Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Elife ; 112022 05 17.
Article in English | MEDLINE | ID: mdl-35578890

ABSTRACT

Thermal adaptation is an extensively used intervention for enhancing or suppressing thermogenic and mitochondrial activity in adipose tissues. As such, it has been suggested as a potential lifestyle intervention for body weight maintenance. While the metabolic consequences of thermal acclimation are not limited to the adipose tissues, the impact on the rest of the tissues in context of their gene expression profile remains unclear. Here, we provide a systematic characterization of the effects in a comparative multi-tissue RNA sequencing approach following exposure of mice to 10 °C, 22 °C, or 34 °C in a panel of organs consisting of spleen, bone marrow, spinal cord, brain, hypothalamus, ileum, liver, quadriceps, subcutaneous-, visceral- and brown adipose tissues. We highlight that transcriptional responses to temperature alterations exhibit a high degree of tissue-specificity both at the gene level and at GO enrichment gene sets, and show that the tissue-specificity is not directed by the distinct basic gene expression pattern exhibited by the various organs. Our study places the adaptation of individual tissues to different temperatures in a whole-organism framework and provides integrative transcriptional analysis necessary for understanding the temperature-mediated biological programming.


Humans, mice and most other mammals are constantly exposed to fluctuations in the temperature of their environment. These fluctuations cause striking metabolic effects in the body, for example, exposure to cold promotes burning of calories to generate heat, thereby reducing how much fat accumulates in the body. On the other hand, warmer temperatures strengthen the bones and protect against a bone disease known as osteoporosis. As such, it has been suggested that exposure to alternating warm or cold temperatures could be a potential lifestyle intervention that conveys various benefits to our health. Our body stores fat in tissues known as adipose tissues, which are found all over the body including under the skin and around our major organs and muscles. Exposure to cold triggers changes in the activities of some genes in the adipose tissues to burn more calories. But it remains unclear how temperature affects the activities of other organs with respect to their expression of genes in the whole-body context. Hadadi, Spiljar et al. used an RNA sequencing approach to study the activities of genes in various tissues of mice exposed to cold (10°C), room temperature (22°C), or mild warm (34°C). The experiments revealed numerous genes whose levels were different in the various organs and temperatures tested. Overall, adipose tissues experienced the biggest changes in gene levels between different temperatures, followed by tissues involved in immune responses, and the brain and spinal cord tissues. Each organ changed gene expression levels in its own way. , and this was not due to the different intimate gene expression profile between the various organs. These findings improve our understanding of how changes in temperature affect mammals by putting the responses of individual tissues into the context of the whole body. Hadadi, Spiljar et al. also generated a web-based, free-to-use application to allow others to view and further analyze the data collected in this work for gene levels in the various organs of interest.


Subject(s)
Cold Temperature , Transcriptome , Acclimatization/genetics , Adipose Tissue, Brown/metabolism , Animals , Mice , Thermogenesis
2.
Sci Transl Med ; 14(640): eabl6058, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35417190

ABSTRACT

In chronic inflammatory diseases of the central nervous system (CNS), immune cells persisting behind the blood-brain barrier are supposed to promulgate local tissue destruction. The drivers of such compartmentalized inflammation remain unclear, but tissue-resident memory T cells (TRM) represent a potentially important cellular player in this process. Here, we investigated whether resting CD8+ TRM persisting after cleared infection with attenuated lymphocytic choriomeningitis virus (LCMV) can initiate immune responses directed against cognate self-antigen in the CNS. We demonstrated that time-delayed conditional expression of the LCMV glycoprotein as neo-self-antigen by glia cells reactivated CD8+ TRM. Subsequently, CD8+ TRM expanded and initiated CNS inflammation and immunopathology in an organ-autonomous manner independently of circulating CD8+ T cells. However, in the absence of CD4+ T cells, TCF-1+ CD8+ TRM failed to expand and differentiate into terminal effectors. Similarly, in human demyelinating CNS autoimmune lesions, we found CD8+ T cells expressing TCF-1 that predominantly exhibited a TRM-like phenotype. Together, our study provides evidence for CD8+ TRM-driven CNS immunopathology and sheds light on why inflammatory processes may evade current immunomodulatory treatments in chronic autoimmune CNS conditions.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Autoantigens , CD4-Positive T-Lymphocytes , Central Nervous System , Humans , Inflammation , Lymphocytic choriomeningitis virus
3.
Cell Metab ; 33(11): 2231-2246.e8, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34687652

ABSTRACT

Autoimmunity is energetically costly, but the impact of a metabolically active state on immunity and immune-mediated diseases is unclear. Ly6Chi monocytes are key effectors in CNS autoimmunity with an elusive role in priming naive autoreactive T cells. Here, we provide unbiased analysis of the immune changes in various compartments during cold exposure and show that this energetically costly stimulus markedly ameliorates active experimental autoimmune encephalomyelitis (EAE). Cold exposure decreases MHCII on monocytes at steady state and in various inflammatory mouse models and suppresses T cell priming and pathogenicity through the modulation of monocytes. Genetic or antibody-mediated monocyte depletion or adoptive transfer of Th1- or Th17-polarized cells for EAE abolishes the cold-induced effects on T cells or EAE, respectively. These findings provide a mechanistic link between environmental temperature and neuroinflammation and suggest competition between cold-induced metabolic adaptations and autoimmunity as energetic trade-off beneficial for the immune-mediated diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Neuroinflammatory Diseases , Adoptive Transfer , Animals , Autoimmunity , Mice , Mice, Inbred C57BL , Th17 Cells
4.
Sci Transl Med ; 11(498)2019 06 26.
Article in English | MEDLINE | ID: mdl-31243152

ABSTRACT

Epidemiological studies associate viral infections during childhood with the risk of developing autoimmune disease during adulthood. However, the mechanistic link between these events remains elusive. We report that transient viral infection of the brain in early life, but not at a later age, precipitates brain autoimmune disease elicited by adoptive transfer of myelin-specific CD4+ T cells at sites of previous infection in adult mice. Early-life infection of mouse brains imprinted a chronic inflammatory signature that consisted of brain-resident memory T cells expressing the chemokine (C-C motif) ligand 5 (CCL5). Blockade of CCL5 signaling via C-C chemokine receptor type 5 prevented the formation of brain lesions in a mouse model of autoimmune disease. In mouse and human brain, CCL5+ TRM were located predominantly to sites of microglial activation. This study uncovers how transient brain viral infections in a critical window in life might leave persisting chemotactic cues and create a long-lived permissive environment for autoimmunity.


Subject(s)
Autoimmune Diseases/immunology , Brain/immunology , Immunologic Memory , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , Chemokine CCL5/metabolism , Disease Susceptibility , HLA-DR Antigens/metabolism , Humans , Lymphocytic choriomeningitis virus/immunology , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology
6.
Front Immunol ; 9: 2827, 2018.
Article in English | MEDLINE | ID: mdl-30555489

ABSTRACT

Tissue-resident-memory CD8+ T cells (TRM) have been described as a non-circulating memory T cell subset that persists at sites of previous infection. While TRM in all non-lymphoid organs probably share a core signature differentiation pathway, certain aspects of their maintenance and effector functions may vary. It is well-established that TRM provide long-lived protective immunity through immediate effector function and accelerated recruitment of circulating immune cells. Besides immune defense against pathogens, other immunological roles of TRM are less well-studied. Likewise, evidence of a putative detrimental role of TRM for inflammatory diseases is only beginning to emerge. In this review, we discuss the protective and harmful role of TRM in organ-specific immunity and immunopathology as well as prospective implications for immunomodulatory therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Animals , Humans , Inflammation/immunology , Organ Specificity/immunology
7.
Cell ; 175(2): 458-471.e19, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30173917

ABSTRACT

Inflammatory disorders of the CNS are frequently accompanied by synaptic loss, which is thought to involve phagocytic microglia and complement components. However, the mechanisms accounting for aberrant synaptic connectivity in the context of CD8+ T cell-driven neuronal damage are poorly understood. Here, we profiled the neuronal translatome in a murine model of encephalitis caused by CD8+ T cells targeting antigenic neurons. Neuronal STAT1 signaling and downstream CCL2 expression were essential for apposition of phagocytes, ensuing synaptic loss and neurological disease. Analogous observations were made in the brains of Rasmussen's encephalitis patients. In this devastating CD8+ T cell-driven autoimmune disease, neuronal STAT1 phosphorylation and CCL2 expression co-clustered with infiltrating CD8+ T cells as well as phagocytes. Taken together, our findings uncover an active role of neurons in coordinating phagocyte-mediated synaptic loss and highlight neuronal STAT1 and CCL2 as critical steps in this process that are amenable to pharmacological interventions.


Subject(s)
Neurons/metabolism , Phagocytosis/physiology , Synapses/physiology , Animals , Brain/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/physiology , Disease Models, Animal , Encephalitis/genetics , Encephalitis/immunology , Encephalitis/physiopathology , Female , Humans , Inflammation/immunology , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Nervous System Diseases/metabolism , Neurons/physiology , Phagocytes/immunology , Phagocytes/metabolism , Phagocytosis/immunology , Phosphorylation , STAT1 Transcription Factor/physiology , Transcriptome/genetics
8.
Immunity ; 48(5): 937-950.e8, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768177

ABSTRACT

Infections are thought to trigger CD8+ cytotoxic T lymphocyte (CTL) responses during autoimmunity. However, the transcriptional programs governing the tissue-destructive potential of CTLs remain poorly defined. In a model of central nervous system (CNS) inflammation, we found that infection with lymphocytic choriomeningitis virus (LCMV), but not Listeria monocytogenes (Lm), drove autoimmunity. The DNA-binding factor TOX was induced in CTLs during LCMV infection and was essential for their encephalitogenic properties, and its expression was inhibited by interleukin-12 during Lm infection. TOX repressed the activity of several transcription factors (including Id2, TCF-1, and Notch) that are known to drive CTL differentiation. TOX also reduced immune checkpoint sensitivity by restraining the expression of the inhibitory checkpoint receptor CD244 on the surface of CTLs, leading to increased CTL-mediated damage in the CNS. Our results identify TOX as a transcriptional regulator of tissue-destructive CTLs in autoimmunity, offering a potential mechanistic link to microbial triggers.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Homeodomain Proteins/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Adult , Aged , Animals , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Signaling Lymphocytic Activation Molecule Family/immunology , T-Lymphocytes, Cytotoxic/immunology
9.
J Exp Med ; 213(8): 1571-87, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27377586

ABSTRACT

Tissue-resident memory T cells (TRM) persist at sites of prior infection and have been shown to enhance pathogen clearance by recruiting circulating immune cells and providing bystander activation. Here, we characterize the functioning of brain-resident memory T cells (bTRM) in an animal model of viral infection. bTRM were subject to spontaneous homeostatic proliferation and were largely refractory to systemic immune cell depletion. After viral reinfection in mice, bTRM rapidly acquired cytotoxic effector function and prevented fatal brain infection, even in the absence of circulating CD8(+) memory T cells. Presentation of cognate antigen on MHC-I was essential for bTRM-mediated protective immunity, which involved perforin- and IFN-γ-dependent effector mechanisms. These findings identify bTRM as an organ-autonomous defense system serving as a paradigm for TRM functioning as a self-sufficient first line of adaptive immunity.


Subject(s)
Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Brain/pathology , Brain/virology , CD8-Positive T-Lymphocytes/pathology , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Knockout
10.
Cell Rep ; 15(5): 1076-1087, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27117419

ABSTRACT

The macroautophagy machinery has been implicated in MHC class II restricted antigen presentation. Here, we report that this machinery assists in the internalization of MHC class I molecules. In the absence of the autophagy factors Atg5 and Atg7, MHC class I surface levels are elevated due to decreased endocytosis and degradation. Internalization of MHC class I molecules occurs less efficiently if AAK1 cannot be recruited via Atg8/LC3B. In the absence of Atg-dependent MHC class I internalization, dendritic cells stimulate CD8(+) T cell responses more efficiently in vitro and in vivo. During viral infections, lack of Atg5 results in enhanced influenza- and LCMV-specific CD8(+) T cell responses in vivo. Elevated influenza-specific CD8(+) T cell responses are associated with better immune control of this infection. Thus, the macroautophagy machinery orchestrates T cell immunity by supporting MHC class II but compromises MHC class I restricted antigen presentation.


Subject(s)
Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 7/genetics , Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , Influenza A Virus, H1N1 Subtype/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Antigen Presentation/immunology , Cells, Cultured , Endocytosis/immunology , Histocompatibility Antigens Class II/immunology , Humans , Lymphocyte Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
J Autoimmun ; 67: 8-18, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26341385

ABSTRACT

Plasmacytoid dendritic cells (pDCs) exhibit both innate and adaptive functions. In particular they are the main source of type I IFNs and directly impact T cell responses through antigen presentation. We have previously demonstrated that during experimental autoimmune encephalomyelitis (EAE) initiation, myelin-antigen presentation by pDCs is associated with suppressive Treg development and results in attenuated EAE. Here, we show that pDCs transferred during acute disease phase confer recovery from EAE. Clinical improvement is associated with migration of injected pDCs into inflamed CNS and is dependent on the subsequent and selective chemerin-mediated recruitment of endogenous pDCs to the CNS. The protective effect requires pDC pre-loading with myelin antigen, and is associated with the modulation of CNS-infiltrating pDC phenotype and inhibition of CNS encephalitogenic T cells. This study may pave the way for novel pDC-based cell therapies in autoimmune diseases, aiming at specifically modulating pathogenic cells that induce and sustain autoimmune inflammation.


Subject(s)
Adoptive Transfer , Chemotaxis/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Animals , Autoantigens/immunology , Cell- and Tissue-Based Therapy , Chemokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Myelin Sheath/immunology , Receptors, Chemokine , Receptors, G-Protein-Coupled/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology
12.
Methods Mol Biol ; 1304: 189-209, 2016.
Article in English | MEDLINE | ID: mdl-25146304

ABSTRACT

Neuropathological techniques such as conventional and immunohistochemical staining of paraffin-embedded tissue sections are instrumental for identification and characterization of aberrations of organ architecture during human inflammatory disorders of the central nervous system (CNS) as in their animal models. Here we describe step-by-step protocols for tissue processing, sectioning, and conventional and immunohistochemical stainings to display as well as quantify CNS inflammation, demyelination, and neuronal damage in experimental autoimmune encephalomyelitis (EAE), an animal model of Multiple Sclerosis (MS).


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Animals , Central Nervous System/pathology , Disease Models, Animal , Immunohistochemistry/methods , Mice , Multiple Sclerosis/pathology
13.
Brain ; 138(Pt 11): 3263-74, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26359290

ABSTRACT

Recent association studies have linked numerous genetic variants with an increased risk for multiple sclerosis, although their functional relevance remains largely unknown. Here we investigated phenotypical and functional consequences of a genetic variant in the CD226 gene that, among other autoimmune diseases, predisposes to multiple sclerosis. Phenotypically, effector and regulatory CD4(+) memory T cells of healthy individuals carrying the predisposing CD226 genetic variant showed, in comparison to carriers of the protective variant, reduced surface expression of CD226 and an impaired induction of CD226 after stimulation. This haplotype-dependent reduction in CD226 expression on memory T cells was abrogated in patients with multiple sclerosis, as CD226 expression was comparable to healthy risk haplotype carriers irrespective of genetic variant. Functionally, FOXP3-positive regulatory T cells from healthy carriers of the genetic protective variant showed superior suppressive capacity, which was again abrogated in multiple sclerosis patients. Mimicking the phenotype of human CD226 genetic risk variant carriers, regulatory T cells derived from Cd226-deficient mice showed similarly reduced inhibitory activity, eventually resulting in an exacerbated disease course of experimental autoimmune encephalomyelitis, the animal model of multiple sclerosis. Therefore, by combining human and mouse analyses we show that CD226 exhibits an important role in the activation of regulatory T cells, with its genetically imposed dysregulation impairing regulatory T cell function.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/genetics , Multiple Sclerosis/genetics , RNA, Messenger/metabolism , T-Lymphocytes, Regulatory/immunology , Adult , Animals , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Forkhead Transcription Factors/metabolism , Genetic Predisposition to Disease , Haplotypes , Heterozygote , Humans , Male , Mice , Mice, Knockout , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Phenotype , Polymorphism, Single Nucleotide , T-Lymphocytes, Regulatory/metabolism
14.
J Neuroimmunol ; 274(1-2): 111-24, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25086877

ABSTRACT

The 2-lysophosphatidylcholine analog edelfosine induces apoptosis in highly proliferating cells, e.g. activated immune cells. We examined mechanisms of action of edelfosine on immune functions in experimental autoimmune encephalomyelitis, a well-accepted animal model for multiple sclerosis. We observed activated caspase-3 expression in lymphoid organs and the central nervous system; however, edelfosine did not induce global apoptosis. Edelfosine improved the disease course and led to reduced frequencies of CD4(+) T cells infiltrating into the central nervous system. Our data suggest edelfosine as an interesting treatment candidate for multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunologic Factors/immunology , Phospholipid Ethers/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Phosphodiesterase Inhibitors/immunology , Phosphodiesterase Inhibitors/pharmacology , Phospholipid Ethers/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
15.
Eur J Immunol ; 44(10): 3119-28, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25043505

ABSTRACT

Recent findings indicate a pathogenic involvement of IL-17-producing CD8(+) T cells in multiple sclerosis (MS). IL-17 production has been attributed to a subset of CD8(+) T cells that belong to the mucosal-associated invariant T (MAIT) cell population. Here, we report a reduction of CD8(+) MAIT cells in the blood of MS patients compared with healthy individuals, which significantly correlated with IL-18 serum levels in MS patients. In vitro stimulation of peripheral blood mononuclear cells from healthy individuals and MS patients with IL-18 specifically activated CD8(+) MAIT cells. Moreover, IL-18 together with T-cell receptor stimulation induced, specifically on CD8(+) MAIT cells, an upregulation of the integrin very late antigen-4 that is essential for the infiltration of CD8(+) T cells into the CNS. Notably, we were able to identify CD8(+) MAIT cells in MS brain lesions by immunohistochemistry while they were almost absent in the cerebrospinal fluid (CSF). In summary, our findings indicate that an IL-18-driven activation of CD8(+) MAIT cells contributes to their CNS infiltration in MS, in turn leading to reduced CD8(+) MAIT-cell frequencies in the blood. Therefore, CD8(+) MAIT cells seem to play a role in the innate arm of immunopathology in MS.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukin-18/blood , Multiple Sclerosis/immunology , T-Lymphocyte Subsets/immunology , Chemotaxis, Leukocyte , Flow Cytometry , Humans , Immunohistochemistry , Lymphocyte Activation/immunology , Multiple Sclerosis/blood , Multiple Sclerosis/pathology
16.
Acta Neuropathol Commun ; 1: 78, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24299111

ABSTRACT

BACKGROUND: In human and animal prion diseases, pathological prion protein, PrPSc, as well as prion infectivity is mainly found in the central nervous system, but also in lymphoid organs and muscle. Pathophysiology of prion colonization of lymphoid organs has been studied intensively, yet how myositis influences prion accumulation in muscle is unknown. RESULT: We have investigated the influence of myositis on PrPSc accumulation and prion infectivity in two distinct mouse models of experimental autoimmune myositis. Furthermore, we have addressed the relevance of PrPC expression in the lymphoreticular system in myositis by generating bone marrow chimeras.Here we show that myositis positively influences muscular PrPSc accumulation at preclinical time points and that PrPC-expression in the lymphoid system is critical for this. In muscle, PrPSc and prion infectivity are uncoupled with detectable PrPSc but no prion infectivity at preclinical time points. Muscle has an intrinsically high ability to clear PrPSc once myositis has ceased, possibly involving autophagy. CONCLUSION: Our findings provide new insights into the pathophysiology of prion colonization in muscle pointing out that myositis leads to enhanced prion colonization of muscle in subclinical prion disease.


Subject(s)
Muscles/metabolism , Nervous System Autoimmune Disease, Experimental/metabolism , PrPSc Proteins/metabolism , Animals , Blotting, Western , Disease Progression , Immunohistochemistry , Mice, Inbred C57BL , Mice, Knockout , Prion Proteins , Prions/genetics , Prions/metabolism , Radiation Chimera , Time Factors
17.
J Exp Med ; 210(10): 2087-103, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-23999498

ABSTRACT

Neurons are postmitotic and thus irreplaceable cells of the central nervous system (CNS). Accordingly, CNS inflammation with resulting neuronal damage can have devastating consequences. We investigated molecular mediators and structural consequences of CD8(+) T lymphocyte (CTL) attack on neurons in vivo. In a viral encephalitis model in mice, disease depended on CTL-derived interferon-γ (IFN-γ) and neuronal IFN-γ signaling. Downstream STAT1 phosphorylation and nuclear translocation in neurons were associated with dendrite and synapse loss (deafferentation). Analogous molecular and structural alterations were also found in human Rasmussen encephalitis, a CTL-mediated human autoimmune disorder of the CNS. Importantly, therapeutic intervention by IFN-γ blocking antibody prevented neuronal deafferentation and clinical disease without reducing CTL responses or CNS infiltration. These findings identify neuronal IFN-γ signaling as a novel target for neuroprotective interventions in CTL-mediated CNS disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendrites/immunology , Interferon-gamma/metabolism , Neurons/immunology , Synapses/immunology , Adolescent , Adult , Animals , Cell Nucleus/metabolism , Child , Humans , Interferon-gamma/antagonists & inhibitors , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/virology , Perforin/genetics , Perforin/metabolism , Phosphorylation , Protein Transport , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , T-Lymphocytes, Cytotoxic/immunology , Young Adult , fas Receptor/genetics , fas Receptor/metabolism , Interferon gamma Receptor
18.
J Immunol ; 191(9): 4531-9, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24062488

ABSTRACT

Multiple sclerosis is considered to be initiated by a deregulated, myelin-specific T cell response. However, the formation of inflammatory CNS lesions and the contribution of different leukocyte subsets in setting up these lesions are still incompletely understood. In this study, we show that, in the mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis, neutrophil granulocytes are important contributors in preparing CNS inflammation. Preclinical single-dose Ab-mediated depletion of neutrophils delayed the onset and continuous depletion attenuated the development of experimental autoimmune encephalomyelitis, whereas the generation of a myelin-specific T cell response remained unaffected. Neutrophil-related enzymes such as myeloperoxidase and neutrophil elastase did not contribute in mounting CNS inflammation, as analyzed by using respective knockout mice and inhibitors. CNS-infiltrating neutrophils secreted proinflammatory molecules and matured bone marrow-derived dendritic cells in vitro, which in turn enhanced their ability to restimulate myelin-specific T cells. This was mirrored in vivo, in which depletion of neutrophils specifically impaired maturation of microglia and macrophages into professional APCs, resulting in a diminished amplification of early CNS inflammation. Therefore, inside the CNS neutrophils provide local cofactors that are required for the maturation of myeloid cells into professional APCs representing an essential step for the local restimulation of myelin-specific T cells and the development of autoimmune disease.


Subject(s)
Antigen-Presenting Cells/immunology , Central Nervous System/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Neutrophils/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation/immunology , Autoimmunity/immunology , Cells, Cultured , Cytokines/metabolism , Inflammation/immunology , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Multiple Sclerosis/immunology , Peroxidase/genetics , Peroxidase/metabolism
19.
Nat Med ; 18(12): 1805-11, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160238

ABSTRACT

In multiple sclerosis, an inflammatory disease of the central nervous system (CNS), axonal and neuronal loss are major causes for irreversible neurological disability. However, which molecules contribute to axonal and neuronal injury under inflammatory conditions remains largely unknown. Here we show that the transient receptor potential melastatin 4 (TRPM4) cation channel is crucial in this process. TRPM4 is expressed in mouse and human neuronal somata, but it is also expressed in axons in inflammatory CNS lesions in experimental autoimmune encephalomyelitis (EAE) in mice and in human multiple sclerosis tissue. Deficiency or pharmacological inhibition of TRPM4 using the antidiabetic drug glibenclamide resulted in reduced axonal and neuronal degeneration and attenuated clinical disease scores in EAE, but this occurred without altering EAE-relevant immune function. Furthermore, Trpm4(-/-) mouse neurons were protected against inflammatory effector mechanisms such as excitotoxic stress and energy deficiency in vitro. Electrophysiological recordings revealed TRPM4-dependent neuronal ion influx and oncotic cell swelling upon excitotoxic stimulation. Therefore, interference with TRPM4 could translate into a new neuroprotective treatment strategy.


Subject(s)
Axons/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Multiple Sclerosis/metabolism , Nerve Degeneration/metabolism , TRPM Cation Channels/metabolism , Analysis of Variance , Animals , Axons/drug effects , Blotting, Western , Cell Proliferation/drug effects , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glyburide/pharmacology , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Patch-Clamp Techniques , Real-Time Polymerase Chain Reaction , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics
20.
Blood ; 120(18): 3793-802, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22976954

ABSTRACT

The devastating effect of ischemic stroke is attenuated in mice lacking conventional and unconventional T cells, suggesting that inflammation enhances tissue damage in cerebral ischemia. We explored the functional role of αß and γδ T cells in a murine model of stroke and distinguished 2 different T cell-dependent proinflammatory pathways in ischemia-reperfusion injury. IFN-γ produced by CD4(+) T cells induced TNF-α production in macrophages, whereas IL-17A secreted by γδ T cells led to neutrophil recruitment. The synergistic effect of TNF-α and IL-17A on astrocytes resulted in enhanced secretion of CXCL-1, a neutrophil chemoattractant. Application of an IL-17A-blocking antibody within 3 hours after stroke induction decreased infarct size and improved neurologic outcome in the murine model. In autoptic brain tissue of patients who had a stroke, we detected IL-17A-positive lymphocytes, suggesting that this aspect of the inflammatory cascade is also relevant in the human brain. We propose that selective targeting of IL-17A signaling might provide a new therapeutic option for the treatment of stroke.


Subject(s)
Interleukin-17/immunology , Neutrophil Infiltration/immunology , Signal Transduction/immunology , Stroke/immunology , T-Lymphocytes/immunology , Animals , Brain Ischemia/immunology , Brain Ischemia/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunohistochemistry , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophils/immunology , Real-Time Polymerase Chain Reaction , Stroke/metabolism , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...