Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cell ; 184(4): 1000-1016.e27, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33508229

ABSTRACT

Despite the established dogma of central nervous system (CNS) immune privilege, neuroimmune interactions play an active role in diverse neurological disorders. However, the precise mechanisms underlying CNS immune surveillance remain elusive; particularly, the anatomical sites where peripheral adaptive immunity can sample CNS-derived antigens and the cellular and molecular mediators orchestrating this surveillance. Here, we demonstrate that CNS-derived antigens in the cerebrospinal fluid (CSF) accumulate around the dural sinuses, are captured by local antigen-presenting cells, and are presented to patrolling T cells. This surveillance is enabled by endothelial and mural cells forming the sinus stromal niche. T cell recognition of CSF-derived antigens at this site promoted tissue resident phenotypes and effector functions within the dural meninges. These findings highlight the critical role of dural sinuses as a neuroimmune interface, where brain antigens are surveyed under steady-state conditions, and shed light on age-related dysfunction and neuroinflammatory attack in animal models of multiple sclerosis.


Subject(s)
Cranial Sinuses/immunology , Cranial Sinuses/physiology , Dura Mater/immunology , Dura Mater/physiology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/metabolism , Antigens/cerebrospinal fluid , Cellular Senescence , Chemokine CXCL12/pharmacology , Dura Mater/blood supply , Female , Homeostasis , Humans , Immunity , Male , Mice, Inbred C57BL , Phenotype , Stromal Cells/cytology , T-Lymphocytes/cytology
2.
Immunol Rev ; 325(1): 152-165, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38809041

ABSTRACT

Multiple sclerosis (MS) affects more than 2.8 million people worldwide but the distribution is not even. Although over 200 gene variants have been associated with susceptibility, studies of genetically identical monozygotic twin pairs suggest that the genetic make-up is responsible for only about 20%-30% of the risk to develop disease, while the rest is contributed by milieu factors. Recently, a new, unexpected player has entered the ranks of MS-triggering or facilitating elements: the human gut microbiota. In this review, we summarize the present knowledge of microbial effects on formation of a pathogenic autoreactive immune response targeting the distant central nervous system and delineate the approaches, both in people with MS and in MS animal models, which have led to this concept. Finally, we propose that a tight combination of investigations of human patients with studies of suitable animal models is the best strategy to functionally characterize disease-associated microbiota and thereby contribute to deciphering pathogenesis of a complex human disease.


Subject(s)
Disease Models, Animal , Gastrointestinal Microbiome , Multiple Sclerosis , Humans , Multiple Sclerosis/immunology , Multiple Sclerosis/etiology , Multiple Sclerosis/microbiology , Animals , Gastrointestinal Microbiome/immunology , Intestines/immunology , Intestines/microbiology , Autoimmunity
3.
Proc Natl Acad Sci U S A ; 120(47): e2300733120, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37956299

ABSTRACT

In multiple sclerosis (MS), pathogenic T cell responses are known to be important drivers of autoimmune inflammation. However, increasing evidence suggests an additional role for B cells, which may contribute to pathogenesis via antigen presentation and production of proinflammatory cytokines. However, these B cell effector functions are not featured well in classical experimental autoimmune encephalomyelitis (EAE) mouse models. Here, we compared properties of myelin oligodendrocyte glycoprotein (MOG)-specific and polyclonal B cells and developed an adjuvant-free cotransfer EAE mouse model, where highly activated, MOG-specific induced germinal center B cells provide the critical stimulus for disease development. We could show that high levels of MOG-specific immunoglobulin G (IgGs) are not required for EAE development, suggesting that antigen presentation and activation of cognate T cells by B cells may be important for pathogenesis. As our model allows for B cell manipulation prior to transfer, we found that overexpression of the proinflammatory cytokine interleukin (IL)-6 by MOG-specific B cells leads to an accelerated EAE onset accompanied by activation/expansion of the myeloid compartment rather than a changed T cell response. Accordingly, knocking out IL-6 or tumor necrosis factor α in MOG-specific B cells via CRISPR-Cas9 did not affect activation of pathogenic T cells. In summary, we generated a tool to dissect pathogenic B cell effector function in EAE development, which should improve our understanding of pathogenic processes in MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Mice , Animals , Cytokines , Autoimmunity , Myelin-Oligodendrocyte Glycoprotein , Interleukin-6 , Mice, Inbred C57BL
4.
Eur J Immunol ; 54(1): e2350561, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37850588

ABSTRACT

Multiple sclerosis (MS) is an immune-mediated inflammatory disease of the CNS. A defining characteristic of MS is the ability of autoreactive T lymphocytes to cross the blood-brain barrier and mediate inflammation within the CNS. Previous work from our lab found the gene Enpp2 to be highly upregulated in murine encephalitogenic T cells. Enpp2 encodes for the protein autotaxin, a secreted glycoprotein that catalyzes the production of lysophosphatidic acid and promotes transendothelial migration of T cells from the bloodstream into the lymphatic system. The present study sought to characterize autotaxin expression in T cells during CNS autoimmune disease and determine its potential therapeutic value. Myelin-activated CD4 T cells upregulated expression of autotaxin in vitro, and ex vivo analysis of CNS-infiltrating CD4 T cells showed significantly higher autotaxin expression compared with cells from healthy mice. In addition, inhibiting autotaxin in myelin-specific T cells reduced their encephalitogenicity in adoptive transfer studies and decreased in vitro cell motility. Importantly, using two mouse models of MS, treatment with an autotaxin inhibitor ameliorated EAE severity, decreased the number of CNS infiltrating T and B cells, and suppressed relapses, suggesting autotaxin may be a promising therapeutic target in the treatment of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Mice , Blood-Brain Barrier , CD4-Positive T-Lymphocytes , Central Nervous System , Mice, Inbred C57BL , Multiple Sclerosis/therapy , Multiple Sclerosis/metabolism
5.
Clin Immunol ; 259: 109898, 2024 02.
Article in English | MEDLINE | ID: mdl-38185267

ABSTRACT

Myelin antigen-reactive Th1 and Th17 cells are critical drivers of central nervous system (CNS) autoimmune inflammation. Transcription factors T-bet and RORγt play a crucial role in the differentiation and function of Th1 and Th17 cells, and impart them a pathogenic role in CNS autoimmune inflammation. Mice deficient in these two factors do not develop experimental autoimmune encephalomyelitis (EAE). While T-bet and RORγt are known to regulate the expression of several cell adhesion and migratory molecules in T cells, their role in supporting Th1 and Th17 trafficking to the CNS is not completely understood. More importantly, once Th1 and Th17 cells reach the CNS, how the function of these transcription factors modulates the local inflammatory response during EAE is unclear. In the present study, we showed that myelin oligodendrocyte glycoprotein 35-55 peptide (MOG35-55)-specific Th1 cells deficient in RORγt could cross the blood-brain barrier (BBB) but failed to induce demyelination, apoptosis of neurons, and EAE. Pathogenic Th17 cell-derived cytokines GM-CSF, TNF-α, IL-17A, and IL-21 significantly increased the surface expression of IL-23R on neuronal cells. Furthermore, we showed that, in EAE, neurons in the brain and spinal cord express IL-23R. IL-23-IL-23R signaling in neuronal cells caused phosphorylation of STAT3 (Ser727 and Tyr705) and induced cleaved caspase 3 and cleaved poly (ADP-ribose) polymerase-1 (PARP-1) molecules in an IL-23R-dependent manner and caused apoptosis. Thus, we provided a mechanism showing that T-bet is required to recruit pathogenic Th17 cells to the CNS and RORγt-mediated inflammatory response to drive the apoptosis of IL-23R+ neurons in the CNS and cause EAE. Understanding detailed molecular mechanisms will help to design better strategies to control neuroinflammation and autoimmunity. ONE SENTENCE SUMMARY: IL-23-IL-23R signaling promotes apoptosis of CNS neurons.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Mice , Animals , Th17 Cells , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Mice, Transgenic , Th1 Cells , Inflammation , Myelin-Oligodendrocyte Glycoprotein , Transcription Factors/metabolism , Interleukin-23/metabolism , Apoptosis , Neurons/metabolism , Neurons/pathology , Mice, Inbred C57BL
6.
Rev Neurol (Paris) ; 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39289136

ABSTRACT

The past two decades have seen a considerable paradigm shift in the way autoimmune CNS disorders are considered, diagnosed, and treated; largely due to the discovery of novel autoantibodies directed at neuroglial surface or intracellular targets. This approach has enabled multiple bona fide CNS autoantibody-associated diseases to thoroughly infiltrate the sphere of clinical neurology, facilitating advances in patient outcomes. This review focusses on the fundamental immunological concepts behind CNS autoantibody-associated diseases. First, we briefly review the broad phenotypic profiles of these conditions. Next, we explore concepts around immune checkpoints and the related B cell lineage. Thirdly, the sources of autoantibody production are discussed alongside triggers of tolerance failure, including neoplasms, infections and iatrogenic therapies. Penultimately, the role of T cells and leucocyte trafficking into the CNS are reviewed. Finally, biological insights from responses to targeted immunotherapies in different CNS autoantibody-associated diseases are summarised. The continued and rapid expansion of the CNS autoantibody-associated field holds promise for further improved diagnostic and therapeutic paradigms, ultimately leading to further improvements in patient outcomes.

7.
J Autoimmun ; 138: 103053, 2023 07.
Article in English | MEDLINE | ID: mdl-37236124

ABSTRACT

Hepatocyte nuclear factor 4 α (HNF4α), a transcription factor (TF) essential for embryonic development, has been recently shown to regulate the expression of inflammatory genes. To characterize HNF4a function in immunity, we measured the effect of HNF4α antagonists on immune cell responses in vitro and in vivo. HNF4α blockade reduced immune activation in vitro and disease severity in the experimental model of multiple sclerosis (MS). Network biology studies of human immune transcriptomes unraveled HNF4α together with SP1 and c-myc as master TF regulating differential expression at all MS stages. TF expression was boosted by immune cell activation, regulated by environmental MS risk factors and higher in MS immune cells compared to controls. Administration of compounds targeting TF expression or function demonstrated non-synergic, interdependent transcriptional control of CNS autoimmunity in vitro and in vivo. Collectively, we identified a coregulatory transcriptional network sustaining neuroinflammation and representing an attractive therapeutic target for MS and other inflammatory disorders.


Subject(s)
Autoimmunity , Hepatocyte Nuclear Factor 4 , Multiple Sclerosis , Humans , Autoimmunity/genetics , Gene Expression Regulation , Gene Regulatory Networks , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Transcriptome , Genes, myc
8.
Brain ; 144(9): 2625-2634, 2021 10 22.
Article in English | MEDLINE | ID: mdl-33848319

ABSTRACT

Although CSF analysis routinely enables the diagnosis of neurological diseases, it is mainly used for the gross distinction between infectious, autoimmune inflammatory, and degenerative disorders of the CNS. To investigate, whether a multi-dimensional cellular blood and CSF characterization can support the diagnosis of clinically similar neurological diseases, we analysed 546 patients with autoimmune neuroinflammatory, degenerative, or vascular conditions in a cross-sectional retrospective study. By combining feature selection with dimensionality reduction and machine learning approaches we identified pan-disease parameters that were altered across all autoimmune neuroinflammatory CNS diseases and differentiated them from other neurological conditions and inter-autoimmunity classifiers that subdifferentiate variants of CNS-directed autoimmunity. Pan-disease as well as diseases-specific changes formed a continuum, reflecting clinical disease evolution. A validation cohort of 231 independent patients confirmed that combining multiple parameters into composite scores can assist the classification of neurological patients. Overall, we showed that the integrated analysis of blood and CSF parameters improves the differential diagnosis of neurological diseases, thereby facilitating early treatment decisions.


Subject(s)
Inflammation Mediators/cerebrospinal fluid , Nervous System Diseases/cerebrospinal fluid , Nervous System Diseases/classification , Biomarkers/cerebrospinal fluid , Cohort Studies , Diagnosis, Differential , Female , Humans , Male , Nervous System Diseases/diagnosis , Retrospective Studies
9.
J Neuroinflammation ; 18(1): 27, 2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33468194

ABSTRACT

BACKGROUND: Follicular regulatory T (TFR) cells are essential for the regulation of germinal center (GC) response and humoral self-tolerance. Dysregulated follicular helper T (TFH) cell-GC-antibody (Ab) response secondary to dysfunctional TFR cells is the root of an array of autoimmune disorders. The contribution of TFR cells to the pathogenesis of multiple sclerosis (MS) and murine experimental autoimmune encephalomyelitis (EAE) remains largely unclear. METHODS: To determine the impact of dysregulated regulatory T cells (Tregs), TFR cells, and Ab responses on EAE, we compared the MOG-induced EAE in mice with a FoxP3-specific ablation of the transcription factor Blimp1 to control mice. In vitro co-culture assays were used to understand how Tregs and Ab regulate the activity of microglia and central nervous system (CNS)-infiltrating myeloid cells. RESULTS: Mice with a FoxP3-specific deletion of Blimp1 developed severe EAE and failed to recover compared to control mice, reflecting conversion of Tregs into interleukin (IL)-17A/granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing effector T cells associated with increased TFH-Ab responses, more IgE deposition in the CNS, and inability to regulate CNS CD11b+ myeloid cells. Notably, serum IgE titers were positively correlated with EAE scores, and culture of CNS CD11b+ cells with sera from these EAE mice enhanced their activation, while transfer of Blimp1-deficient TFR cells promoted Ab production, activation of CNS CD11b+ cells, and EAE. CONCLUSIONS: Blimp1 is essential for the maintenance of TFR cells and Ab responses in EAE. Dysregulated TFR cells and Ab responses promote CNS autoimmunity.


Subject(s)
Antibody Formation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Cell Differentiation/immunology , Germinal Center , Mice , Mice, Inbred C57BL
10.
Eur J Immunol ; 49(11): 2030-2043, 2019 11.
Article in English | MEDLINE | ID: mdl-31318439

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are found in the CNS during neuroinflammation and have been reported to exert regulatory functions in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). However, the mechanisms of entry of pDCs into the CNS as well as their phenotype and innate functional properties, once recruited into the CNS, have not been thoroughly examined. Herein, we show that pDCs rapidly accumulate into the brain and spinal cord during the acute phase of EAE, and maintain the expression of numerous phenotypic markers typical of peripheral pDCs. Functionally, CNS-pDCs constitutively expressed IRF7 and were able to rapidly produce type I IFNs and IL-12p40 upon ex vivo TLR-9 stimulation. Using adoptive transfer experiments, we provide evidence that CNS-pDC are recruited from the blood and accumulate into the CNS during the acute phase of EAE. Accumulation of pDCs into the CNS was strongly inhibited in the absence of CD29, but not CD18, suggesting a major role for ß1 but not ß2 integrins. Indeed, blocking the CD49d α4-integrins during acute EAE drastically diminished CNS-pDC numbers. Together, our results demonstrate that circulating pDCs are actively recruited into the CNS during acute EAE through a mechanism largely dependent on CD49d/CD29-integrins.


Subject(s)
Brain/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4/immunology , Integrin beta1/immunology , Spinal Cord/immunology , Adoptive Transfer , Animals , Brain/drug effects , Brain/pathology , Cell Movement/immunology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Dendritic Cells/transplantation , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Regulation , Immunity, Innate , Integrin alpha4/genetics , Integrin beta1/genetics , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/genetics , Interferon Type I/immunology , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Peptide Fragments/administration & dosage , Pertussis Toxin/administration & dosage , Signal Transduction , Spinal Cord/drug effects , Spinal Cord/pathology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
11.
J Neuroinflammation ; 17(1): 79, 2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32143718

ABSTRACT

The gut microbiota regulates the host immune and nervous systems and plays an important role in the pathogenesis of autoimmune neurological disease multiple sclerosis (MS). There are considerable efforts currently being undertaken to develop therapies for MS based on the modulation of microbiota. Evidence from experimental models suggests that the manipulation of microbiota through diet or antibiotics prior to the disease development limits disease susceptibility. However, it is currently unclear if microbiota manipulation therapies would also have an impact on ongoing neurological disease. Here, we examined the effect of antibiotic-based microbiota modulation in spontaneous experimental autoimmune encephalomyelitis (EAE) mouse models of MS before and after the onset of autoimmune disease. Prophylactic antibiotic treatment led to a significant reduction of susceptibility to spontaneous EAE. In contrast, antibiotic treatment after the onset of spontaneous EAE did not show a significant amelioration. These results reveal that the perturbation of gut bacteria alters disease susceptibility but has minimal impact on the ongoing neurological disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Gastrointestinal Microbiome/drug effects , Neuroimmunomodulation/physiology , Animals , Anti-Bacterial Agents/pharmacology , Brain/pathology , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Neuroimmunomodulation/drug effects , Spinal Cord/pathology
12.
J Neuroinflammation ; 17(1): 102, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32248814

ABSTRACT

Multiple sclerosis (MS) is the most common autoimmune disease of the CNS. The etiology of MS is still unclear but it is widely recognized that both genetic and environmental factors contribute to its pathogenesis. Immune signaling and responses are critically regulated by ubiquitination, a posttranslational modification that is promoted by ubiquitinating enzymes and inhibited by deubiquitinating enzymes (DUBs). Genome-wide association studies (GWASs) identified that polymorphisms in or in the vicinity of two human DUB genes TNFAIP3 and USP18 were associated with MS susceptibility. Studies with experimental autoimmune encephalomyelitis (EAE), an animal model of MS, have provided biological rationale for the correlation between these DUBs and MS. Additional studies have shown that other DUBs are also involved in EAE by controlling distinct cell populations. Therefore, DUBs are emerging as crucial regulators of MS/EAE and might become potential therapeutic targets for the clinical treatment of MS.


Subject(s)
Autoimmunity/immunology , Deubiquitinating Enzymes/metabolism , Encephalomyelitis, Autoimmune, Experimental/enzymology , Multiple Sclerosis/enzymology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Multiple Sclerosis/immunology
13.
J Neuroinflammation ; 16(1): 49, 2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30808363

ABSTRACT

BACKGROUND: Quinoline-3-carboxamides, such as laquinimod, ameliorate CNS autoimmunity in patients and reduce tumor cell metastasis experimentally. Previous studies have focused on the immunomodulatory effect of laquinimod on myeloid cells. The data contained herein suggest that quinoline-3-carboxamides improve the immunomodulatory and anti-tumor effects of NK cells by upregulating the adhesion molecule DNAX accessory molecule-1 (DNAM-1). METHODS: We explored how NK cell activation by laquinimod inhibits CNS autoimmunity in experimental autoimmune encephalomyelitis (EAE), the most utilized model of MS, and improves immunosurveillance of experimental lung melanoma metastasis. Functional manipulations included in vivo NK and DC depletion experiments and in vitro assays of NK cell function. Clinical, histological, and flow cytometric read-outs were assessed. RESULTS: We demonstrate that laquinimod activates natural killer (NK) cells via the aryl hydrocarbon receptor and increases their DNAM-1 cell surface expression. This activation improves the cytotoxicity of NK cells against B16F10 melanoma cells and augments their immunoregulatory functions in EAE by interacting with CD155+ dendritic cells (DC). Noteworthy, the immunosuppressive effect of laquinimod-activated NK cells was due to decreasing MHC class II antigen presentation by DC and not by increasing DC killing. CONCLUSIONS: This study clarifies how DNAM-1 modifies the bidirectional crosstalk of NK cells with CD155+ DC, which can be exploited to suppress CNS autoimmunity and strengthen tumor surveillance.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Autoimmunity/drug effects , Dendritic Cells/drug effects , Immunologic Surveillance/immunology , Killer Cells, Natural/drug effects , Quinolones/pharmacology , Animals , Antigens, Differentiation, T-Lymphocyte/immunology , Autoimmunity/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Transgenic , Quinolines/agonists , Receptors, Aryl Hydrocarbon/agonists , Receptors, Virus/immunology
14.
Int J Mol Sci ; 20(1)2019 Jan 07.
Article in English | MEDLINE | ID: mdl-30621022

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) leading to demyelination and axonal damage. It often affects young adults and can lead to neurological disability. Interferon ß (IFNß) preparations represent widely used treatment regimens for patients with relapsing-remitting MS (RRMS) with therapeutic efficacy in reducing disease progression and frequency of acute exacerbations. In mice, IFNß therapy has been shown to ameliorate experimental autoimmune encephalomyelitis (EAE), an animal model of MS while genetic deletion of IFNß or its receptor augments clinical severity of disease. However, the complex mechanism of action of IFNß in CNS autoimmunity has not been fully elucidated. Here, we review our current understanding of the origin, phenotype, and function of microglia and CNS immigrating macrophages in the pathogenesis of MS and EAE. In addition, we highlight the emerging roles of microglia as IFNß-producing cells and vice versa the impact of IFNß on microglia in CNS autoimmunity. We finally discuss recent progress in unraveling the underlying molecular mechanisms of IFNß-mediated effects in EAE.


Subject(s)
Autoimmunity , Central Nervous System/immunology , Interferon-beta/metabolism , Microglia/metabolism , Neuroprotective Agents/metabolism , Animals , Disease Models, Animal , Humans
15.
J Neurochem ; 146(2): 122-132, 2018 07.
Article in English | MEDLINE | ID: mdl-29574788

ABSTRACT

It is widely accepted that development of autoimmunity in the central nervous system (CNS) is triggered by autoreactive T cells, that are activated in the periphery and gain the capacity to migrate through endothelial cells at the blood-brain barrier (BBB) into the CNS. Upon local reactivation, an inflammatory cascade is initiated, that subsequently leads to a recruitment of additional immune cells ultimately causing demyelination and axonal damage. Even though the interaction of immune cells with the BBB has been in the focus of research for many years, the exact mechanisms of how immune cells enter and exit the CNS remains poorly understood. In this line, the factors deciding immune cell entry routes, lesion formation, cellular composition as well as distribution within the CNS have also not been elucidated. The following factors have been proposed to represent key determinants for lesion evaluation and distribution: (i) presence and density of (auto) antigens in the CNS, (ii) local immune milieu at sites of lesion development and resolution, (iii) trafficking routes and specific trafficking requirements, especially at the BBB and (iv) characteristics and phenotypes of CNS infiltrating cells and cell subsets (e.g. features of T helper subtypes or CD8 cells). The heterogeneity of lesion development within inflammatory demyelinating diseases remains poorly understood until today, but here especially orphan inflammatory CNS disorders such as neuromyelitis optica spectrum disorder (NMOSD), Rasmussen encephalitis or SUSAC syndrome might give important insights in critical determinants of lesion topography. Finally, investigating the interaction of T cells with the BBB using in vitro approaches or tracking of T cells in vivo in animals or even human patients, as well as the discovery of lymphatic vasculature in the CNS are teaching us new aspects during the development of CNS autoimmunity. In this review, we discuss recent findings which help to unravel mechanisms underlying lesion topography and might lead to new diagnostic or therapeutic approaches in neuroinflammatory disorders including multiple sclerosis (MS).


Subject(s)
Autoimmunity/immunology , Demyelinating Autoimmune Diseases, CNS/immunology , Demyelinating Autoimmune Diseases, CNS/pathology , Animals , Blood-Brain Barrier/physiopathology , Humans
16.
Brain Behav Immun ; 66: 382-393, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28642092

ABSTRACT

The CC chemokine ligand 17 (CCL17) and its cognate CC chemokine receptor 4 (CCR4) are known to control leukocyte migration, maintenance of TH17 cells, and regulatory T cell (Treg) expansion in vivo. In this study we characterized the expression and functional role of CCL17 in the pathogenesis of experimental autoimmune encephalomyelitis (EAE). Using a CCL17/EGFP reporter mouse model, we could show that CCL17 expression in the CNS can be found in a subset of classical dendritic cells (DCs) that immigrate into the CNS during the effector phase of MOG-induced EAE. CCL17 deficient (CCL17-/-) mice exhibited an ameliorated disease course upon MOG-immunization, associated with reduced immigration of IL-17 producing CD4+ T cells and peripheral DCs into the CNS. CCL17-/- DCs further showed equivalent MHC class II and costimulatory molecule expression and an equivalent capacity to secrete IL-23 and induce myelin-reactive TH17 cells when compared to wildtype DCs. In contrast, their transmigration in an in vitro model of the blood-brain barrier was markedly impaired. In addition, peripheral Treg cells were enhanced in CCL17-/- mice at peak of disease pointing towards an immunoregulatory function of CCL17 in EAE. Our study identifies CCL17 as a unique modulator of EAE pathogenesis regulating DC trafficking as well as peripheral Treg cell expansion in EAE. Thus, CCL17 operates at distinct levels and on different cell subsets during immune response in EAE, a property harboring therapeutic potential for the treatment of CNS autoimmunity.


Subject(s)
Chemokine CCL17/metabolism , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Cell Movement , Chemokine CCL17/genetics , Female , Interleukin-23/metabolism , Mice, Inbred C57BL , Mice, Knockout , Spleen/immunology , Spleen/physiopathology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism
17.
Int J Mol Sci ; 18(11)2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29099057

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS). It affects more than two million people worldwide, mainly young adults, and may lead to progressive neurological disability. Chemokines and their receptors have been shown to play critical roles in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a murine disease model induced by active immunization with myelin proteins or transfer of encephalitogenic CD4⁺ T cells that recapitulates clinical and neuropathological features of MS. Chemokine ligand-receptor interactions orchestrate leukocyte trafficking and influence multiple pathophysiological cellular processes, including antigen presentation and cytokine production by dendritic cells (DCs). The C-C class chemokines 17 (CCL17) and 22 (CCL22) and their C-C chemokine receptor 4 (CCR4) have been shown to play an important role in homeostasis and inflammatory responses. Here, we provide an overview of the involvement of CCR4 and its ligands in CNS autoimmunity. We review key clinical studies of MS together with experimental studies in animals that have demonstrated functional roles of CCR4, CCL17, and CCL22 in EAE pathogenesis. Finally, we discuss the therapeutic potential of newly developed CCR4 antagonists and a humanized anti-CCR4 antibody for treatment of MS.


Subject(s)
Autoimmune Diseases of the Nervous System/immunology , Autoimmunity , Chemokine CCL17/immunology , Chemokine CCL22/immunology , Receptors, CCR4/immunology , Animals , Autoimmune Diseases of the Nervous System/drug therapy , Autoimmune Diseases of the Nervous System/pathology , Autoimmunity/immunology , Central Nervous System/drug effects , Central Nervous System/immunology , Central Nervous System/pathology , Chemokine CCL17/analysis , Chemokine CCL22/analysis , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Molecular Targeted Therapy/methods , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Receptors, CCR4/analysis
18.
J Autoimmun ; 67: 90-101, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26584738

ABSTRACT

The increasing incidence in Multiple Sclerosis (MS) during the last decades in industrialized countries might be linked to a change in dietary habits. Nowadays, enhanced salt content is an important characteristic of Western diet and increased dietary salt (NaCl) intake promotes pathogenic T cell responses contributing to central nervous system (CNS) autoimmunity. Given the importance of macrophage responses for CNS disease propagation, we addressed the influence of salt consumption on macrophage responses in CNS autoimmunity. We observed that EAE-diseased mice receiving a NaCl-high diet showed strongly enhanced macrophage infiltration and activation within the CNS accompanied by disease aggravation during the effector phase of EAE. NaCl treatment of macrophages elicited a strong pro-inflammatory phenotype characterized by enhanced pro-inflammatory cytokine production, increased expression of immune-stimulatory molecules, and an antigen-independent boost of T cell proliferation. This NaCl-induced pro-inflammatory macrophage phenotype was accompanied by increased activation of NF-kB and MAPK signaling pathways. The pathogenic relevance of NaCl-conditioned macrophages is illustrated by the finding that transfer into EAE-diseased animals resulted in significant disease aggravation compared to untreated macrophages. Importantly, also in human monocytes, NaCl promoted a pro-inflammatory phenotype that enhanced human T cell proliferation. Taken together, high dietary salt intake promotes pro-inflammatory macrophages that aggravate CNS autoimmunity. Together with other studies, these results underline the need to further determine the relevance of increased dietary salt intake for MS disease severity.


Subject(s)
Autoimmunity , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Sodium Chloride, Dietary/administration & dosage , Animals , Autoimmunity/drug effects , Biomarkers , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Humans , Immunophenotyping , MAP Kinase Signaling System , Macrophage Activation/drug effects , Macrophages/drug effects , Mice , Monocytes/immunology , Monocytes/metabolism , Phenotype
19.
Acta Neuropathol ; 132(3): 413-31, 2016 09.
Article in English | MEDLINE | ID: mdl-27383204

ABSTRACT

Innate immune responses by myeloid cells decisively contribute to perpetuation of central nervous system (CNS) autoimmunity and their pharmacologic modulation represents a promising strategy to prevent disease progression in Multiple Sclerosis (MS). Based on our observation that peripheral immune cells from relapsing-remitting and primary progressive MS patients exhibited strongly decreased levels of the bile acid receptor FXR (farnesoid-X-receptor, NR1H4), we evaluated its potential relevance as therapeutic target for control of established CNS autoimmunity. Pharmacological FXR activation promoted generation of anti-inflammatory macrophages characterized by arginase-1, increased IL-10 production, and suppression of T cell responses. In mice, FXR activation ameliorated CNS autoimmunity in an IL-10-dependent fashion and even suppressed advanced clinical disease upon therapeutic administration. In analogy to rodents, pharmacological FXR activation in human monocytes from healthy controls and MS patients induced an anti-inflammatory phenotype with suppressive properties including control of effector T cell proliferation. We therefore, propose an important role of FXR in control of T cell-mediated autoimmunity by promoting anti-inflammatory macrophage responses.


Subject(s)
Autoimmunity/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-10/immunology , Myeloid Cells/metabolism , Receptors, Cytoplasmic and Nuclear/immunology , T-Lymphocytes/cytology , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , T-Lymphocytes/metabolism
20.
J Neuroimmunol ; 387: 578282, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38183947

ABSTRACT

Multiple sclerosis (MS) is a demyelinating disease characterized by infiltration of autoreactive T cells into the central nervous system (CNS). In order to understand how activated, autoreactive T cells are able to cross the blood brain barrier, the unique molecular characteristics of pathogenic T cells need to be more thoroughly examined. In previous work, our laboratory found autotaxin (ATX) to be upregulated by activated autoreactive T cells in the mouse model of MS. ATX is a secreted glycoprotein that promotes T cell chemokinesis and transmigration through catalysis of lysophoshphatidic acid (LPA). ATX is elevated in the serum of MS patients during active disease phases, and we previously found that inhibiting ATX decreases severity of neurological deficits in the mouse model. In this study, ATX expression was found to be lower in MS patient immune cells during rest, but significantly increased during early activation in a manner not seen in healthy controls. The ribosomal binding protein HuR, which stabilizes ATX mRNA, was also increased in MS patients in a similar pattern to that of ATX, suggesting it may be helping regulate ATX levels after activation. The proinflammatory cytokine interleukin-23 (IL-23) was shown to induce prolonged ATX expression in MS patient Th1 and Th17 cells. Finally, through ChIP, re-ChIP analysis, we show that IL-23 may be signaling through pSTAT3/pSTAT4 heterodimers to induce expression of ATX. Taken together, these findings elucidate cell types that may be contributing to elevated serum ATX levels in MS patients and identify potential drivers of sustained expression in encephalitogenic T cells.


Subject(s)
Multiple Sclerosis , Animals , Mice , Humans , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Central Nervous System/metabolism , Disease Models, Animal , Cytokines , Interleukin-23 , Lysophospholipids/genetics , Lysophospholipids/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL