Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Immunol ; 19(5): 1-7, 2018 05.
Article in English | MEDLINE | ID: mdl-29662171

ABSTRACT

The cytokine transforming growth factor-ß (TGF-ß) regulates the development and homeostasis of several tissue-resident macrophage populations, including microglia. TGF-ß is not critical for microglia survival but is required for the maintenance of the microglia-specific homeostatic gene signature1,2. Under defined host conditions, circulating monocytes can compete for the microglial niche and give rise to long-lived monocyte-derived macrophages residing in the central nervous system (CNS)3-5. Whether monocytes require TGF-ß for colonization of the microglial niche and maintenance of CNS integrity is unknown. We found that abrogation of TGF-ß signaling in CX3CR1+ monocyte-derived macrophages led to rapid onset of a progressive and fatal demyelinating motor disease characterized by myelin-laden giant macrophages throughout the spinal cord. Tgfbr2-deficient macrophages were characterized by high expression of genes encoding proteins involved in antigen presentation, inflammation and phagocytosis. TGF-ß is thus crucial for the functional integration of monocytes into the CNS microenvironment.


Subject(s)
Brain/immunology , Demyelinating Diseases/immunology , Macrophages/pathology , Spinal Cord/immunology , Transforming Growth Factor beta/immunology , Animals , Brain/metabolism , Brain/pathology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Signal Transduction , Spinal Cord/metabolism , Spinal Cord/pathology , Transforming Growth Factor beta/metabolism
2.
EMBO Rep ; 23(7): e54499, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35593064

ABSTRACT

Targeting myeloid cells, especially microglia, for the treatment of neuroinflammatory diseases such as multiple sclerosis (MS), is underappreciated. Our in silico drug screening reveals topoisomerase 1 (TOP1) inhibitors as promising drug candidates for microglial modulation. We show that TOP1 is highly expressed in neuroinflammatory conditions, and TOP1 inhibition using camptothecin (CPT) and its FDA-approved analog topotecan (TPT) reduces inflammatory responses in microglia/macrophages and ameliorates neuroinflammation in vivo. Transcriptomic analyses of sorted microglia from LPS-challenged mice reveal an altered transcriptional phenotype following TPT treatment. To target myeloid cells, we design a nanosystem using ß-glucan-coated DNA origami (MyloGami) loaded with TPT (TopoGami). MyloGami shows enhanced specificity to myeloid cells while preventing the degradation of the DNA origami scaffold. Myeloid-specific TOP1 inhibition using TopoGami significantly suppresses the inflammatory response in microglia and mitigates MS-like disease progression. Our findings suggest that TOP1 inhibition in myeloid cells represents a therapeutic strategy for neuroinflammatory diseases and that the myeloid-specific nanosystems we designed may also benefit the treatment of other diseases with dysfunctional myeloid cells.


Subject(s)
Neuroinflammatory Diseases , Topoisomerase I Inhibitors , Animals , DNA , Macrophages , Mice , Topoisomerase I Inhibitors/pharmacology , Topotecan/pharmacology
3.
Proc Natl Acad Sci U S A ; 117(6): 3103-3113, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31980518

ABSTRACT

Neutrophils are the most abundant immune cells found in actively inflamed joints of patients with rheumatoid arthritis (RA), and most animal models for RA depend on neutrophils for the induction of joint inflammation. Exogenous IL-4 and IL-13 protect mice from antibody-mediated joint inflammation, although the mechanism is not understood. Neutrophils display a very strong basal expression of STAT6, which is responsible for signaling following exposure to IL-4 and IL-13. Still, the role of IL-4 and IL-13 in neutrophil biology has not been well studied. This can be explained by the low neutrophil surface expression of the IL-4 receptor α-chain (IL-4Rα), essential for IL-4- and IL-13-induced STAT6 signaling. Here we identify that colony stimulating factor 3 (CSF3), released during acute inflammation, mediates potent STAT3-dependent neutrophil IL-4Rα up-regulation during sterile inflammatory conditions. We further demonstrate that IL-4 limits neutrophil migration to inflamed joints, and that CSF3 combined with IL-4 or IL-13 results in a prominent neutrophil up-regulation of the inhibitory Fcγ receptor (FcγR2b). Taking these data together, we demonstrate that the IL-4 and CSF3 pathways are linked and play important roles in regulating proinflammatory neutrophil behavior.


Subject(s)
Arthritis/metabolism , Interleukin-4 , Neutrophil Infiltration/physiology , Neutrophils/metabolism , Receptors, IgG/metabolism , Animals , Disease Models, Animal , Interleukin-4/genetics , Interleukin-4/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
4.
Proc Natl Acad Sci U S A ; 114(9): E1678-E1687, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28196884

ABSTRACT

Vitamin D exerts multiple immunomodulatory functions and has been implicated in the etiology and treatment of several autoimmune diseases, including multiple sclerosis (MS). We have previously reported that in juvenile/adolescent rats, vitamin D supplementation protects from experimental autoimmune encephalomyelitis (EAE), a model of MS. Here we demonstrate that this protective effect associates with decreased proliferation of CD4+ T cells and lower frequency of pathogenic T helper (Th) 17 cells. Using transcriptome, methylome, and pathway analyses in CD4+ T cells, we show that vitamin D affects multiple signaling and metabolic pathways critical for T-cell activation and differentiation into Th1 and Th17 subsets in vivo. Namely, Jak/Stat, Erk/Mapk, and Pi3K/Akt/mTor signaling pathway genes were down-regulated upon vitamin D supplementation. The protective effect associated with epigenetic mechanisms, such as (i) changed levels of enzymes involved in establishment and maintenance of epigenetic marks, i.e., DNA methylation and histone modifications; (ii) genome-wide reduction of DNA methylation, and (iii) up-regulation of noncoding RNAs, including microRNAs, with concomitant down-regulation of their protein-coding target RNAs involved in T-cell activation and differentiation. We further demonstrate that treatment of myelin-specific T cells with vitamin D reduces frequency of Th1 and Th17 cells, down-regulates genes in key signaling pathways and epigenetic machinery, and impairs their ability to transfer EAE. Finally, orthologs of nearly 50% of candidate MS risk genes and 40% of signature genes of myelin-reactive T cells in MS changed their expression in vivo in EAE upon supplementation, supporting the hypothesis that vitamin D may modulate risk for developing MS.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Vitamin D/pharmacology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Epigenesis, Genetic/drug effects , Genomics/methods , Lymphocyte Activation/drug effects , Multiple Sclerosis/drug therapy , Rats , Signal Transduction/genetics , Signal Transduction/immunology , Th1 Cells/drug effects , Th17 Cells/drug effects , Up-Regulation/drug effects
5.
Proc Natl Acad Sci U S A ; 113(8): 2188-93, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26862169

ABSTRACT

Multiple sclerosis (MS) is the most common chronic inflammatory disease of the central nervous system and also is regarded as an autoimmune condition. However, the antigenic targets of the autoimmune response in MS have not yet been deciphered. In an effort to mine the autoantibody repertoire within MS, we profiled 2,169 plasma samples from MS cases and population-based controls using bead arrays built with 384 human protein fragments selected from an initial screening with 11,520 antigens. Our data revealed prominently increased autoantibody reactivity against the chloride-channel protein anoctamin 2 (ANO2) in MS cases compared with controls. This finding was corroborated in independent assays with alternative protein constructs and by epitope mapping with peptides covering the identified region of ANO2. Additionally, we found a strong interaction between the presence of ANO2 autoantibodies and the HLA complex MS-associated DRB1*15 allele, reinforcing a potential role for ANO2 autoreactivity in MS etiopathogenesis. Furthermore, immunofluorescence analysis in human MS brain tissue showed ANO2 expression as small cellular aggregates near and inside MS lesions. Thus this study represents one of the largest efforts to characterize the autoantibody repertoire within MS. The findings presented here demonstrate that an ANO2 autoimmune subphenotype may exist in MS and lay the groundwork for further studies focusing on the pathogenic role of ANO2 autoantibodies in MS.


Subject(s)
Autoantibodies/blood , Chloride Channels/immunology , Membrane Proteins/immunology , Multiple Sclerosis/immunology , Adolescent , Adult , Aged , Anoctamins , Autoantigens/blood , Autoantigens/immunology , Autoantigens/metabolism , Brain/immunology , Brain/metabolism , Case-Control Studies , Chloride Channels/blood , Chloride Channels/metabolism , Epitope Mapping , Female , HLA-DRB1 Chains/genetics , Humans , Immunohistochemistry , Male , Membrane Proteins/blood , Membrane Proteins/metabolism , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/genetics , Peptide Fragments/blood , Peptide Fragments/immunology , Young Adult
6.
J Neuroinflammation ; 12: 194, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26511327

ABSTRACT

BACKGROUND: Ιn multiple sclerosis (MS), axonal damage leads to permanent neurological disabilities and the spreading of the autoimmune response to axonal antigens is implicated in disease progression. Experimental autoimmune encephalomyelitis (EAE) provides an animal model that mimics MS. Using different EAE models, we investigated the pathophysiological basis of epitope spreading to neurofascin, a protein localized at the node of Ranvier and its regulation by non-MHC genes. METHODS: We used two different EAE models in DA rat; one which is induced with myelin oligodendrocyte glycoprotein (MOG) which leads to disease characterized by profound demyelination, and the second which is induced with myelin basic protein (MBP) peptide 63-88 which results in severe central nervous system (CNS) inflammation but little or no demyelination. We determined anti-neurofascin antibody levels during the course of disease. Furthermore, the anti-neurofascin IgG response was correlated with clinical parameters in 333 (DAxPVG.1AV1) x DA rats on which we performed linkage analysis to determine if epitope spreading to neurofascin was affected by non-MHC genes. RESULTS: Spreading of the antibody response to neurofascin occurred in demyelinating MOG-induced EAE but not in EAE induced with MBP peptide 63-88. Anti-neurofascin IgG levels correlated with disease severity in (DAxPVG.1AV1) x DA rats, and a genomic region on chromosome 3 was found to influence this response. CONCLUSIONS: Inter-molecular epitope spreading to neurofascin correlates with disease severity in MOG-EAE is dependent on extensive demyelination and is influenced by non-MHC genes. The findings presented here may shed light on factors involved in the severity of MS and its genetics.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Myelin-Oligodendrocyte Glycoprotein/immunology , Nerve Growth Factors/genetics , Nerve Growth Factors/immunology , Animals , Demyelinating Diseases/drug therapy , Demyelinating Diseases/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Epitopes , Female , Immunoglobulin G/immunology , Inflammation/chemically induced , Inflammation/pathology , Male , Myelin Basic Protein/pharmacology , Peptides/pharmacology , Rats
7.
Neurobiol Dis ; 71: 220-33, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25149263

ABSTRACT

Multiple sclerosis (MS) is the most common chronic inflammatory demyelinating disease of the central nervous system (CNS) in young adults. Chronic treatments with histone deacetylase inhibitors (HDACis) have been reported to ameliorate experimental autoimmune encephalomyelitis (EAE), a rodent model of MS, by targeting immune responses. We have recently shown that the HDAC inhibition/knockdown in the presence of thyroid hormone (T3) can also promote oligodendrocyte (OL) differentiation and expression of myelin genes in neural stem cells (NSCs) and oligodendrocyte precursors (OPCs). In this study, we found that treatment with an HDACi, valproic acid (VPA), and T3, alone or in combination, directly affects encephalitogenic CD4+ T cells. VPA, but not T3, compromised their proliferation, while both molecules reduced the frequency of IL-17-producing cells. Transfer of T3, VPA and VPA/T3 treated encephalitogenic CD4+ T cells into naïve rats induced less severe EAE, indicating that the effects of these molecules are persistent and do not require their maintenance after the initial stimuli. Thus, we investigated the effect of acute treatment with VPA and l-thyroxine (T4), a precursor of T3, on myelin oligodendrocyte glycoprotein-induced EAE in Dark Agouti rats, a close mimic of MS. We found that a brief treatment after disease onset led to sustained amelioration of EAE and prevention of inflammatory demyelination in the CNS accompanied with a higher expression of myelin-related genes in the brain. Furthermore, the treatment modulated immune responses, reduced the number of CD4+ T cells and affected the Th1 differentiation program in the brain. Our data indicate that an acute treatment with VPA and T4 after the onset of EAE can produce persistent clinically relevant therapeutic effects by limiting the pathogenic immune reactions while promoting myelin gene expression.


Subject(s)
Brain/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme Inhibitors/therapeutic use , Thyroxine/therapeutic use , Valproic Acid/therapeutic use , Analysis of Variance , Animals , Brain/pathology , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/etiology , Flow Cytometry , Interleukin-17/metabolism , Ki-67 Antigen/metabolism , Myelin Basic Protein/immunology , Myelin Basic Protein/toxicity , Peptide Fragments/immunology , Peptide Fragments/toxicity , Rats
8.
PLoS Pathog ; 8(6): e1002753, 2012.
Article in English | MEDLINE | ID: mdl-22761571

ABSTRACT

Herpes simplex encephalitis (HSE) is a fatal infection of the central nervous system (CNS) predominantly caused by Herpes simplex virus type 1. Factors regulating the susceptibility to HSE are still largely unknown. To identify host gene(s) regulating HSE susceptibility we performed a genome-wide linkage scan in an intercross between the susceptible DA and the resistant PVG rat. We found one major quantitative trait locus (QTL), Hse1, on rat chromosome 4 (confidence interval 24.3-31 Mb; LOD score 29.5) governing disease susceptibility. Fine mapping of Hse1 using recombinants, haplotype mapping and sequencing, as well as expression analysis of all genes in the interval identified the calcitonin receptor gene (Calcr) as the main candidate, which also is supported by functional studies. Thus, using unbiased genetic approach variability in Calcr was identified as potentially critical for infection and viral spread to the CNS and subsequent HSE development.


Subject(s)
Encephalitis, Herpes Simplex/genetics , Genetic Predisposition to Disease/genetics , Neurons/virology , Receptors, Calcitonin/genetics , Animals , Chromosome Mapping/methods , Flow Cytometry , Genome-Wide Association Study , Genotype , Haplotypes , Herpesvirus 1, Human , Quantitative Trait Loci , Rats , Real-Time Polymerase Chain Reaction , Receptors, Calcitonin/metabolism , Transfection
9.
Nat Commun ; 15(1): 383, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38195627

ABSTRACT

Microglia harness an unutilized health-promoting potential in age-related neurodegenerative and neuroinflammatory diseases, conditions like progressive multiple sclerosis (MS). Our research unveils an microglia population emerging in the cortical brain regions of aging mice, marked by ERK1/2, Akt, and AMPK phosphorylation patterns and a transcriptome indicative of activated autophagy - a process critical for cellular adaptability. By deleting the core autophagy gene Ulk1 in microglia, we reduce this population in the central nervous system of aged mice. Notably, this population is found dependent on IL-34, rather than CSF1, although both are ligands for CSF1R. When aging mice are exposed to autoimmune neuroinflammation, the loss of autophagy-dependent microglia leads to neural and glial cell death and increased mortality. Conversely, microglial expansion mediated by IL-34 exhibits a protective effect. These findings shed light on an autophagy-dependent neuroprotective microglia population as a potential target for treating age-related neuroinflammatory conditions, including progressive MS.


Subject(s)
Central Nervous System , Microglia , Animals , Mice , Neuroglia , Autophagy/genetics , Interleukins
10.
Brain Behav Immun ; 27(1): 109-22, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23044177

ABSTRACT

Increasing evidence suggests that genetic background affects outcome of traumatic brain injuries (TBI). Still, there is limited detailed knowledge on what pathways/processes are affected by genetic heterogeneity. The inbred rat strains DA and PVG differ in neuronal survival following TBI. We here carried out global expressional profiling to identify differentially regulated pathways governing the response to an experimental controlled brain contusion injury. One of the most differentially regulated molecular networks concerned immune cell trafficking. Subsequent characterization of the involved cells using flow cytometry demonstrated greater infiltration of neutrophils and monocytes, as well as a higher degree of microglia activation in DA compared to PVG rats. In addition, DA rats displayed a higher number of NK cells and a higher ratio of CD161bright compared to CD161dim NK cells. Local expression of complement pathway molecules such as C1 and C3 was higher in DA and both the key complement component C3 and membrane-attack complex (MAC) could be demonstrated on axons and nerve cells. A stronger activation of the complement system in DA was associated with higher cerebrospinal fluid levels of neurofilament-light, a biomarker for nerve/axonal injury. In summary, we demonstrate substantial differences between DA and PVG rats in activation of inflammatory pathways; in particular, immune cell influx and complement activation associated with neuronal/axonal injury after TBI. These findings suggest genetic influences acting on inflammatory activation to be of importance in TBI and motivate further efforts using experimental forward genetics to identify genes/pathways that affect outcome.


Subject(s)
Brain Injuries , Complement Activation , Leukocytes , RNA, Messenger/analysis , Rats, Inbred Strains , Animals , Brain Injuries/genetics , Brain Injuries/immunology , Cell Movement/genetics , Complement Activation/genetics , Complement Activation/immunology , Complement C1q/genetics , Complement C1q/immunology , Complement C3/genetics , Complement C3/immunology , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/immunology , Complement System Proteins/genetics , Complement System Proteins/immunology , Cytokines/genetics , Cytokines/immunology , Gene Expression Profiling , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Leukocytes/cytology , Leukocytes/immunology , Microglia/cytology , Microglia/immunology , Monocytes/cytology , Monocytes/immunology , NK Cell Lectin-Like Receptor Subfamily B/genetics , NK Cell Lectin-Like Receptor Subfamily B/immunology , Neutrophils/cytology , Neutrophils/immunology , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred Strains/genetics , Rats, Inbred Strains/immunology
11.
Sci Adv ; 9(20): eadg3032, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37196088

ABSTRACT

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system, for which and Epstein-Barr virus (EBV) infection is a likely prerequisite. Due to the homology between Epstein-Barr nuclear antigen 1 (EBNA1) and alpha-crystallin B (CRYAB), we examined antibody reactivity to EBNA1 and CRYAB peptide libraries in 713 persons with MS (pwMS) and 722 matched controls (Con). Antibody response to CRYAB amino acids 7 to 16 was associated with MS (OR = 2.0), and combination of high EBNA1 responses with CRYAB positivity markedly increased disease risk (OR = 9.0). Blocking experiments revealed antibody cross-reactivity between the homologous EBNA1 and CRYAB epitopes. Evidence for T cell cross-reactivity was obtained in mice between EBNA1 and CRYAB, and increased CRYAB and EBNA1 CD4+ T cell responses were detected in natalizumab-treated pwMS. This study provides evidence for antibody cross-reactivity between EBNA1 and CRYAB and points to a similar cross-reactivity in T cells, further demonstrating the role of EBV adaptive immune responses in MS development.


Subject(s)
Epstein-Barr Virus Infections , Multiple Sclerosis , alpha-Crystallins , Animals , Mice , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human
12.
J Cell Mol Med ; 16(1): 129-41, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21342435

ABSTRACT

The syntaxin 11 (STX11) gene is mutated in a proportion of patients with familial haemophagocytic lymphohistiocytosis (FHL) and exocytosis of cytotoxic granules is impaired in STX11-deficient NK cells. However, the subcellular localization, regulation of expression and molecular function of STX11 in NK cells and other cytotoxic lymphocytes remain unknown. Here we demonstrate that STX11 expression is strictly controlled by several mechanisms in a cell-type-specific manner and that the enzymatic activity of the proteasome is required for STX11 expression in NK cells. In resting NKL cells, STX11 was localized in the cation-dependent mannose-6-phosphate receptor (CD-M6PR)-containing compartment, which was clearly distinct from cytotoxic granules or Rab27a-expressing vesicles. These subcellular structures appeared to fuse at the contact area with NK-sensitive target cells as demonstrated by partial colocalization of STX11 with perforin and Rab27a. Although STX11-deficent allo-specific cytotoxic T-lymphocytes efficiently lysed target cells and released cytotoxic granules, they exhibited a significantly lower extent of spontaneous association of perforin with Rab27a as compared with STX11-expressing T cells. Thus, our results suggest that STX11 promotes the fusion of Rab27a-expressing vesicles with cytotoxic granules and reveal an additional level of complexity in the spatial/temporal segregation of subcellular structures participating in the process of granule-mediated cytotoxicity.


Subject(s)
Cytoplasmic Granules/metabolism , Immunological Synapses/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Qa-SNARE Proteins/metabolism , Cell Line , Humans , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/physiopathology , Perforin/metabolism , Proteasome Endopeptidase Complex/metabolism , Qa-SNARE Proteins/genetics , Subcellular Fractions/metabolism , T-Lymphocytes, Cytotoxic/immunology , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab27 GTP-Binding Proteins
14.
Sci Adv ; 8(17): eabn1823, 2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35476434

ABSTRACT

Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS), in which pathological T cells, likely autoimmune, play a key role. Despite its central importance, the autoantigen repertoire remains largely uncharacterized. Using a novel in vitro antigen delivery method combined with the Human Protein Atlas library, we screened for T cell autoreactivity against 63 CNS-expressed proteins. We identified four previously unreported autoantigens in MS: fatty acid-binding protein 7, prokineticin-2, reticulon-3, and synaptosomal-associated protein 91, which were verified to induce interferon-γ responses in MS in two cohorts. Autoreactive profiles were heterogeneous, and reactivity to several autoantigens was MS-selective. Autoreactive T cells were predominantly CD4+ and human leukocyte antigen-DR restricted. Mouse immunization induced antigen-specific responses and CNS leukocyte infiltration. This represents one of the largest systematic efforts to date in the search for MS autoantigens, demonstrates the heterogeneity of autoreactive profiles, and highlights promising targets for future diagnostic tools and immunomodulatory therapies in MS.

15.
Sci Immunol ; 5(52)2020 10 16.
Article in English | MEDLINE | ID: mdl-33067381

ABSTRACT

Multiple sclerosis (MS) is a leading cause of incurable progressive disability in young adults caused by inflammation and neurodegeneration in the central nervous system (CNS). The capacity of microglia to clear tissue debris is essential for maintaining and restoring CNS homeostasis. This capacity diminishes with age, and age strongly associates with MS disease progression, although the underlying mechanisms are still largely elusive. Here, we demonstrate that the recovery from CNS inflammation in a murine model of MS is dependent on the ability of microglia to clear tissue debris. Microglia-specific deletion of the autophagy regulator Atg7, but not the canonical macroautophagy protein Ulk1, led to increased intracellular accumulation of phagocytosed myelin and progressive MS-like disease. This impairment correlated with a microglial phenotype previously associated with neurodegenerative pathologies. Moreover, Atg7-deficient microglia showed notable transcriptional and functional similarities to microglia from aged wild-type mice that were also unable to clear myelin and recover from disease. In contrast, induction of autophagy in aged mice using the disaccharide trehalose found in plants and fungi led to functional myelin clearance and disease remission. Our results demonstrate that a noncanonical form of autophagy in microglia is responsible for myelin degradation and clearance leading to recovery from MS-like disease and that boosting this process has a therapeutic potential for age-related neuroinflammatory conditions.


Subject(s)
Autophagy-Related Protein 7/deficiency , Encephalomyelitis, Autoimmune, Experimental/immunology , Microglia/immunology , Multiple Sclerosis/immunology , Phagocytosis/immunology , Animals , Autophagy/immunology , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein-1 Homolog/deficiency , Autophagy-Related Protein-1 Homolog/genetics , Brain/cytology , Brain/immunology , Brain/pathology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Male , Mice , Mice, Knockout , Microglia/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/metabolism , Primary Cell Culture , Spinal Cord/cytology , Spinal Cord/immunology , Spinal Cord/pathology
16.
PLoS Pathog ; 3(6): e80, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17559303

ABSTRACT

Although malaria and Epstein-Barr (EBV) infection are recognized cofactors in the genesis of endemic Burkitt lymphoma (BL), their relative contribution is not understood. BL, the most common paediatric cancer in equatorial Africa, is a high-grade B cell lymphoma characterized by c-myc translocation. EBV is a ubiquitous B lymphotropic virus that persists in a latent state after primary infection, and in Africa, most children have sero-converted by 3 y of age. Malaria infection profoundly affects the B cell compartment, inducing polyclonal activation and hyper-gammaglobulinemia. We recently identified the cystein-rich inter-domain region 1alpha (CIDR1alpha) of the Plasmodium falciparum membrane protein 1 as a polyclonal B cell activator that preferentially activates the memory compartment, where EBV is known to persist. Here, we have addressed the mechanisms of interaction between CIDR1alpha and EBV in the context of B cells. We show that CIDR1alpha binds to the EBV-positive B cell line Akata and increases the number of cells switching to the viral lytic cycle as measured by green fluorescent protein (GFP) expression driven by a lytic promoter. The virus production in CIDR1alpha-exposed cultures was directly proportional to the number of GFP-positive Akata cells (lytic EBV) and to the increased expression of the EBV lytic promoter BZLF1. Furthermore, CIDR1alpha stimulated the production of EBV in peripheral blood mononuclear cells derived from healthy donors and children with BL. Our results suggest that P. falciparum antigens such as CIDR1alpha can directly induce EBV reactivation during malaria infection that may increase the risk of BL development for children living in malaria-endemic areas. To our knowledge, this is the first report to show that a microbial protein can drive a latently infected B cell into EBV replication.


Subject(s)
Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Malaria/virology , Plasmodium falciparum/genetics , Virus Activation/genetics , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Cell Line, Tumor , Child , Child, Preschool , DNA, Viral/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Erythrocytes/parasitology , Gene Expression Regulation, Viral , Herpesvirus 4, Human/immunology , Humans , Leukocytes, Mononuclear/virology , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Recurrence , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Activation/immunology , Virus Replication
17.
Nat Med ; 24(12): 1837-1844, 2018 12.
Article in English | MEDLINE | ID: mdl-30420755

ABSTRACT

Multiple sclerosis (MS) is characterized by an immune system attack targeting myelin, which is produced by oligodendrocytes (OLs). We performed single-cell transcriptomic analysis of OL lineage cells from the spinal cord of mice induced with experimental autoimmune encephalomyelitis (EAE), which mimics several aspects of MS. We found unique OLs and OL precursor cells (OPCs) in EAE and uncovered several genes specifically alternatively spliced in these cells. Surprisingly, EAE-specific OL lineage populations expressed genes involved in antigen processing and presentation via major histocompatibility complex class I and II (MHC-I and -II), and in immunoprotection, suggesting alternative functions of these cells in a disease context. Importantly, we found that disease-specific oligodendroglia are also present in human MS brains and that a substantial number of genes known to be susceptibility genes for MS, so far mainly associated with immune cells, are expressed in the OL lineage cells. Finally, we demonstrate that OPCs can phagocytose and that MHC-II-expressing OPCs can activate memory and effector CD4-positive T cells. Our results suggest that OLs and OPCs are not passive targets but instead active immunomodulators in MS. The disease-specific OL lineage cells, for which we identify several biomarkers, may represent novel direct targets for immunomodulatory therapeutic approaches in MS.


Subject(s)
Cell Lineage/genetics , Immune System , Multiple Sclerosis/genetics , Transcriptome/genetics , Alternative Splicing/genetics , Animals , Antigen Presentation/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gene Expression Regulation/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II/genetics , Humans , Mice , Multiple Sclerosis/physiopathology , Myelin Sheath/genetics , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/pathology , Oligodendroglia/metabolism , Single-Cell Analysis
18.
19.
J Leukoc Biol ; 99(3): 437-46, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26516183

ABSTRACT

Dendritic cells are professional APCs that play a central role in the initiation of immune responses. The limited ex vivo availability of dendritic cells inspires the widespread use of bone marrow-derived dendritic cells as an alternative in research. However, the functional characteristics of bone marrow-derived dendritic cells are incompletely understood. Therefore, we compared functional and phenotypic characteristics of rat bone marrow-derived dendritic cells generated with GM-CSF/IL-4 or FLT3 ligand bone marrow-derived dendritic cells. A comparison of surface markers revealed that FLT3 ligand-bone marrow-derived dendritic cells expressed signal regulatory protein α, CD103, and CD4 and baseline levels of MHC class II, CD40, and CD86, which were highly up-regulated upon stimulation. Conversely, GM-CSF/IL-4-bone marrow-derived dendritic cells constitutively expressed signal regulatory protein α, CD11c, and CD11b but only mildly up-regulated MHC class II, CD40, or CD86 following stimulation. Expression of dendritic cell-associated core transcripts was restricted to FLT3 ligand-bone marrow-derived dendritic cells . GM-CSF/IL-4-bone marrow-derived dendritic cells were superior at phagocytosis but were outperformed by FLT3 ligand-bone marrow-derived dendritic cells at antigen presentation and T cell stimulation in vitro. Stimulated GM-CSF/IL-4-bone marrow-derived dendritic cells secreted more TNF, CCL5, CCL20, and NO, whereas FLT3 ligand-bone marrow-derived dendritic cells secreted more IL-6 and IL-12. Finally, whereas GM-CSF/IL-4-bone marrow-derived dendritic cell culture supernatants added to resting T cell cultures promoted forkhead box p3(+) regulatory T cell populations, FLT3 ligand-bone marrow-derived dendritic cell culture supernatants drove Th17 differentiation. We conclude that rat GM-CSF/IL-4-bone marrow-derived dendritic cells and FLT3 ligand-bone marrow-derived dendritic cells are functionally distinct. Our data support the current rationale that FLT3 ligand-bone marrow-derived dendritic cells mostly resemble classic dendritic cells but comprise additional minor subpopulations, whereas GM-CSF/IL-4-bone marrow-derived dendritic cells resemble monocyte-derived inflammatory dendritic cells (iNOS-positive monocyte-derived cells).


Subject(s)
Bone Marrow Cells/physiology , Dendritic Cells/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-4/pharmacology , Membrane Proteins/pharmacology , Animals , Phenotype , Rats , Rats, Inbred Lew
20.
J Inflamm Res ; 8: 211-25, 2015.
Article in English | MEDLINE | ID: mdl-26622189

ABSTRACT

Multiple sclerosis (MS) is a complex autoimmune condition with firmly established genetic and environmental components. Genome-wide association studies (GWAS) have revealed a large number of genetic polymorphisms in the vicinity of, and within, genes that associate to disease. However, the significance of these single-nucleotide polymorphisms in disease and possible mechanisms of action remain, with a few exceptions, to be established. While the animal model for MS, experimental autoimmune encephalomyelitis (EAE), has been instrumental in understanding immunity in general and mechanisms of MS disease in particular, much of the translational information gathered from the model in terms of treatment development (glatiramer acetate and natalizumab) has been extensively summarized. In this review, we would thus like to cover the work done in EAE from a GWAS perspective, highlighting the research that has addressed the role of different GWAS genes and their pathways in EAE pathogenesis. Understanding the contribution of these pathways to disease might allow for the stratification of disease subphenotypes in patients and in turn open the possibility for new and individualized treatment approaches in the future.

SELECTION OF CITATIONS
SEARCH DETAIL