Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
Cell ; 187(15): 4043-4060.e30, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38878778

ABSTRACT

Inflammation-induced neurodegeneration is a defining feature of multiple sclerosis (MS), yet the underlying mechanisms remain unclear. By dissecting the neuronal inflammatory stress response, we discovered that neurons in MS and its mouse model induce the stimulator of interferon genes (STING). However, activation of neuronal STING requires its detachment from the stromal interaction molecule 1 (STIM1), a process triggered by glutamate excitotoxicity. This detachment initiates non-canonical STING signaling, which leads to autophagic degradation of glutathione peroxidase 4 (GPX4), essential for neuronal redox homeostasis and thereby inducing ferroptosis. Both genetic and pharmacological interventions that target STING in neurons protect against inflammation-induced neurodegeneration. Our findings position STING as a central regulator of the detrimental neuronal inflammatory stress response, integrating inflammation with glutamate signaling to cause neuronal cell death, and present it as a tractable target for treating neurodegeneration in MS.


Subject(s)
Inflammation , Membrane Proteins , Multiple Sclerosis , Neurons , Animals , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Membrane Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Mice , Humans , Inflammation/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Signal Transduction , Autophagy , Mice, Inbred C57BL , Glutamic Acid/metabolism , Ferroptosis , Disease Models, Animal , Female , Male
2.
Cell ; 185(4): 585-602.e29, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35051368

ABSTRACT

The relevance of extracellular magnesium in cellular immunity remains largely unknown. Here, we show that the co-stimulatory cell-surface molecule LFA-1 requires magnesium to adopt its active conformation on CD8+ T cells, thereby augmenting calcium flux, signal transduction, metabolic reprogramming, immune synapse formation, and, as a consequence, specific cytotoxicity. Accordingly, magnesium-sufficiency sensed via LFA-1 translated to the superior performance of pathogen- and tumor-specific T cells, enhanced effectiveness of bi-specific T cell engaging antibodies, and improved CAR T cell function. Clinically, low serum magnesium levels were associated with more rapid disease progression and shorter overall survival in CAR T cell and immune checkpoint antibody-treated patients. LFA-1 thus directly incorporates information on the composition of the microenvironment as a determinant of outside-in signaling activity. These findings conceptually link co-stimulation and nutrient sensing and point to the magnesium-LFA-1 axis as a therapeutically amenable biologic system.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Magnesium/metabolism , Animals , Bacterial Infections/immunology , Caloric Restriction , Cell Line, Tumor , Cytotoxicity, Immunologic , HEK293 Cells , Humans , Immunologic Memory , Immunological Synapses/metabolism , Immunotherapy , Lymphocyte Activation/immunology , MAP Kinase Signaling System , Magnesium/administration & dosage , Male , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism
3.
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
4.
Immunity ; 56(4): 813-828.e10, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36809763

ABSTRACT

T cell factor 1 (Tcf-1) expressing CD8+ T cells exhibit stem-like self-renewing capacity, rendering them key for immune defense against chronic viral infection and cancer. Yet, the signals that promote the formation and maintenance of these stem-like CD8+ T cells (CD8+SL) remain poorly defined. Studying CD8+ T cell differentiation in mice with chronic viral infection, we identified the alarmin interleukin-33 (IL-33) as pivotal for the expansion and stem-like functioning of CD8+SL as well as for virus control. IL-33 receptor (ST2)-deficient CD8+ T cells exhibited biased end differentiation and premature loss of Tcf-1. ST2-deficient CD8+SL responses were restored by blockade of type I interferon signaling, suggesting that IL-33 balances IFN-I effects to control CD8+SL formation in chronic infection. IL-33 signals broadly augmented chromatin accessibility in CD8+SL and determined these cells' re-expansion potential. Our study identifies the IL-33-ST2 axis as an important CD8+SL-promoting pathway in the context of chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-33 , Lymphocytic Choriomeningitis , Animals , Mice , Alarmins/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus , Mice, Inbred C57BL , Persistent Infection , T Cell Transcription Factor 1/metabolism
5.
Nat Immunol ; 20(6): 701-710, 2019 06.
Article in English | MEDLINE | ID: mdl-31110314

ABSTRACT

Cachexia represents a leading cause of morbidity and mortality in various cancers, chronic inflammation and infections. Understanding of the mechanisms that drive cachexia has remained limited, especially for infection-associated cachexia (IAC). In the present paper we describe a model of reversible cachexia in mice with chronic viral infection and identify an essential role for CD8+ T cells in IAC. Cytokines linked to cancer-associated cachexia did not contribute to IAC. Instead, virus-specific CD8+ T cells caused morphologic and molecular changes in the adipose tissue, which led to depletion of lipid stores. These changes occurred at a time point that preceded the peak of the CD8+ T cell response and required T cell-intrinsic type I interferon signaling and antigen-specific priming. Our results link systemic antiviral immune responses to adipose-tissue remodeling and reveal an underappreciated role of CD8+ T cells in IAC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cachexia/etiology , Virus Diseases/complications , Virus Diseases/immunology , Adipose Tissue/diagnostic imaging , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adipose Tissue/virology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cachexia/diagnostic imaging , Cachexia/metabolism , Cachexia/pathology , Chronic Disease , Cytokines/blood , Cytokines/metabolism , Female , Interferon Type I/metabolism , Lipid Metabolism , Lipolysis , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus , Magnetic Resonance Imaging/methods , Male , Mice , Signal Transduction , Virus Diseases/virology
6.
Nat Immunol ; 19(12): 1341-1351, 2018 12.
Article in English | MEDLINE | ID: mdl-30374128

ABSTRACT

Polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) have been characterized in the context of malignancies. Here we show that PMN-MDSCs can restrain B cell accumulation during central nervous system (CNS) autoimmunity. Ly6G+ cells were recruited to the CNS during experimental autoimmune encephalomyelitis (EAE), interacted with B cells that produced the cytokines GM-CSF and interleukin-6 (IL-6), and acquired properties of PMN-MDSCs in the CNS in a manner dependent on the signal transducer STAT3. Depletion of Ly6G+ cells or dysfunction of Ly6G+ cells through conditional ablation of STAT3 led to the selective accumulation of GM-CSF-producing B cells in the CNS compartment, which in turn promoted an activated microglial phenotype and lack of recovery from EAE. The frequency of CD138+ B cells in the cerebrospinal fluid (CSF) of human subjects with multiple sclerosis was negatively correlated with the frequency of PMN-MDSCs in the CSF. Thus PMN-MDSCs might selectively control the accumulation and cytokine secretion of B cells in the inflamed CNS.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Myeloid-Derived Suppressor Cells/immunology , Adolescent , Adult , Animals , Central Nervous System/immunology , Female , Humans , Male , Mice , Middle Aged , Young Adult
7.
Nature ; 627(8003): 407-415, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38383779

ABSTRACT

Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.


Subject(s)
Aquaporin 4 , Autoantibodies , Autoantigens , B-Lymphocytes , Immune Tolerance , Neuromyelitis Optica , Animals , Humans , Mice , AIRE Protein , Aquaporin 4/deficiency , Aquaporin 4/genetics , Aquaporin 4/immunology , Aquaporin 4/metabolism , Autoantibodies/immunology , Autoantigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD40 Antigens/immunology , Germinal Center/cytology , Germinal Center/immunology , Neuromyelitis Optica/immunology , Neuromyelitis Optica/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thyroid Epithelial Cells/immunology , Thyroid Epithelial Cells/metabolism , Transcriptome
8.
Cell ; 156(1-2): 277-90, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439382

ABSTRACT

Central nervous system myelin is a multilayered membrane sheath generated by oligodendrocytes for rapid impulse propagation. However, the underlying mechanisms of myelin wrapping have remained unclear. Using an integrative approach of live imaging, electron microscopy, and genetics, we show that new myelin membranes are incorporated adjacent to the axon at the innermost tongue. Simultaneously, newly formed layers extend laterally, ultimately leading to the formation of a set of closely apposed paranodal loops. An elaborated system of cytoplasmic channels within the growing myelin sheath enables membrane trafficking to the leading edge. Most of these channels close with ongoing development but can be reopened in adults by experimentally raising phosphatidylinositol-(3,4,5)-triphosphate levels, which reinitiates myelin growth. Our model can explain assembly of myelin as a multilayered structure, abnormal myelin outfoldings in neurological disease, and plasticity of myelin biogenesis observed in adult life.


Subject(s)
Axons/metabolism , Myelin Sheath/metabolism , Animals , Cells, Cultured , Central Nervous System/metabolism , Mice , Neuroglia/metabolism , Oligodendroglia/metabolism , Zebrafish
9.
Nat Immunol ; 16(1): 67-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25419628

ABSTRACT

Immune responses are tightly regulated to ensure efficient pathogen clearance while avoiding tissue damage. Here we report that Setdb2 was the only protein lysine methyltransferase induced during infection with influenza virus. Setdb2 expression depended on signaling via type I interferons, and Setdb2 repressed expression of the gene encoding the neutrophil attractant CXCL1 and other genes that are targets of the transcription factor NF-κB. This coincided with occupancy by Setdb2 at the Cxcl1 promoter, which in the absence of Setdb2 displayed diminished trimethylation of histone H3 Lys9 (H3K9me3). Mice with a hypomorphic gene-trap construct of Setdb2 exhibited increased infiltration of neutrophils during sterile lung inflammation and were less sensitive to bacterial superinfection after infection with influenza virus. This suggested that a Setdb2-mediated regulatory crosstalk between the type I interferons and NF-κB pathways represents an important mechanism for virus-induced susceptibility to bacterial superinfection.


Subject(s)
Histone-Lysine N-Methyltransferase/immunology , NF-kappa B/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Pneumonia/immunology , Superinfection/immunology , Animals , Chemokine CXCL1/immunology , Disease Susceptibility , Female , Interferon Type I/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/virology , Pneumonia/enzymology , Pneumonia/virology , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Specific Pathogen-Free Organisms , Superinfection/enzymology , Superinfection/microbiology
10.
Immunity ; 48(2): 380-395.e6, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29426702

ABSTRACT

Individual reports suggest that the central nervous system (CNS) contains multiple immune cell types with diverse roles in tissue homeostasis, immune defense, and neurological diseases. It has been challenging to map leukocytes across the entire brain, and in particular in pathology, where phenotypic changes and influx of blood-derived cells prevent a clear distinction between reactive leukocyte populations. Here, we applied high-dimensional single-cell mass and fluorescence cytometry, in parallel with genetic fate mapping systems, to identify, locate, and characterize multiple distinct immune populations within the mammalian CNS. Using this approach, we revealed that microglia, several subsets of border-associated macrophages and dendritic cells coexist in the CNS at steady state and exhibit disease-specific transformations in the immune microenvironment during aging and in models of Alzheimer's disease and multiple sclerosis. Together, these data and the described framework provide a resource for the study of disease mechanisms, potential biomarkers, and therapeutic targets in CNS disease.


Subject(s)
Aging/immunology , Central Nervous System/immunology , Leukocytes/immunology , Macrophages/immunology , Animals , Dendritic Cells/immunology , Mice , Mice, Inbred C57BL , Microglia/immunology , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/immunology , Single-Cell Analysis
11.
Immunity ; 48(3): 514-529.e6, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29548672

ABSTRACT

Microglia as tissue macrophages contribute to the defense and maintenance of central nervous system (CNS) homeostasis. Little is known about the epigenetic signals controlling microglia function in vivo. We employed constitutive and inducible mutagenesis in microglia to delete two class I histone deacetylases, Hdac1 and Hdac2. Prenatal ablation of Hdac1 and Hdac2 impaired microglial development. Mechanistically, the promoters of pro-apoptotic and cell cycle genes were hyperacetylated in absence of Hdac1 and Hdac2, leading to increased apoptosis and reduced survival. In contrast, Hdac1 and Hdac2 were not required for adult microglia survival during homeostasis. In a mouse model of Alzheimer's disease, deletion of Hdac1 and Hdac2 in microglia, but not in neuroectodermal cells, resulted in a decrease in amyloid load and improved cognitive impairment by enhancing microglial amyloid phagocytosis. Collectively, we report a role for epigenetic factors that differentially affect microglia development, homeostasis, and disease that could potentially be utilized therapeutically.


Subject(s)
Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Homeostasis , Microglia/immunology , Microglia/metabolism , Neurodegenerative Diseases/genetics , Neurogenesis/genetics , Animals , Apoptosis , Cell Proliferation , Disease Models, Animal , Epigenesis, Genetic , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Histones/metabolism , Memory Disorders/genetics , Memory Disorders/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/psychology , Neurogenesis/immunology , Phagocytosis/immunology , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Spatial Learning , Transcriptome
12.
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
13.
EMBO J ; 41(19): e108536, 2022 10 04.
Article in English | MEDLINE | ID: mdl-35924455

ABSTRACT

During development, hematopoietic stem cells (HSCs) are produced from the hemogenic endothelium and will expand in a transient hematopoietic niche. Prostaglandin E2 (PGE2) is essential during vertebrate development and HSC specification, but its precise source in the embryo remains elusive. Here, we show that in the zebrafish embryo, PGE2 synthesis genes are expressed by distinct stromal cell populations, myeloid (neutrophils, macrophages), and endothelial cells of the caudal hematopoietic tissue. Ablation of myeloid cells, which produce the PGE2 precursor prostaglandin H2 (PGH2), results in loss of HSCs in the caudal hematopoietic tissue, which could be rescued by exogeneous PGE2 or PGH2 supplementation. Endothelial cells contribute by expressing the PGH2 import transporter slco2b1 and ptges3, the enzyme converting PGH2 into PGE2. Of note, differential niche cell expression of PGE2 biosynthesis enzymes is also observed in the mouse fetal liver. Taken altogether, our data suggest that the triad composed of neutrophils, macrophages, and endothelial cells sequentially and synergistically contributes to blood stem cell expansion during vertebrate development.


Subject(s)
Hemangioblasts , Zebrafish , Animals , Dinoprostone/metabolism , Hematopoietic Stem Cells/metabolism , Mice , Prostaglandin H2/metabolism
14.
Blood ; 141(14): 1755-1767, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36574344

ABSTRACT

CD4+FOXP3+ regulatory T cells (Tregs) have demonstrated efficacy in the prevention and treatment of graft-versus-host disease (GVHD). Preclinical and clinical studies indicate that Tregs are able to protect from GVHD without interfering with the graft-versus-tumor (GVT) effect of hematopoietic cell transplantation (HCT), although the underlying molecular mechanisms are largely unknown. To elucidate Treg suppressive function during in vivo suppression of acute GVHD, we performed paired T-cell receptor (TCRα and ΤCRß genes) repertoire sequencing and RNA sequencing analysis on conventional T cells (Tcons) and Tregs before and after transplantation in a major histocompatibility complex -mismatched mouse model of HCT. We show that both Tregs and Tcons underwent clonal restriction, and Tregs did not interfere with the activation of alloreactive Tcon clones and the breadth of their TCR repertoire but markedly suppressed their expansion. Transcriptomic analysis revealed that Tregs predominantly affected the transcriptome of CD4 Tcons and, to a lesser extent, that of CD8 Tcons, thus modulating the transcription of genes encoding pro- and anti-inflammatory molecules as well as enzymes involved in metabolic processes, inducing a switch from glycolysis to oxidative phosphorylation. Finally, Tregs did not interfere with the induction of gene sets involved in the GVT effect. Our results shed light onto the mechanisms of acute GVHD suppression by Tregs and will support the clinical translation of this immunoregulatory approach.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Animals , Mice , T-Lymphocytes, Regulatory/pathology , Transcriptome , Graft vs Host Disease/genetics , Graft vs Host Disease/prevention & control , Graft vs Host Disease/pathology , Proteins/genetics
15.
EMBO Rep ; 24(3): e55328, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36715148

ABSTRACT

The vasculature is a key regulator of leukocyte trafficking into the central nervous system (CNS) during inflammatory diseases including multiple sclerosis (MS). However, the impact of endothelial-derived factors on CNS immune responses remains unknown. Bioactive lipids, in particular oxysterols downstream of Cholesterol-25-hydroxylase (Ch25h), promote neuroinflammation but their functions in the CNS are not well-understood. Using floxed-reporter Ch25h knock-in mice, we trace Ch25h expression to CNS endothelial cells (ECs) and myeloid cells and demonstrate that Ch25h ablation specifically from ECs attenuates experimental autoimmune encephalomyelitis (EAE). Mechanistically, inflamed Ch25h-deficient CNS ECs display altered lipid metabolism favoring polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) expansion, which suppresses encephalitogenic T lymphocyte proliferation. Additionally, endothelial Ch25h-deficiency combined with immature neutrophil mobilization into the blood circulation nearly completely protects mice from EAE. Our findings reveal a central role for CNS endothelial Ch25h in promoting neuroinflammation by inhibiting the expansion of immunosuppressive myeloid cell populations.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Oxysterols , Mice , Animals , Endothelial Cells/metabolism , Oxysterols/metabolism , Neuroinflammatory Diseases , Central Nervous System/metabolism , Mice, Inbred C57BL
16.
Nature ; 571(7764): 265-269, 2019 07.
Article in English | MEDLINE | ID: mdl-31207605

ABSTRACT

Cytotoxic T cells are essential mediators of protective immunity to viral infection and malignant tumours and are a key target of immunotherapy approaches. However, prolonged exposure to cognate antigens often attenuates the effector capacity of T cells and limits their therapeutic potential1-4. This process, known as T cell exhaustion or dysfunction1, is manifested by epigenetically enforced changes in gene regulation that reduce the expression of cytokines and effector molecules and upregulate the expression of inhibitory receptors such as programmed cell-death 1 (PD-1)5-8. The underlying molecular mechanisms that induce and stabilize the phenotypic and functional features of exhausted T cells remain poorly understood9-12. Here we report that the development and maintenance of populations of exhausted T cells in mice requires the thymocyte selection-associated high mobility group box (TOX) protein13-15. TOX is induced by high antigen stimulation of the T cell receptor and correlates with the presence of an exhausted phenotype during chronic infections with lymphocytic choriomeningitis virus in mice and hepatitis C virus in humans. Removal of its DNA-binding domain reduces the expression of PD-1 at the mRNA and protein level, augments the production of cytokines and results in a more polyfunctional T cell phenotype. T cells with this deletion initially mediate increased effector function and cause more severe immunopathology, but ultimately undergo a massive decline in their quantity, notably among the subset of TCF-1+ self-renewing T cells. Altogether, we show that TOX is a critical factor for the normal progression of T cell dysfunction and the maintenance of exhausted T cells during chronic infection, and provide a link between the suppression of effector function intrinsic to CD8 T cells and protection against immunopathology.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/virology , High Mobility Group Proteins/metabolism , Homeodomain Proteins/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Animals , Cell Proliferation , Chronic Disease , Cytokines/immunology , Cytokines/metabolism , Epigenesis, Genetic , Female , Gene Expression Regulation/immunology , Hepacivirus/immunology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Immunologic Memory , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Phenotype , Thymocytes/cytology , Thymocytes/immunology , Transcription, Genetic
17.
Immunity ; 43(5): 974-86, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26588782

ABSTRACT

Tissue damage caused by viral hepatitis is a major cause of morbidity and mortality worldwide. Using a mouse model of viral hepatitis, we identified virus-induced early transcriptional changes in the redox pathways in the liver, including downregulation of superoxide dismutase 1 (Sod1). Sod1(-/-) mice exhibited increased inflammation and aggravated liver damage upon viral infection, which was independent of T and NK cells and could be ameliorated by antioxidant treatment. Type I interferon (IFN-I) led to a downregulation of Sod1 and caused oxidative liver damage in Sod1(-/-) and wild-type mice. Genetic and pharmacological ablation of the IFN-I signaling pathway protected against virus-induced liver damage. These results delineate IFN-I mediated oxidative stress as a key mediator of virus-induced liver damage and describe a mechanism of innate-immunity-driven pathology, linking IFN-I signaling with antioxidant host defense and infection-associated tissue damage. VIDEO ABSTRACT.


Subject(s)
Hepatocytes/immunology , Interferon Type I/immunology , Oxidative Stress/immunology , Superoxide Dismutase/immunology , Animals , Antioxidants/metabolism , Hepatitis, Viral, Animal/immunology , Immunity, Innate/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Liver/immunology , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Signal Transduction/immunology , Superoxide Dismutase-1 , T-Lymphocytes/immunology , Transcription, Genetic/immunology
18.
Proc Natl Acad Sci U S A ; 118(46)2021 11 16.
Article in English | MEDLINE | ID: mdl-34772811

ABSTRACT

Chronic viral infections subvert protective B cell immunity. An early type I interferon (IFN-I)-driven bias to short-lived plasmablast differentiation leads to clonal deletion, so-called "decimation," of antiviral memory B cells. Therefore, prophylactic countermeasures against decimation remain an unmet need. We show that vaccination-induced CD4 T cells prevented the decimation of naïve and memory B cells in chronically lymphocytic choriomeningitis virus (LCMV)-infected mice. Although these B cell responses were largely T independent when IFN-I was blocked, preexisting T help assured their sustainability under conditions of IFN-I-driven inflammation by instructing a germinal center B cell transcriptional program. Prevention of decimation depended on T cell-intrinsic Bcl6 and Tfh progeny formation. Antigen presentation by B cells, interactions with antigen-specific T helper cells, and costimulation by CD40 and ICOS were also required. Importantly, B cell-mediated virus control averted Th1-driven immunopathology in LCMV-challenged animals with preexisting CD4 T cell immunity. Our findings show that vaccination-induced Tfh cells represent a cornerstone of effective B cell immunity to chronic virus challenge, pointing the way toward more effective B cell-based vaccination against persistent viral diseases.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Persistent Infection/immunology , Vaccines/immunology , Virus Diseases/immunology , Animals , Antibodies, Viral/immunology , Antigen Presentation/immunology , Antiviral Agents/immunology , Cells, Cultured , Germinal Center/immunology , Inflammation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Memory B Cells/immunology , Mice , Proto-Oncogene Proteins c-bcl-6/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Vaccination/methods
19.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34782467

ABSTRACT

Nondegradative ubiquitin chains attached to specific targets via Lysine 63 (K63) residues have emerged to play a fundamental role in synaptic function. The K63-specific deubiquitinase CYLD has been widely studied in immune cells and lately also in neurons. To better understand if CYLD plays a role in brain and synapse homeostasis, we analyzed the behavioral profile of CYLD-deficient mice. We found that the loss of CYLD results in major autism-like phenotypes including impaired social communication, increased repetitive behavior, and cognitive dysfunction. Furthermore, the absence of CYLD leads to a reduction in hippocampal network excitability, long-term potentiation, and pyramidal neuron spine numbers. By providing evidence that CYLD can modulate mechanistic target of rapamycin (mTOR) signaling and autophagy at the synapse, we propose that synaptic K63-linked ubiquitination processes could be fundamental in understanding the pathomechanisms underlying autism spectrum disorder.


Subject(s)
Autophagy/physiology , Hippocampus/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Autism Spectrum Disorder , Autistic Disorder , Deubiquitinating Enzyme CYLD , Female , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins , Nerve Tissue Proteins , Neurons/metabolism , Synapses/metabolism , Ubiquitin/metabolism , Ubiquitination
20.
Eur J Immunol ; 52(2): 297-311, 2022 02.
Article in English | MEDLINE | ID: mdl-34727578

ABSTRACT

Plasma cells and their secreted antibodies play a central role in the long-term protection against chronic viral infection. However, due to experimental limitations, a comprehensive description of linked genotypic, phenotypic, and antibody repertoire features of plasma cells (gene expression, clonal frequency, virus specificity, and affinity) has been challenging to obtain. To address this, we performed single-cell transcriptome and antibody repertoire sequencing of the murine BM plasma cell population following chronic lymphocytic choriomeningitis virus infection. Our single-cell sequencing approach recovered full-length and paired heavy- and light-chain sequence information for thousands of plasma cells and enabled us to perform recombinant antibody expression and specificity screening. Antibody repertoire analysis revealed that, relative to protein immunization, chronic infection led to increased levels of clonal expansion, class-switching, and somatic variants. Furthermore, antibodies from the highly expanded and class-switched (IgG) plasma cells were found to be specific for multiple viral antigens and a subset of clones exhibited cross-reactivity to nonviral and autoantigens. Integrating single-cell transcriptome data with antibody specificity suggested that plasma cell transcriptional phenotype was correlated to viral antigen specificity. Our findings demonstrate that chronic viral infection can induce and sustain plasma cell clonal expansion, combined with significant somatic hypermutation, and can generate cross-reactive antibodies.


Subject(s)
Antibodies, Viral , Clonal Selection, Antigen-Mediated , Immunoglobulin Heavy Chains , Immunoglobulin Light Chains , Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus/immunology , Plasma Cells/immunology , Single-Cell Analysis , Animals , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Chronic Disease , Female , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL