells ,Inflammation ,virus ,Biology ,Lymphocytic Choriomeningitis ,liver ,Lymphocytic choriomeningitis ,Arginine ,Cell Line ,03 medical and health sciences ,hepatocyte ,urea cycle ,medicine ,Animals ,Vero Cells ,Ornithine Carbamoyltransferase ,medicine.disease ,infection ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,inflammation ,Hepatocytes ,Drug metabolism ,CD8 - Abstract
Summary Infections induce complex host responses linked to antiviral defense, inflammation, and tissue damage and repair. We hypothesized that the liver, as a central metabolic hub, may orchestrate systemic metabolic changes during infection. We infected mice with chronic lymphocytic choriomeningitis virus (LCMV), performed RNA sequencing and proteomics of liver tissue, and integrated these data with serum metabolomics at different infection phases. Widespread reprogramming of liver metabolism occurred early after infection, correlating with type I interferon (IFN-I) responses. Viral infection induced metabolic alterations of the liver that depended on the interferon alpha/beta receptor (IFNAR1). Hepatocyte-intrinsic IFNAR1 repressed the transcription of metabolic genes, including Otc and Ass1, which encode urea cycle enzymes. This led to decreased arginine and increased ornithine concentrations in the circulation, resulting in suppressed virus-specific CD8+ T cell responses and ameliorated liver pathology. These findings establish IFN-I-induced modulation of hepatic metabolism and the urea cycle as an endogenous mechanism of immunoregulation. Video Abstract Download : Download video (18MB)
- Published
- 2019
9. Immunological gene signatures in B cell follicle reticular cells are highly conserved across organs and species
- Author
-
Mechthild Lütge, Lucas Onder, Hung-Wei Cheng, Yves Stanossek, Angelina De Martin, Lisa Spannagel, Charlotte Soneson, Mark Robinson, Natalia Pikor, and Burkhard Ludewig
- Subjects
Immunology ,Immunology and Allergy - Abstract
Secondary lymphoid organs (SLO) such as the spleen, lymph nodes and Peyer’s patches are strategically positioned to survey bodily surfaces and to support the generation of cellular and humoral immunity. The movement and interaction of antigens, antigen presenting cells, B and T lymphocytes within SLOs is coordinated by specialized fibroblastic reticular cells (FRCs) that form dedicated microenvironments and provide essential niche molecules such as the chemokine CXCL13. High-resolution transcriptomic analysis of Cxcl13-expressing cells in mouse models has previously enabled the molecular characterization of heterogenous B cell-interacting reticular cells (BRC) in lymph nodes. However, it remains unknown to what extent the molecular identity of niche-forming BRCs is conserved across SLOs. Here, we employed single cell RNA-sequencing of Cxcl13-expressing cells from murine lymph node, spleen and Peyer’s patch to compare the molecular identity of BRCs across SLOs. While structural and developmental genes dominated organ-specific gene signatures, we found conserved gene signatures reflecting crucial immunomodulatory functions. The highest conservation was observed in follicular dendritic cells, a BRC subset specialized in the capture and presentation of antigen. Moreover, immunomodulatory gene signatures were preserved in BRCs from human lymph nodes and palatine tonsils highlighting the important role of BRC-defined microenvironments in steering efficient immune responses in SLOs across species.
- Published
- 2021
- Full Text
- View/download PDF
10. Integration of Th17- and Lymphotoxin-Derived Signals Initiates Meningeal-Resident Stromal Cell Remodeling to Propagate Neuroinflammation
- Author
-
Hanane Touil, Brendan Heiden, Samuel K. Ludwin, Paul O'Connor, Jennifer L. Gommerman, Valera Castanov, Deepali Malhotra, Robert Kay, Shannon J. Turley, Alexandre Prat, Natalia Pikor, Amit Bar-Or, Lesley A. Ward, Joy Qu, Susan J. Armstrong, Georgina Galicia, Jillian L. Astarita, Valeria Ramaglia, Louis Boon, Leslie Summers-Deluca, and Claudia X. Dominguez
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Male ,Lymphotoxin alpha ,Chemokine ,Encephalomyelitis, Autoimmune, Experimental ,Stromal cell ,Immunology ,Inflammation ,Polymerase Chain Reaction ,Extracellular matrix ,Mice ,03 medical and health sciences ,Meninges ,Multiple Sclerosis, Relapsing-Remitting ,0302 clinical medicine ,medicine ,Animals ,Humans ,Immunology and Allergy ,Lymphotoxin-alpha ,Neuroinflammation ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,biology ,Flow Cytometry ,Immunohistochemistry ,Cell biology ,Infectious Diseases ,medicine.anatomical_structure ,Lymphotoxin ,biology.protein ,Th17 Cells ,Female ,Stromal Cells ,medicine.symptom ,Signal Transduction ,030215 immunology - Abstract
SummaryTertiary lymphoid tissues (TLTs) have been observed in the meninges of multiple sclerosis (MS) patients, but the stromal cells and molecular signals that support TLTs remain unclear. Here, we show that T helper 17 (Th17) cells induced robust TLTs within the brain meninges that were associated with local demyelination during experimental autoimmune encephalitis (EAE). Th17-cell-induced TLTs were underpinned by a network of stromal cells producing extracellular matrix proteins and chemokines, enabling leukocytes to reside within, rather than simply transit through, the meninges. Within the CNS, interactions between lymphotoxin αβ (LTαβ) on Th17 cells and LTβR on meningeal radio-resistant cells were necessary for the propagation of de novo interleukin-17 responses, and activated T cells from MS patients expressed elevated levels of LTβR ligands. Therefore, input from both Th17 cells and the lymphotoxin pathway induce the formation of an immune-competent stromal cell niche in the meninges.
- Published
- 2015
- Full Text
- View/download PDF
11. Origin and Differentiation Trajectories of Fibroblastic Reticular Cells in the Splenic White Pulp
- Author
-
Thomas Hehlgans, Thomas Rülicke, Mario Novkovic, Jun-ichi Miyazaki, Natalia Pikor, Elke Scandella, Mark D. Robinson, Lucas Onder, Hung-Wei Cheng, Mechthild Lütge, Charlotte Soneson, and Burkhard Ludewig
- Subjects
medicine.anatomical_structure ,Stromal cell ,Lymphotoxin ,Reticular cell ,Cell ,medicine ,Biology ,Progenitor cell ,Embryonic stem cell ,Mural cell ,Progenitor ,Cell biology - Abstract
The splenic white pulp is underpinned by poorly characterized stromal cells that demarcate distinct immune cell microenvironments. Here, definition of the embryonic origin and tracing of the differentiation trajectories of fibroblastic reticular cells (FRCs) was enabled by the establishment of FRC-specific fate-mapping in mice. We found that all reticular cell subsets descend from pluripotent progenitors that emerge at embryonic day 19.5 from Sca-1 periarterial progenitors. Commitment of FRC progenitors was concluded during the first week of postnatal life through occupation of niches along developing central arterioles. Single cell transcriptomic analysis facilitated deconvolution of FRC differentiation trajectories and indicated that perivascular reticular cells function both as adult lymphoid organizer cells and mural cell progenitors. Finally, the lymphotoxin-β receptor-independent sustenance of postnatal progenitor stemness unveiled that systemic immune surveillance in the splenic white pulp is governed through subset specification of reticular cells from a pluripotent periarterial progenitor cell.
- Published
- 2018
- Full Text
- View/download PDF
12. Lymphatic Endothelial Cells Control Initiation of Lymph Node Organogenesis
- Author
-
Thomas Rülicke, Shinichiro Sawa, Lucas Onder, Jennifer L. Gommerman, Elke Scandella, Klaus Pfeffer, Thomas Hehlgans, Burkhard Ludewig, Hung Wei Cheng, Christopher G. Mueller, Ari Waisman, Mario Novkovic, Natalia Pikor, Burkhard Becher, Urs Mörbe, Cantonal Hospital St. Gallen (KSSG), Brustzentrum Kantonsspital St. Gallen, Institute of Medical Microbiology and Hospital Hygiene (University of Düsseldorf), Heinrich Heine Universität Düsseldorf = Heinrich Heine University [Düsseldorf], Universität Zürich [Zürich] = University of Zurich (UZH), Department of Internal Medicine, Johannes Gutenberg - Universität Mainz (JGU), Directors's Laboratory, University of Zurich, and Ludewig, Burkhard
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,government.form_of_government ,Organogenesis ,[SDV]Life Sciences [q-bio] ,Immunology ,610 Medicine & health ,Mice, Transgenic ,Biology ,Choristoma ,10263 Institute of Experimental Immunology ,03 medical and health sciences ,Mice ,Immune system ,Lymphotoxin beta Receptor ,medicine ,Lymph node stromal cell ,Immunology and Allergy ,Animals ,Lymph node ,Cells, Cultured ,ComputingMilieux_MISCELLANEOUS ,2403 Immunology ,Receptor Activator of Nuclear Factor-kappa B ,Mesenchymal stem cell ,NF-kappa B ,Endothelial Cells ,Cell Differentiation ,Mesenchymal Stem Cells ,2725 Infectious Diseases ,Embryo, Mammalian ,Cell biology ,Mice, Inbred C57BL ,Haematopoiesis ,Lymphatic Endothelium ,Receptors, Lysosphingolipid ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Lymphatic system ,2723 Immunology and Allergy ,government ,570 Life sciences ,biology ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Lymph ,Lymph Nodes ,Signal Transduction - Abstract
Lymph nodes (LNs) are strategically situated throughout the body at junctures of the blood vascular and lymphatic systems to direct immune responses against antigens draining from peripheral tissues. The current paradigm describes LN development as a programmed process that is governed through the interaction between mesenchymal lymphoid tissue organizer (LTo) cells and hematopoietic lymphoid tissue inducer (LTi) cells. Using cell-type-specific ablation of key molecules involved in lymphoid organogenesis, we found that initiation of LN development is dependent on LTi-cell-mediated activation of lymphatic endothelial cells (LECs) and that engagement of mesenchymal stromal cells is a succeeding event. LEC activation was mediated mainly by signaling through receptor activator of NF-κB (RANK) and the non-canonical NF-κB pathway and was steered by sphingosine-1-phosphate-receptor-dependent retention of LTi cells in the LN anlage. Finally, the finding that pharmacologically enforced interaction between LTi cells and LECs promotes ectopic LN formation underscores the central LTo function of LECs.
- Published
- 2017
- Full Text
- View/download PDF
13. Association of Checkpoint Inhibitor–Induced Toxic Effects With Shared Cancer and Tissue Antigens in Non–Small Cell Lung Cancer
- Author
-
Christoph Driessen, Christoph Jakob Ackermann, Omar Ali, Alfred Zippelius, Wolfram Jochum, Hung-Wei Cheng, Lukas Flatz, Mirjam Fässler, Ahmed N. Hegazy, Sergey Nikolaev, Mübeccel Akdis, Mike Recher, Natalia Pikor, Jonathan H. Goldman, Willem van de Veen, Burkhard Ludewig, Heinz Läubli, Jacques Neefjes, Martin Früh, David Bomze, Sandra S. Ring, Stefan Diem, Cristina Del Carmen Gil Cruz, Fiamma Berner, Rebekka Niederer, Antonio Cozzio, Fabienne Hartmann, Daniel E. Speiser, Petra Baumgaertner, and Gideon Hönger
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,T-Lymphocytes ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Pembrolizumab ,Antibodies, Monoclonal, Humanized ,medicine.disease_cause ,Autoimmune Diseases ,Autoimmunity ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Antigens, Neoplasm ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Humans ,Medicine ,030212 general & internal medicine ,Adverse effect ,Prospective cohort study ,Lung cancer ,Aged ,business.industry ,Correction ,Cancer ,Immunotherapy ,Middle Aged ,medicine.disease ,Nivolumab ,Treatment Outcome ,030220 oncology & carcinogenesis ,Female ,business - Abstract
Immunotherapy with checkpoint inhibitors targeting the PD-1 (programmed cell death 1) axis has brought notable progress in patients with non-small cell lung cancer (NSCLC) and other cancers. However, autoimmune toxic effects are frequent and poorly understood, making it important to understand the pathophysiologic processes of autoimmune adverse effects induced by checkpoint inhibitor therapy.To gain mechanistic insight into autoimmune skin toxic effects induced by anti-PD-1 treatment in patients with non-small cell lung cancer.This prospective cohort study was conducted from July 1, 2016, to December 31, 2018. Patients (n = 73) with non-small cell lung cancer who received anti-PD-1 therapy (nivolumab or pembrolizumab) were recruited from 4 different centers in Switzerland (Kantonsspital St Gallen, Spital Grabs, Spital Wil, and Spital Flawil). Peripheral blood mononuclear cells, tumor biopsy specimens and biopsies from sites of autoimmune skin toxic effects were collected over a 2-year period, with patient follow-up after 1 year.Response to treatment, overall survival, progression-free survival, and development of autoimmune toxic effects (based on standard laboratory values and clinical examinations).Of the cohort of 73 patients with NSCLC (mean [SD] age, 68.1 [8.9] years; 44 [60%] men), 25 (34.2% [95% CI, 24.4%-45.7%]) developed autoimmune skin toxic effects, which were more frequent in patients with complete remission or partial remission (68.2% [95% CI, 47.3%-83.6%]) than those with progressive or stable disease (19.6% [95% CI, 11.0%-32.5%]) (χ2 = 14.02, P .001). Nine T-cell antigens shared between tumor tissue and skin were identified. These antigens were able to stimulate CD8+ and CD4+ T cells in vitro. Several of the antigen-specific T cells found in blood samples were also present in autoimmune skin lesions and lung tumors of patients who responded to anti-PD-1 therapy.These findings highlight a potential mechanism of checkpoint inhibitor-mediated autoimmune toxic effects and describe the association between toxic effects and response to therapy; such an understanding will help in controlling adverse effects, deciphering new cancer antigens, and further improving immunotherapy.
- Published
- 2019
- Full Text
- View/download PDF
14. CCL19-producing fibroblastic stromal cells restrain lung carcinoma growth by promoting local antitumor T-cell responses
- Author
-
Regulo Rodriguez, Maximilian Boesch, Lucas Onder, Jovana Cupovic, Mario Novkovic, Natalia Pikor, Ignazio Tarantino, Wolfram Jochum, Hung-Wei Cheng, Tino Schneider, Burkhard Ludewig, and Martin Brutsche
- Subjects
0301 basic medicine ,Lung Neoplasms ,Stromal cell ,T-Lymphocytes ,T cell ,Immunology ,Mice, Transgenic ,Biology ,Carcinoma, Lewis Lung ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Immunology and Allergy ,Lung cancer ,Tumor microenvironment ,Lewis lung carcinoma ,Cancer ,Dendritic cell ,Fibroblasts ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Chemokine CCL19 ,Stromal Cells ,Transcriptome - Abstract
Background A particular characteristic of non–small cell lung cancer is the composition of the tumor microenvironment with a very high proportion of fibroblastic stromal cells (FSCs). Objective Lapses in our basic knowledge of fibroblast phenotype and function in the tumor microenvironment make it difficult to define whether FSC subsets exist that exhibit either tumor-promoting or tumor-suppressive properties. Methods We used gene expression profiling of lung versus tumor FSCs from patients with non–small cell lung cancer. Moreover, CCL19-expressing FSCs were studied in transgenic mouse models by using a lung cancer metastasis model. Results CCL19 mRNA expression in human tumor FSCs correlates with immune cell infiltration and intratumoral accumulation of CD8 + T cells. Mechanistic dissection in murine lung carcinoma models revealed that CCL19-expressing FSCs form perivascular niches to promote accumulation of CD8 + T cells in the tumor. Targeted ablation of CCL19-expressing tumor FSCs reduced immune cell recruitment and resulted in unleashed tumor growth. Conclusion These data suggest that a distinct population of CCL19-producing FSCs fosters the development of an immune-stimulating intratumoral niche for immune cells to control cancer growth.
- Published
- 2018
- Full Text
- View/download PDF
15. Meningeal Tertiary Lymphoid Tissues and Multiple Sclerosis: A gathering place for diverse types of Immune Cells during CNS autoimmunity
- Author
-
Amit Bar-Or, Alexandre Prat, Natalia Pikor, and Jennifer L. Gommerman
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,Stromal cell ,T cell ,Mini Review ,Experimental autoimmune encephalomyelitis (EAE) ,Immunology ,experimental autoimmune encephalomyelitis ,Tertiary Lymphoid Tissues (TLT) ,Context (language use) ,multiple sclerosis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Meninges ,Medicine ,Immunology and Allergy ,Neuroinflammation ,B cell ,business.industry ,Multiple sclerosis ,Experimental autoimmune encephalomyelitis ,Follicle-like Structures (FLS) ,tertiary lymphoid tissues ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,multiple sclerosis (MS) ,Stromal Cells ,business ,lcsh:RC581-607 ,030217 neurology & neurosurgery - Abstract
Collections of leukocytes in the meningeal space have been documented in Multiple Sclerosis (MS). These meningeal aggregates, which in the context of other autoimmune diseases have often been termed Tertiary Lymphoid Tissues (TLT), have been associated with sub-pial cortical damage and disease progression. However, the key molecular and cellular signals required for their formation and maintenance, remain unclear. Herein we review TLT structures in other disease states in order to provide a framework for understanding these structures in the MS meninges. We then assess the evidence that the meningeal compartment serves as an important nexus for immune cells as well as a location for drainage of antigen into the cervical lymph node compartment. Extrapolating what is known about the molecular and cellular cues that initiate the formation of leukocyte aggregates in non-lymphoid tissues, we speculate on what signals lead to the formation and maintenance of meningeal TLT structures. Referring to the animal model of MS (Experimental Autoimmune Encephalomyelitis - EAE), we also explore what is known about these structures in supporting B cell and T cell responses during neuroinflammation. Lastly, we examine the evidence that connects these structures to ongoing neuropathology. Collectively, our review points to the meningeal compartment as an important player in neuroinflammatory processes. Moreover, we hypothesize that in order to gain insights into pro- and anti-inflammatory properties of lymphocytes in MS, one must understand the cellular scaffolds that support lymphocyte retention within the meninges, thus highlighting the importance of non-immune cells (stromal cells) in the neuroinflammatory process.
- Published
- 2016
- Full Text
- View/download PDF
16. B cells in the Multiple Sclerosis Central Nervous System: Trafficking and contribution to CNS-compartmentalized inflammation
- Author
-
Alexandre Prat, Jennifer L. Gommerman, Amit Bar-Or, Natalia Pikor, Hanane Touil, and Laure Michel
- Subjects
Central Nervous System ,lcsh:Immunologic diseases. Allergy ,B cells ,Multiple Sclerosis ,business.industry ,Multiple sclerosis ,Immunology ,Antigen presentation ,Central nervous system ,Meninges ,Inflammation ,Review ,medicine.disease ,Cerebrospinal fluid ,medicine.anatomical_structure ,trafficking ,medicine ,Immunology and Allergy ,Cytokine secretion ,meningeal inflammation ,medicine.symptom ,business ,lcsh:RC581-607 ,B cell - Abstract
Clinical trial results of peripheral B cell depletion indicate abnormal pro-inflammatory B cell properties, and particularly antibody-independent functions, contribute to relapsing MS disease activity. However, potential roles of B cells in progressive forms of disease continue to be debated. Prior work indicates that presence of B cells is fostered within the inflamed MS central nervous system (CNS) environment, and that B cell-rich immune-cell collections may be present within the meninges of patients. A potential association is reported between such meningeal immune-cell collections and the sub-pial pattern of cortical injury that is now considered important in progressive disease. Elucidating the characteristics of B cells that populate the MS CNS, how they traffic into the CNS and how they may contribute to progressive forms of the disease has become of considerable interest. Here, we will review characteristics of human B cells identified within distinct CNS sub-compartments of patients with MS, including the cerebrospinal fluid (CSF), parenchymal lesions and meninges, as well as the relationship between B cell populations identified in these sub-compartments and the periphery. We will further describe the different barriers of the CNS and the possible mechanisms of migration of B cells across these barriers. Finally, we will consider the range of human B cell responses (including potential for antibody production, cytokine secretion and antigen presentation) that may contribute to propagating inflammation and injury cascades thought to underlie MS progression.
- Published
- 2015
- Full Text
- View/download PDF
17. Secondary B cell receptor diversification is necessary for T cell mediated neuro-inflammation during experimental autoimmune encephalomyelitis
- Author
-
Alberto Martin, Georgina Galicia, Jennifer L. Gommerman, Bryant Boulianne, and Natalia Pikor
- Subjects
CD4-Positive T-Lymphocytes ,Mouse ,T-Lymphocytes ,lcsh:Medicine ,Mice ,0302 clinical medicine ,Infectious Diseases of the Nervous System ,Molecular Cell Biology ,Encephalomyelitis ,lcsh:Science ,0303 health sciences ,B-Lymphocytes ,Multidisciplinary ,biology ,Experimental autoimmune encephalomyelitis ,Brain ,Animal Models ,medicine.anatomical_structure ,Neurology ,Medicine ,Cytokines ,Cellular Types ,Research Article ,Multiple Sclerosis ,Encephalomyelitis, Autoimmune, Experimental ,T cell ,Immune Cells ,B-cell receptor ,Receptors, Cell Surface ,Myelin oligodendrocyte glycoprotein ,Autoimmune Diseases ,03 medical and health sciences ,Model Organisms ,Cytidine Deaminase ,medicine ,Animals ,Humans ,Biology ,B cell ,030304 developmental biology ,Multiple sclerosis ,lcsh:R ,medicine.disease ,Demyelinating Disorders ,Immunoglobulin M ,Immunology ,biology.protein ,Cytokine secretion ,Clinical Immunology ,Immunization ,Myelin-Oligodendrocyte Glycoprotein ,lcsh:Q ,Lymph Nodes ,030217 neurology & neurosurgery ,Conformational epitope - Abstract
Background Clinical studies of B cell depletion in Multiple Sclerosis (MS) have revealed that B Lymphocytes are involved in the neuro-inflammatory process, yet it remains unclear how B cells can exert pro- and anti-inflammatory functions during MS. Experimental Autoimmune Encephalomyelitis (EAE) is an animal model of MS whereby myelin-specific T cells become activated and subsequently migrate to the Central Nervous System (CNS) where they perform pro-inflammatory functions such as cytokine secretion. Typically EAE is induced by immunization of mice of a susceptible genetic background with peptide antigen emulsified in Complete Freund's Adjuvant. However, novel roles for B-lymphocytes in EAE may also be explored by immunization with full-length myelin oligodendrocyte glycoprotein (MOG) that contains the B cell conformational epitope. Here we show that full length MOG immunization promotes a chronic disease in mice that depends on antigen-driven secondary diversification of the B cell receptor. Methods Activation-Induced Deaminase (AID) is an enzyme that is essential for antigen-driven secondary diversification of the B cell receptor. We immunized AID−/− mice with the extracellular domain (amino acids 1–120) of recombinant human MOG protein (rhMOG) and examined the incidence and severity of disease in AID−/− versus wild type mice. Corresponding with these clinical measurements, we also evaluated parameters of T cell activation in the periphery and the CNS as well as the generation of anti-MOG antibodies (Ab). Conclusions AID−/− mice exhibit reduced severity and incidence of EAE. This suggests that the secondary diversification of the B cell receptor is required for B cells to exert their full encephalogenic potential during rhMOG-induced EAE, and possibly also during MS.
- Published
- 2013
18. A sphingosine-1-phosphate receptor 1-directed agonist reduces central nervous system inflammation in a plasmacytoid dendritic cell-dependent manner
- Author
-
Natalia Pikor, Jordan A. Schwartz, Mario A. Ostrowski, Allen Jian, Leslie Summers-Deluca, Jennifer L. Gommerman, Georgina Galicia-Rosas, Barbara Nuesslein-Hildesheim, and Olga L. Rojas
- Subjects
Agonist ,Encephalomyelitis, Autoimmune, Experimental ,medicine.drug_class ,Sphingosine-1-phosphate receptor ,Encephalomyelitis ,T-Lymphocytes ,Immunology ,Inflammation ,Plasmacytoid dendritic cell ,Thiophenes ,Biology ,Proinflammatory cytokine ,Mice ,medicine ,Immunology and Allergy ,Animals ,Receptor ,Sphingosine-1-Phosphate Receptors ,Cell Aggregation ,B-Lymphocytes ,Experimental autoimmune encephalomyelitis ,Anti-Inflammatory Agents, Non-Steroidal ,Dendritic Cells ,medicine.disease ,Receptors, Lysosphingolipid ,beta-Alanine ,Female ,medicine.symptom - Abstract
Gradients of the sphingolipid sphingosine-1-phosphate (S1P) are responsible for the egress of lymphocytes from lymph nodes by activating the S1P1 receptor expressed on the surface of lymphocytes. Small molecule drugs that downregulate S1P receptors induce the sequestration of lymphocytes within lymph nodes, thus preventing lymphocytes from accessing sites of inflammation. In particular, FTY720, a pan-S1P receptor agonist, has been efficacious in the treatment of multiple sclerosis as well as its animal model, experimental autoimmune encephalomyelitis (EAE), by virtue of its ability to restrain lymphocytes within the lymph nodes, thus precluding their migration into the CNS. However, multiple leukocyte subsets express S1P receptors of varying types, and although it is beneficial to prevent transmigration of proinflammatory lymphocytes into the CNS, allowing access of regulatory leukocyte subsets to the CNS is desirable. In this study, we show that an S1P1-specific agonist (AUY954) is clinically efficacious in ameliorating pre-established EAE in SJL/J mice. Efficacy of AUY954 correlated with a reduction of lymphocytes in the CNS, but access of plasmacytoid dendritic cells (pDCs) to the CNS was unimpaired, and the presence of pDCs was found to be an important cofactor in mediating the clinical efficacy of AUY954. These results indicate that pDCs are important in quieting autoimmune responses during EAE, and that trafficking inhibitors that are permissive for pDC accumulation in the CNS may be of therapeutic value for the treatment of multiple sclerosis.
- Published
- 2012
19. Lymphotoxin-sensitive microenvironments in homeostasis and inflammation
- Author
-
Bryant Boulianne, Elisa A. Porfilio, Jennifer L. Gommerman, and Natalia Pikor
- Subjects
lcsh:Immunologic diseases. Allergy ,Stromal cell ,Follicle-like Structures (FLS) ,Immunology ,Inflammation ,Biology ,Fibroblastic Reticular Cell (FRC) ,follicular dendritic cell ,Lymph Node (LN) ,Mini Review Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,lymphotoxin ,fibroblastic reticular cell ,Lymph node stromal cell ,medicine ,Immunology and Allergy ,Lymphopoiesis ,030304 developmental biology ,0303 health sciences ,Follicular Dendritic Cell (FDC) ,Follicular dendritic cells ,ymphotoxin (LT) ,chemokine ,lymph node ,Cell biology ,follicle-like structures ,Lymphatic system ,Lymphotoxin ,medicine.symptom ,lcsh:RC581-607 ,030215 immunology - Abstract
Stromal cell micro-microenvironments within lymphoid tissues are designed to support immune cell homeostasis and to regulate ongoing immune responses to pathogens. Such stromal cell networks have been best characterized within lymphoid tissues including the spleen and peripheral lymph nodes, and systems for classifying stromal cell phenotypes and functions are emerging. In response to inflammation, stromal cell networks within lymphoid tissues change in order to accommodate and regulate lymphocyte activation. Local inflammation in non-lymphoid tissues can also induce de novo formation of lymphoid aggregates, which we term here "follicle-like structures". Of note, the stromal cell networks that underpin such follicles are not as well characterized and may be different depending on the anatomical site. However, one common element that is integral to the maintenance of stromal cell environments, either in lymphoid tissue or extra-lymphoid sites, is the constitutive regulation of stromal cell phenotype and/or function by the Lymphotoxin (LT) pathway. Here we discuss how the LT pathway influences stromal cell environments both in homeostasis and in the context of inflammation in lymphoid and non-lymphoid tissues.
- Published
- 2012
- Full Text
- View/download PDF
20. B cells in MS: Why, where and how?
- Author
-
Natalia Pikor and Jennifer L. Gommerman
- Subjects
Clinically isolated syndrome ,biology ,business.industry ,Multiple sclerosis ,Experimental autoimmune encephalomyelitis ,Germinal center ,General Medicine ,medicine.disease ,Myelin basic protein ,Myelin oligodendrocyte glycoprotein ,Myelin ,medicine.anatomical_structure ,Neurology ,Immunology ,biology.protein ,Medicine ,Neurology (clinical) ,business ,Neuroinflammation - Abstract
Multiple sclerosis (MS) is a chronic disease of the central nervous system (CNS), in which auto-aggressive lymphocytes participate in inflammation that causes myelin destruction. Although T lymphocytes have been viewed as important culprits in the inflammatory cascade that results in MS, clinical trial results and animal model data support a role for B lymphocytes in MS pathology. In spite of these encouraging results, the mechanism behind why B cell depletion might be effective for MS treatment remains unknown. Herein we summarize the state of our knowledge for how B cells and their antibody products may influence the initiation and or propagation of MS, drawing from human studies and animal model data.
- Published
- 2011
21. Substrain Differences Reveal Novel Disease-Modifying Gene Candidates That Alter the Clinical Course of a Rodent Model of Multiple Sclerosis
- Author
-
Georgina Galicia-Rosas, Dustin Defreitas, Lucy R. Osborne, Jennifer O’Leary, Lesley A. Ward, Jennifer L. Gommerman, Shalina S. Ousman, Natalia Pikor, Leslie Summers deLuca, and Trisha M. Finlay
- Subjects
Encephalomyelitis, Autoimmune, Experimental ,DNA Copy Number Variations ,Immunology ,Copy number analysis ,Endogeny ,Mice, Inbred Strains ,Biology ,Severity of Illness Index ,Article ,Myelin ,Mice ,Multiple Sclerosis, Relapsing-Remitting ,Adjuvants, Immunologic ,Species Specificity ,immune system diseases ,Cell Line, Tumor ,Weight Loss ,medicine ,Immunology and Allergy ,Animals ,Genetic Predisposition to Disease ,Myelin Proteolipid Protein ,B cell ,Cells, Cultured ,Multiple sclerosis ,Experimental autoimmune encephalomyelitis ,Mycobacterium tuberculosis ,medicine.disease ,Phenotype ,Neuronal Apoptosis-Inhibitory Protein ,Peptide Fragments ,Myelin proteolipid protein ,nervous system diseases ,Disease Models, Animal ,medicine.anatomical_structure ,Acute Disease ,Female - Abstract
Experimental autoimmune encephalomyelitis (EAE) is a rodent model of multiple sclerosis that is executed in animals by immunization with myelin Ag in adjuvant. The SJL/J autoimmune-prone strain of mouse has been used to model relapsing–remitting multiple sclerosis. However, significant variations in peak scores, timing of onset, and incidence are observed among laboratories, with the postacute (relapse) phase of the disease exhibiting significant inconsistency. We characterized two substrains of SJL/J mice that exhibit profoundly different EAE disease parameters. Induction of EAE in the first SJL/J substrain resulted in many cases of chronic EAE that was dominated by an aggressive B cell response to the immunizing Ag and to endogenous CNS Ags. In contrast, the other SJL/J substrain exhibited a relapsing–remitting form of EAE concomitant with an elevated number of cytokine-producing CD4+ T cells in the CNS. Exploiting these interstrain differences, we performed a genome-wide copy number analysis on the two disparate SJL/J substrains and discovered numerous gene-dosage differences. In particular, one inflammation-associated gene, Naip1, was present at a higher copy number in the SJL/J substrain that exhibited relapsing–remitting EAE. These results demonstrate that substrain differences, perhaps at the level of genomic copy number, can account for variability in the postacute phase of EAE and may drive chronic versus relapsing disease.
- Published
- 2010
22. A Th17/Lymphotoxin axis enables cytokine production by brain meningeal stromal cells and propagates de novo CNS-intrinsic T cell responses during inflammation
- Author
-
Robert Kay, Jennifer L. Gommerman, Natalia Pikor, Jillian L. Astarita, and Shannon J. Turley
- Subjects
Microglia ,medicine.medical_treatment ,T cell ,CD3 ,Immunology ,Experimental autoimmune encephalomyelitis ,Congenic ,Biology ,medicine.disease ,Myelin oligodendrocyte glycoprotein ,Immune system ,Cytokine ,medicine.anatomical_structure ,Neurology ,medicine ,biology.protein ,Immunology and Allergy ,Neurology (clinical) - Abstract
Background: What does it require to be at risk for multiple sclerosis (MS)? To investigate the mechanisms involved we apply experimental autoimmune encephalomyelitis (EAE) as a model of MS in rats. By cross-breeding the EAE-susceptible DA strain and the EAE-resistant PVG strain we identified several quantitative trait loci (QTL) governing the susceptibility to EAE. Approach and results: The R11R6 congenic strain has an EAEsusceptible DA background, but comprises a gene region on chromosome 4 from the EAE-resistant PVG and is itself resistant to active EAE immunization, but not passive transfer. The identified region could contain potentially eight genes encoding for C-type lectin (CLEC) family members: Clec4a, Clec4a2, Clec4a3, Clec4a1, Clec4b2, Clec4n, Cle4d and Clec4e. Using backcrossing strategies coupled with SNP genotyping we narrowed the difference to four CLECs from PVG origin: Clec4b2, Clec4n, Clec4d and Clec4e. Expression of these and other CLECs is associated to myeloid cells. In vitro-generated bone marrow-derived dendritic cells (BMDCs), monocyte-derived DCs (Mo-DCs) or microglia from R11R6 exhibited comparatively lower expression of Clec4d and Clec4e. Surprisingly, despite having dramatic resistance to EAE induction, the R11R6 congenic showed no defect in immunization per se: We observed neither differences in lymph node cell counts, nor frequencies of T-cells, B-cells or DCs. Ex-vivo re-stimulation with the immunogen (Myelin Oligodendrocyte Glycoprotein, MOG) yielded equivalent proliferation between DA and R11R6.We then assessed the inflammatory response in the CNS before onset, at onset and at the peak of the first relapse. We found few differences in the frequency of microglia, lymphocytes and granulocytes/macrophages (Gran/Ma) cells in the CNS. R11R6 rats had significantly more lymphocytes in the brain at onset and significantly less Gran/Ma at peak both in the brain and spinal cord. Therewas no significant difference in the frequency of CD3 T cells in the CNS. However, lymphocytes from R11R6 strain proliferated less and infiltrating CD4CD3 lymphocytes were less frequent in the spinal cord at peak. They also produced drastically less IL-17 at onset and peak in both brain and spinal cord. Furthermore, granulocyte-marker positive cells from the R11R6 strain expressed significantly lower levels of MHC II. Conclusions: Though R11R6 rat have no defect in mounting an immune response and generate CNS-reactive T-cells, they exhibit a dramatic confinement of the subsequent inflammatory response. As the only genetic difference in this congenic strain is the origin and expression of CLEC genes, we conclude that the expression of CLECs on myeloid cells governs the susceptibility to EAE in rats. As CLEC receptors are important for sensing pathogens and tissue damage, we assume their regulatory involvement in shaping the extent of inflammation during EAE. Currently, we are investigating the exact mechanisms in further detail.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.