Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Eur J Immunol ; 52(1): 161-177, 2022 01.
Article in English | MEDLINE | ID: mdl-34524684

ABSTRACT

The migration of CD4+ effector/memory T cells across the blood-brain barrier (BBB) is a critical step in MS or its animal model, EAE. T-cell diapedesis across the BBB can occur paracellular, via the complex BBB tight junctions or transcellular via a pore through the brain endothelial cell body. Making use of primary mouse brain microvascular endothelial cells (pMBMECs) as in vitro model of the BBB, we here directly compared the transcriptome profile of pMBMECs favoring transcellular or paracellular T-cell diapedesis by RNA sequencing (RNA-seq). We identified the atypical chemokine receptor 1 (Ackr1) as one of the main candidate genes upregulated in pMBMECs favoring transcellular T-cell diapedesis. We confirmed upregulation of ACKR1 protein in pMBMECs promoting transcellular T-cell diapedesis and in venular endothelial cells in the CNS during EAE. Lack of endothelial ACKR1 reduced transcellular T-cell diapedesis across pMBMECs under physiological flow in vitro. Combining our previous observation that endothelial ACKR1 contributes to EAE pathogenesis by shuttling chemokines across the BBB, the present data support that ACKR1 mediated chemokine shuttling enhances transcellular T-cell diapedesis across the BBB during autoimmune neuroinflammation.


Subject(s)
Blood-Brain Barrier , CD4-Positive T-Lymphocytes , Duffy Blood-Group System , Encephalomyelitis, Autoimmune, Experimental , Memory T Cells , Multiple Sclerosis , Receptors, Cell Surface , Transendothelial and Transepithelial Migration , Animals , Mice , Blood-Brain Barrier/immunology , CD4-Positive T-Lymphocytes/immunology , Duffy Blood-Group System/genetics , Duffy Blood-Group System/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/genetics , Inflammation/immunology , Memory T Cells/immunology , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Transendothelial and Transepithelial Migration/genetics , Transendothelial and Transepithelial Migration/immunology
2.
J Neuroinflammation ; 19(1): 304, 2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36527076

ABSTRACT

BACKGROUND: Listeria monocytogenes (Lm) is a bacterial pathogen of major concern for humans and ruminants due to its neuroinvasive potential and its ability to cause deadly encephalitis (neurolisteriosis). On one hand, polymorphonuclear neutrophils (PMN) are key players in the defense against Lm, but on the other hand intracerebral infiltration with PMN is associated with significant neural tissue damage. Lm-PMN interactions in neurolisteriosis are poorly investigated, and factors inducing PMN chemotaxis to infectious foci containing Lm in the central nervous system (CNS) remain unidentified. METHODS: In this study, we assessed bovine PMN chemotaxis towards Lm and supernatants of infected endogenous brain cell populations in ex vivo chemotaxis assays, to identify chemotactic stimuli for PMN chemotaxis towards Lm in the brain. In addition, microglial secretion of IL-8 was assessed both ex vivo and in situ. RESULTS: Our data show that neither Lm cell wall components nor intact bacteria elicit chemotaxis of bovine PMN ex vivo. Moreover, astrocytes and neural cells fail to induce bovine PMN chemotaxis upon infection. In contrast, supernatant from Lm infected microglia readily induced chemotaxis of bovine PMN. Microglial expression and secretion of IL-8 was identified during early Lm infection in vitro and in situ, although IL-8 blocking with a specific antibody could not abrogate PMN chemotaxis towards Lm infected microglial supernatant. CONCLUSIONS: These data provide evidence that host-derived rather than bacterial factors trigger PMN chemotaxis to bacterial foci in the CNS, that microglia have a primary role as initiators of bovine PMN chemotaxis into the brain during neurolisteriosis and that blockade of these factors could be a therapeutic target to limit intrathecal PMN chemotaxis and PMN associated damage in neurolisteriosis.


Subject(s)
Listeria monocytogenes , Humans , Animals , Cattle , Microglia , Neutrophils/metabolism , Chemotaxis , Interleukin-8/metabolism , Chemotaxis, Leukocyte
3.
Proc Natl Acad Sci U S A ; 114(4): E524-E533, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28069965

ABSTRACT

Activated leukocyte cell adhesion molecule (ALCAM) is a cell adhesion molecule found on blood-brain barrier endothelial cells (BBB-ECs) that was previously shown to be involved in leukocyte transmigration across the endothelium. In the present study, we found that ALCAM knockout (KO) mice developed a more severe myelin oligodendrocyte glycoprotein (MOG)35-55-induced experimental autoimmune encephalomyelitis (EAE). The exacerbated disease was associated with a significant increase in the number of CNS-infiltrating proinflammatory leukocytes compared with WT controls. Passive EAE transfer experiments suggested that the pathophysiology observed in active EAE was linked to the absence of ALCAM on BBB-ECs. In addition, phenotypic characterization of unimmunized ALCAM KO mice revealed a reduced expression of BBB junctional proteins. Further in vivo, in vitro, and molecular analysis confirmed that ALCAM is associated with tight junction molecule assembly at the BBB, explaining the increased permeability of CNS blood vessels in ALCAM KO animals. Collectively, our data point to a biologically important function of ALCAM in maintaining BBB integrity.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Endothelial Cells/metabolism , Activated-Leukocyte Cell Adhesion Molecule/genetics , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Homeostasis , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Severity of Illness Index , Spinal Cord/metabolism , Tight Junction Proteins/metabolism
4.
Int J Mol Sci ; 20(6)2019 Mar 21.
Article in English | MEDLINE | ID: mdl-30901861

ABSTRACT

Multiple sclerosis (MS) is the most common inflammatory disorder of the central nervous system (CNS) in young adults leading to severe disability. Besides genetic traits, environmental factors contribute to MS pathogenesis. Cigarette smoking increases the risk of MS in an HLA-dependent fashion, but the underlying mechanisms remain unknown. Here, we explored the effect of cigarette smoke exposure on spontaneous and induced models of experimental autoimmune encephalomyelitis (EAE) by evaluating clinical disease and, when relevant, blood leukocytes and histopathology. In the relapsing-remitting (RR) transgenic model in SJL/J mice, we observed very low incidence in both smoke-exposed and control groups. In the optico-spinal encephalomyelitis (OSE) double transgenic model in C57BL/6 mice, the early onset of EAE prevented a meaningful evaluation of the effects of cigarette smoke. In EAE models induced by immunization, daily exposure to cigarette smoke caused a delayed onset of EAE followed by a protracted disease course in SJL/J mice. In contrast, cigarette smoke exposure ameliorated the EAE clinical score in C57BL/6J mice. Our exploratory studies therefore show that genetic background influences the effects of cigarette smoke on autoimmune neuroinflammation. Importantly, our findings expose the challenge of identifying an animal model for studying the influence of cigarette smoke in MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/diagnosis , Encephalomyelitis, Autoimmune, Experimental/etiology , Genetic Background , Smoking/adverse effects , Age of Onset , Animals , Biopsy , Brain/metabolism , Brain/pathology , Disease Models, Animal , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/metabolism , Immunohistochemistry , Mice , Multiple Sclerosis/diagnosis , Multiple Sclerosis/etiology , Multiple Sclerosis/metabolism , Phenotype , Risk Assessment , Risk Factors , Severity of Illness Index , Spinal Cord/metabolism , Spinal Cord/pathology
5.
Eur J Immunol ; 46(10): 2481-2483, 2016 10.
Article in English | MEDLINE | ID: mdl-27469626

ABSTRACT

Considering the 3R rules of animal experimentation, we asked if refined scoring of experimental autoimmune encephalomyelitis (EAE) in mice improves documentation of clinical EAE allowing to perform a more powerful statistical analysis. Surprisingly, refined EAE scoring failed to improve statistical outcome comparing the overall disease courses between two groups of mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/diagnosis , Multiple Sclerosis/diagnosis , Research Design , Animals , Disease Models, Animal , Disease Progression , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Observer Variation , Severity of Illness Index
6.
Eur J Immunol ; 45(4): 1043-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25545837

ABSTRACT

The extravasation of CD4(+) effector/memory T cells (TEM cells) across the blood-brain barrier (BBB) is a crucial step in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) or multiple sclerosis (MS). Endothelial ICAM-1 and ICAM-2 are essential for CD4(+) TEM cell crawling on the BBB prior to diapedesis. Here, we investigated the influence of cell surface levels of endothelial ICAM-1 in determining the cellular route of CD4(+) TEM -cell diapedesis across cytokine treated primary mouse BBB endothelial cells under physiological flow. Inflammatory conditions, inducing high levels of endothelial ICAM-1, promoted rapid initiation of transcellular diapedesis of CD4(+) T cells across the BBB, while intermediate levels of endothelial ICAM-1 favored paracellular CD4(+) T-cell diapedesis. Importantly, the route of T-cell diapedesis across the BBB was independent of loss of BBB barrier properties. Unexpectedly, a low number of CD4(+) TEM cells was found to cross the inflamed BBB in the absence of endothelial ICAM-1 and ICAM-2 via an obviously alternatively regulated transcellular pathway. In vivo, this translated to the development of ameliorated EAE in ICAM-1(null) //ICAM-2(-/-) C57BL/6J mice. Taken together, our study demonstrates that cell surface levels of endothelial ICAM-1 rather than the inflammatory stimulus or BBB integrity influence the pathway of T-cell diapedesis across the BBB.


Subject(s)
Blood-Brain Barrier/immunology , CD4-Positive T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Intercellular Adhesion Molecule-1/metabolism , Transendothelial and Transepithelial Migration/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion/immunology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Endothelial Cells/immunology , Female , Immunologic Memory/immunology , Inflammation/immunology , Intercellular Adhesion Molecule-1/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/immunology
7.
Cells ; 13(5)2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38474388

ABSTRACT

Dendritic cell (DC) migration from peripheral tissues via afferent lymphatic vessels to draining lymph nodes (dLNs) is important for the organism's immune regulation and immune protection. Several lymphatic endothelial cell (LEC)-expressed adhesion molecules have thus far been found to support transmigration and movement within the lymphatic vasculature. In this study, we investigated the contribution of CD112, an adhesion molecule that we recently found to be highly expressed in murine LECs, to this process. Performing in vitro assays in the murine system, we found that transmigration of bone marrow-derived dendritic cells (BM-DCs) across or adhesion to murine LEC monolayers was reduced when CD112 was absent on LECs, DCs, or both cell types, suggesting the involvement of homophilic CD112-CD112 interactions. While CD112 was highly expressed in murine dermal LECs, CD112 levels were low in endogenous murine dermal DCs and BM-DCs. This might explain why we observed no defect in the in vivo lymphatic migration of adoptively transferred BM-DCs or endogenous DCs from the skin to dLNs. Compared to murine DCs, human monocyte-derived DCs expressed higher CD112 levels, and their migration across human CD112-expressing LECs was significantly reduced upon CD112 blockade. CD112 expression was also readily detected in endogenous human dermal DCs and LECs by flow cytometry and immunofluorescence. Upon incubating human skin punch biopsies in the presence of CD112-blocking antibodies, DC emigration from the tissue into the culture medium was significantly reduced, indicating impaired lymphatic migration. Overall, our data reveal a contribution of CD112 to human DC migration.


Subject(s)
Langerhans Cells , Lymphatic Vessels , Nectins , Animals , Humans , Mice , Cell Movement/physiology , Endothelium, Lymphatic , Langerhans Cells/physiology , Nectins/metabolism
8.
Brain Struct Funct ; 225(9): 2885-2895, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33128675

ABSTRACT

Analysis of the angioarchitecture and quantification of the conduit vessels and microvasculature is of paramount importance for understanding the physiological and pathological processes within the central nervous system (CNS). Most of the available in vivo imaging methods lack penetration depth and/or resolution. Some ex vivo methods may provide better resolution, but are mainly destructive, as they are designed for imaging the CNS tissues after their removal from the skull or vertebral column. The removal procedure inevitably alters the in situ relations of the investigated structures and damages the dura mater and leptomeninges. µAngiofil, a polymer-based contrast agent, permits a qualitatively novel postmortem microangio-computed tomography (microangioCT) approach with excellent resolution and, therefore, visualization of the smallest brain capillaries. The datasets obtained empower a rather straightforward quantitative analysis of the vascular tree, including the microvasculature. The µAngiofil has an excellent filling capacity as well as a radio-opacity higher than the one of bone tissue, which allows imaging the cerebral microvasculature even within the intact skull or vertebral column. This permits in situ visualization and thus investigation of the dura mater and leptomeningeal layers as well as their blood supply in their original geometry. Moreover, the methodology introduced here permits correlative approaches, i.e., microangioCT followed by classical histology, immunohistochemistry and even electron microscopy. The experimental approach presented here makes use of common desktop microCT scanners, rendering it a promising everyday tool for the evaluation of the (micro)vasculature of the central nervous system in preclinical and basic research.


Subject(s)
Brain/anatomy & histology , Brain/blood supply , Computed Tomography Angiography/methods , X-Ray Microtomography/methods , Animals , Brain/diagnostic imaging , Gliosarcoma/diagnostic imaging , Gliosarcoma/pathology , Image Processing, Computer-Assisted/methods , Mice, Inbred C57BL , Rats , Rats, Wistar , Swine/anatomy & histology , Swine, Miniature/anatomy & histology
9.
Sci Rep ; 9(1): 11714, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406267

ABSTRACT

The interleukin 7 receptor alpha chain (IL-7Rα) is predominately expressed by lymphocytes, and activation by its ligand IL-7 supports the development and maintenance of T cells and boosts T-cell mediated immunity. We recently reported that lymphatic endothelial cells (LECs) in dermal lymphatics also express IL-7 and its receptor chains (IL-7Rα and CD132) and that IL-7 supports lymphatic drainage. This suggested that activation of IL-7Rα signaling in lymphatics could exert inflammation-resolving activity, by promoting the clearance of excess tissue fluid. Here we investigated how the potentially opposing effects of IL-7Rα signaling in immune cells and in the lymphatic vasculature would affect the development and progression of psoriasis-like skin inflammation. We found that during acute and chronic skin inflammation mice with an endothelial-specific deletion of IL-7Rα (IL-7RαΔEC mice) developed more edema compared to control mice, as a consequence of impaired lymphatic drainage. However, systemic treatment of wild-type mice with IL-7 exacerbated edema and immune cell infiltration in spite of increasing lymphatic drainage, whereas treatment with IL-7Rα blocking antibody ameliorated inflammatory symptoms. These data identify IL-7Rα signaling as a new pathway in psoriasis-like skin inflammation and show that its pro-inflammatory effects on the immune compartment override its anti-inflammatory, drainage-enhancing effects on the endothelium.


Subject(s)
Antibodies, Neutralizing/pharmacology , CD4-Positive T-Lymphocytes/immunology , Endothelial Cells/immunology , Interleukin-7/immunology , Psoriasis/drug therapy , Receptors, Interleukin-7/immunology , Skin/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Gene Expression Regulation , Humans , Imiquimod/administration & dosage , Inflammation , Interleukin-7/genetics , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphatic Vessels/drug effects , Lymphatic Vessels/immunology , Lymphatic Vessels/pathology , Male , Mice , Mice, Inbred C57BL , Organ Specificity , Oxazolone/administration & dosage , Psoriasis/chemically induced , Psoriasis/genetics , Psoriasis/pathology , Receptors, Interleukin-7/antagonists & inhibitors , Receptors, Interleukin-7/genetics , Signal Transduction , Skin/drug effects , Skin/pathology , Tetradecanoylphorbol Acetate/administration & dosage , Tetradecanoylphorbol Acetate/analogs & derivatives
10.
Front Immunol ; 10: 3056, 2019.
Article in English | MEDLINE | ID: mdl-31993059

ABSTRACT

In experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), myelin-specific T cells are activated in the periphery and differentiate in T helper (Th) 1 and Th17 effector cells, which cross the blood-brain barrier (BBB) to reach the central nervous system (CNS), where they induce neuroinflammation. Here, we explored the role of intercellular adhesion molecule-1 (ICAM-1) and ICAM-2 in the activation of naïve myelin-specific T cells and in the subsequent migration of differentiated encephalitogenic Th1 and Th17 cells across the BBB in vitro and in vivo. While on antigen-presenting cells ICAM-1, but not ICAM-2 was required for the activation of naïve CD4+ T cells, endothelial ICAM-1 and ICAM-2 mediated both Th1 and Th17 cell migration across the BBB. ICAM-1/-2-deficient mice developed ameliorated typical and atypical EAE transferred by encephalitogenic Th1 and Th17 cells, respectively. Our study underscores important yet cell-specific contributions for ICAM-1 and ICAM-2 in EAE pathogenesis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Transendothelial and Transepithelial Migration/immunology , Animals , Antigens, CD/metabolism , Blood-Brain Barrier/metabolism , Cell Adhesion Molecules/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Th1 Cells/metabolism , Th17 Cells/metabolism
11.
Front Immunol ; 8: 406, 2017.
Article in English | MEDLINE | ID: mdl-28443093

ABSTRACT

T-cell migration across the blood-brain barrier (BBB) is a crucial step in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Two-photon intravital microscopy (2P-IVM) has been established as a powerful tool to study cell-cell interactions in inflammatory EAE lesions in living animals. In EAE, central nervous system inflammation is strongly pronounced in the spinal cord, an organ in which 2P-IVM imaging is technically very challenging and has been limited to the lumbar spinal cord. Here, we describe a novel spinal cord window preparation allowing to use 2P-IVM to image immune cell interactions with the cervical spinal cord microvascular endothelium during EAE. We describe differences in the angioarchitecture of the cervical spinal cord versus the lumbar spinal cord, which will entail different hemodynamic parameters in these different vascular beds. Using T cells as an example, we demonstrate the suitability of this novel methodology in imaging the post-arrest multistep T-cell extravasation across the cervical spinal cord microvessels. The novel methodology includes an outlook to the analysis of the cellular pathway of T-cell diapedesis across the BBB by establishing visualization of endothelial junctions in this vascular bed.

SELECTION OF CITATIONS
SEARCH DETAIL