Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 18(1): 246, 2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34711216

ABSTRACT

BACKGROUND: Cognitive deficits are common in patients with sepsis. Previous studies in sepsis-associated encephalopathy (SAE) implicated the C-X-C chemokine receptor type (CXCR) 5. The present study used a mouse model of SAE to examine whether CXCR5 down-regulation could attenuate cognitive deficits. METHODS: Sepsis was induced in adult male C57BL/6 J and CXCR5-/- mice by cecal ligation and puncture (CLP). At 14-18 days after surgery, animals were tested in a Morris water maze, followed by a fear conditioning test. Transmission electron microscopy of hippocampal sections was used to assess levels of autophagy. Primary microglial cultures challenged with lipopolysaccharide (LPS) were used to examine the effects of short interfering RNA targeting CXCR5, and to investigate the possible involvement of the p38MAPK/NF-κB/STAT3 signaling pathway. RESULTS: CLP impaired learning and memory and up-regulated CXCR5 in hippocampal microglia. CLP activated hippocampal autophagy, as reflected by increases in numbers of autophagic vacuoles, conversion of microtubule-associated protein 1 light chain 3 (LC3) from form I to form II, accumulation of beclin-1 and autophagy-related gene-5, and a decrease in p62 expression. CLP also shifted microglial polarization to the M1 phenotype, and increased levels of IL-1ß, IL-6 and phosphorylated p38MAPK. CXCR5 knockout further enhanced autophagy but partially reversed all the other CLP-induced effects, including cognitive deficits. Similar effects on autophagy and cytokine expression were observed after knocking down CXCR5 in LPS-challenged primary microglial cultures; this knockdown also partially reversed LPS-induced up-regulation of phosphorylated NF-κB and STAT3. The p38MAPK agonist P79350 partially reversed the effects of CXCR5 knockdown in microglial cultures. CONCLUSIONS: CXCR5 may act via p38MAPK/NF-κB/STAT3 signaling to inhibit hippocampal autophagy during sepsis and thereby contribute to cognitive dysfunction. Down-regulating CXCR5 can restore autophagy and mitigate the proinflammatory microenvironment in the hippocampus.


Subject(s)
Cognitive Dysfunction/metabolism , NF-kappa B/metabolism , Receptors, CXCR5/deficiency , STAT3 Transcription Factor/metabolism , Sepsis-Associated Encephalopathy/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Autophagy/physiology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/prevention & control , Down-Regulation/physiology , Male , Maze Learning/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , NF-kappa B/genetics , Receptors, CXCR5/genetics , STAT3 Transcription Factor/genetics , Sepsis-Associated Encephalopathy/genetics , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/genetics
2.
Nat Commun ; 12(1): 4452, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294713

ABSTRACT

Atherosclerosis-associated cardiovascular disease is one of the main causes of death and disability among patients with diabetes mellitus. However, little is known about the impact of S-nitrosylation in diabetes-accelerated atherosclerosis. Here, we show increased levels of S-nitrosylation of guanine nucleotide-binding protein G(i) subunit alpha-2 (SNO-GNAI2) at Cysteine 66 in coronary artery samples from diabetic patients with atherosclerosis, consistently with results from mice. Mechanistically, SNO-GNAI2 acted by coupling with CXCR5 to dephosphorylate the Hippo pathway kinase LATS1, thereby leading to nuclear translocation of YAP and promoting an inflammatory response in endothelial cells. Furthermore, Cys-mutant GNAI2 refractory to S-nitrosylation abrogated GNAI2-CXCR5 coupling, alleviated atherosclerosis in diabetic mice, restored Hippo activity, and reduced endothelial inflammation. In addition, we showed that melatonin treatment restored endothelial function and protected against diabetes-accelerated atherosclerosis by preventing GNAI2 S-nitrosylation. In conclusion, SNO-GNAI2 drives diabetes-accelerated atherosclerosis by coupling with CXCR5 and activating YAP-dependent endothelial inflammation, and reducing SNO-GNAI2 is an efficient strategy for alleviating diabetes-accelerated atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Diabetic Angiopathies/etiology , Diabetic Angiopathies/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Cysteine/chemistry , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , GTP-Binding Protein alpha Subunit, Gi2/chemistry , GTP-Binding Protein alpha Subunit, Gi2/genetics , Hippo Signaling Pathway , Human Umbilical Vein Endothelial Cells , Humans , Male , Melatonin/pharmacology , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Nitric Oxide Synthase Type II/metabolism , Nitroso Compounds/chemistry , Nitroso Compounds/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Transcription Factors/metabolism , YAP-Signaling Proteins
3.
Front Immunol ; 10: 1903, 2019.
Article in English | MEDLINE | ID: mdl-31474986

ABSTRACT

Previous research has shown that CXCR5-/- mice develop retinal degeneration (RD) with age, a characteristic related to age macular degeneration (AMD). RD in these mice is not well-understood, and in this study, we sought to characterize further the RD phenotype and to gain mechanistic insights into the function of CXCR5 in the retina. CXCR5-/- and WT control mice were used. Fundus images demonstrated a significant (p < 0.001) increase of hypo-pigmented spots in the retina of aged CXCR5-/- mice compared with WT control mice. PAS staining indicated localization of deposits in the sub-retinal pigment epithelia (RPE) layer. AMD-associated proteins Cryab, amyloid beta, and C3d were detected within the RPE/sub-RPE tissues by immunofluorescence (IF). In addition, western blot analysis of COX-2, Arg1, and VEGF-a revealed an increase in the signaling of these molecules within the RPE/choroid complex. Transmission electron microscopy (TEM) indicated a drusen-like structure of sub-RPE deposits with an accumulation of vacuolated cellular debris. Loss of photoreceptors was detected by peanut lectin staining and was corroborated by a reduction in MAP2 signaling. Loss of blood-retinal barrier integrity was demonstrated by a reduction of ZO-1 expression. Inflammatory cells were detected in the sub-RPE space, with an increase in IBA-1 positive microglia cells on the surface of the RPE. Mass spectrometry analysis of CXCR5-/- mouse RPE/choroid proteins extracts, separated by SDS-page and incubated with autologous serum, identified autoantibodies against AMD-associated proteins: Cryaa, Cryab, and Anxa2. In vitro evaluations in BV-2 cell culture indicated a significant increase in production of Arg-1 (p < 0.001) and COX-2 (p < 0.01) in the presence of anti-CXCR5 antibody when compared with Igg-treated control BV-2 cells stimulated with IL-4 and TNFα/IFNγ, respectively. Anti-CXCR5 antibody treatment without stimulating agents did not affect Arg-1 and COX-2 expression; this suggests that CXCR5 may have a regulatory role in microglia cells activation. These results indicate that with age, CXCR5-/- mice develop RD characterized by microglia dysfunction, increased production of CXCL13 in the RPE progressive photoreceptor, neuronal loss, and sub-RPE deposition of cellular debris, resulting in the production of immunogenic proteins and autoimmune-mediated RD.


Subject(s)
Autoimmunity/immunology , Disease Models, Animal , Macular Degeneration/immunology , Receptors, CXCR5/immunology , Retinal Degeneration/immunology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Animals , Autoimmunity/genetics , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , Cell Line , Cyclooxygenase 2/immunology , Cyclooxygenase 2/metabolism , Fluorescent Antibody Technique , Macular Degeneration/genetics , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/immunology , Microfilament Proteins/metabolism , Microglia/cytology , Microglia/immunology , Microglia/metabolism , Microscopy, Electron, Transmission , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , Retinal Degeneration/genetics , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/ultrastructure , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism , Zonula Occludens-1 Protein/immunology , Zonula Occludens-1 Protein/metabolism , alpha-Crystallin B Chain/immunology , alpha-Crystallin B Chain/metabolism
4.
Transplantation ; 103(9): 1809-1820, 2019 09.
Article in English | MEDLINE | ID: mdl-30830040

ABSTRACT

BACKGROUND: We previously reported the novel activity of alloprimed CD8 T cells that suppress posttransplant alloantibody production. The purpose of the study is to investigate the expression and role of CXCR5 on antibody-suppressor CD8 T-cell function. METHODS: C57BL/6 mice were transplanted with FVB/N hepatocytes. Alloprimed CD8 T cells were retrieved on day 7 from hepatocyte transplant recipients. Unsorted or flow-sorted (CXCR5CXCR3 and CXCR3CXCR5) alloprimed CD8 T-cell subsets were analyzed for in vitro cytotoxicity and capacity to inhibit in vivo alloantibody production following adoptive transfer into C57BL/6 or high alloantibody-producing CD8 knock out (KO) hepatocyte transplant recipients. Alloantibody titer was assessed in CD8 KO mice reconstituted with naive CD8 T cells retrieved from C57BL/6, CXCR5 KO, or CXCR3 KO mice. Antibody suppression by ovalbumin (OVA)-primed monoclonal OVA-specific t-cell receptor transgenic CD8+ T cells (OT-I) CXCR5 or CXCR3 CD8 T-cell subsets was also investigated. RESULTS: Alloprimed CXCR5CXCR3CD8 T cells mediated in vitro cytotoxicity of alloprimed "self" B cells, while CXCR3CXCR5CD8 T cells did not. Only flow-sorted alloprimed CXCR5CXCR3CD8 T cells (not flow-sorted alloprimed CXCR3CXCR5CD8 T cells) suppressed alloantibody production and enhanced graft survival when transferred into transplant recipients. Unlike CD8 T cells from wild-type or CXCR3 KO mice, CD8 T cells from CXCR5 KO mice do not develop alloantibody-suppressor function. Similarly, only flow-sorted CXCR5CXCR3 (and not CXCR3CXCR5) OVA-primed OT-I CD8 T cells mediated in vivo suppression of anti-OVA antibody production. CONCLUSIONS: These data support the conclusion that expression of CXCR5 by antigen-primed CD8 T cells is critical for the function of antibody-suppressor CD8 T cells.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , B-Lymphocytes/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Rejection/prevention & control , Hepatocytes/transplantation , Liver Transplantation , Receptors, CXCR5/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , CD8 Antigens/deficiency , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Female , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Survival , Hepatocytes/immunology , Hepatocytes/metabolism , Liver Transplantation/adverse effects , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/deficiency , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , Signal Transduction , Time Factors
5.
Sci Rep ; 7(1): 8933, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28827539

ABSTRACT

The chemokine receptor CXCR5 is primarily expressed on B cells and Tfh cells and facilitates their migration towards B cell follicles. In the present study we investigated the role of the CXCL13/CXCR5 axis in the pathogenesis of rheumatoid arthritis (RA) and specifically addressed the impact of CXCR5-mediated T and B cell migration in this disease. Employing collagen-induced arthritis (CIA) we identify CXCR5 as an absolutely essential factor for the induction of inflammatory autoimmune arthritis. Cxcr5-deficient mice and mice selectively lacking Cxcr5 on T cells were completely resistant to CIA, showed impaired germinal center responses and failed to mount an IgG1 antibody response to collagen II. Selective ablation of CXCR5 expression in B cells also led to suppression of CIA owing to diminished GC responses in secondary lymphoid organs (SLO) and impaired anti-collagen II antibody production. Chimeric mice harboring Cxcr5-proficient and Cxcr5-deficient immune cells revealed SLO and not the synovial tissue as the compartment where CXCR5-mediated cell migration induces autoimmune inflammation in arthritis. Thus our data demonstrate that CXCR5-mediated co-localization of Tfh cells and B cells in SLOs is absolutely essential for the induction of RA and identify CXCR5 and Tfh cells as promising therapeutic targets for the treatment of RA.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Receptors, CXCR5/deficiency , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Arthritis, Rheumatoid/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Disease Models, Animal , Disease Susceptibility , Humans , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Mice , Mice, Knockout , Organ Specificity/genetics , Phenotype
6.
Nature ; 542(7639): 110-114, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28150777

ABSTRACT

CD4+ T cells are central mediators of autoimmune pathology; however, defining their key effector functions in specific autoimmune diseases remains challenging. Pathogenic CD4+ T cells within affected tissues may be identified by expression of markers of recent activation. Here we use mass cytometry to analyse activated T cells in joint tissue from patients with rheumatoid arthritis, a chronic immune-mediated arthritis that affects up to 1% of the population. This approach revealed a markedly expanded population of PD-1hiCXCR5-CD4+ T cells in synovium of patients with rheumatoid arthritis. However, these cells are not exhausted, despite high PD-1 expression. Rather, using multidimensional cytometry, transcriptomics, and functional assays, we define a population of PD-1hiCXCR5- 'peripheral helper' T (TPH) cells that express factors enabling B-cell help, including IL-21, CXCL13, ICOS, and MAF. Like PD-1hiCXCR5+ T follicular helper cells, TPH cells induce plasma cell differentiation in vitro through IL-21 secretion and SLAMF5 interaction (refs 3, 4). However, global transcriptomics highlight differences between TPH cells and T follicular helper cells, including altered expression of BCL6 and BLIMP1 and unique expression of chemokine receptors that direct migration to inflamed sites, such as CCR2, CX3CR1, and CCR5, in TPH cells. TPH cells appear to be uniquely poised to promote B-cell responses and antibody production within pathologically inflamed non-lymphoid tissues.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , B-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Arthritis, Rheumatoid/blood , B-Lymphocytes/pathology , Cell Differentiation , Cell Movement , Chemokine CXCL13/metabolism , Gene Expression Profiling , Humans , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukins/metabolism , Macrophage-Activating Factors , Positive Regulatory Domain I-Binding Factor 1 , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, CXCR5/deficiency , Receptors, CXCR5/metabolism , Receptors, Chemokine/metabolism , Repressor Proteins/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism , Synovial Fluid/immunology , T-Lymphocytes, Helper-Inducer/metabolism
7.
Nature ; 537(7620): 412-428, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27501245

ABSTRACT

During chronic viral infection, virus-specific CD8(+) T cells become exhausted, exhibit poor effector function and lose memory potential. However, exhausted CD8(+) T cells can still contain viral replication in chronic infections, although the mechanism of this containment is largely unknown. Here we show that a subset of exhausted CD8(+) T cells expressing the chemokine receptor CXCR5 has a critical role in the control of viral replication in mice that were chronically infected with lymphocytic choriomeningitis virus (LCMV). These CXCR5(+) CD8(+) T cells were able to migrate into B-cell follicles, expressed lower levels of inhibitory receptors and exhibited more potent cytotoxicity than the CXCR5(-) [corrected] subset. Furthermore, we identified the Id2-E2A signalling axis as an important regulator of the generation of this subset. In patients with HIV, we also identified a virus-specific CXCR5(+) CD8(+) T-cell subset, and its number was inversely correlated with viral load. The CXCR5(+) subset showed greater therapeutic potential than the CXCR5(-) [corrected] subset when adoptively transferred to chronically infected mice, and exhibited synergistic reduction of viral load when combined with anti-PD-L1 treatment. This study defines a unique subset of exhausted CD8(+) T cells that has a pivotal role in the control of viral replication during chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Germinal Center/cytology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Receptors, CXCR5/metabolism , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation , Chronic Disease , Female , Germinal Center/immunology , HIV Infections/immunology , HIV Infections/virology , Humans , Inhibitor of Differentiation Protein 2/metabolism , Lymphocytic choriomeningitis virus/growth & development , Male , Mice , Receptors, CXCR5/deficiency , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , Viral Load/immunology , Virus Replication/immunology
8.
Clin Exp Immunol ; 185(1): 22-32, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26990531

ABSTRACT

The recruitment of immune cells to sites of tissue inflammation is orchestrated by chemokine/chemokine receptor networks. Among these, the CXCL13/CXCR5 axis is thought to be involved critically in systemic lupus erythematosus (SLE) and lupus nephritis pathogenesis. Beyond B cell abnormalities, another hallmark of SLE disease is the occurrence of aberrant T cell responses. In particular, double-negative (DN) T cells are expanded in the peripheral blood of patients with SLE and in lupus-prone mice. DN T cells induce immunoglobulin production, secrete proinflammatory cytokines and infiltrate inflamed tissue, including kidneys. We aimed to investigate how CXCR5 deficiency changes immune cell trafficking in murine lupus. We therefore crossed CXCR5(-/-) mice with B6/lpr mice, a well-established murine lupus model. B cell numbers and B cellular immune responses were diminished in CXCR5-deficient B6/lpr mice. In addition, we observed reduced accumulation of DN T cells in spleen and lymph nodes, paralleled by reduced splenomegaly and lymphadenopathy. In-vivo migration assays revealed reduced migration of CXCR5-deficient DN T cells into lymph nodes, and ex-vivo-activated CXCR5-deficient DN T cells failed to infiltrate kidneys of recipients. Moreover, DN T cells and B cells of CXCR5-deficient B6/lpr mice failed to migrate towards CXCL13 in vitro. We propose that CXCR5 is involved critically in B cell trafficking and germinal cell (GC) formation in murine lupus and in guiding pathogenic DN T cells into lymphoid organs and kidneys, and we therefore describe new pathomechanisms for the CXCL13/CXCR5 axis in SLE.


Subject(s)
B-Lymphocytes/immunology , Kidney/immunology , Lupus Nephritis/immunology , Lymphadenopathy/immunology , Receptors, CXCR5/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/pathology , Cell Movement , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Disease Models, Animal , Disease Progression , Female , Gene Expression Regulation/immunology , Humans , Kidney/pathology , Lupus Nephritis/genetics , Lupus Nephritis/mortality , Lupus Nephritis/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphadenopathy/genetics , Lymphadenopathy/mortality , Lymphadenopathy/pathology , Male , Mice , Mice, Transgenic , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , Signal Transduction , Spleen/immunology , Spleen/pathology , Survival Analysis , T-Lymphocytes/pathology
9.
Nat Cell Biol ; 17(5): 580-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25915127

ABSTRACT

The generation of haematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) will depend on the accurate recapitulation of embryonic haematopoiesis. In the early embryo, HSCs develop from the haemogenic endothelium (HE) and are specified in a Notch-dependent manner through a process named endothelial-to-haematopoietic transition (EHT). As HE is associated with arteries, it is assumed that it represents a subpopulation of arterial vascular endothelium (VE). Here we demonstrate at a clonal level that hPSC-derived HE and VE represent separate lineages. HE is restricted to the CD34(+)CD73(-)CD184(-) fraction of day 8 embryoid bodies and it undergoes a NOTCH-dependent EHT to generate RUNX1C(+) cells with multilineage potential. Arterial and venous VE progenitors, in contrast, segregate to the CD34(+)CD73(med)CD184(+) and CD34(+)CD73(hi)CD184(-) fractions, respectively. Together, these findings identify HE as distinct from VE and provide a platform for defining the signalling pathways that regulate their specification to functional HSCs.


Subject(s)
Arteries/physiology , Cell Differentiation , Cell Lineage , Endothelial Progenitor Cells/physiology , Hematopoietic Stem Cells/physiology , Multipotent Stem Cells/physiology , Pluripotent Stem Cells/physiology , 5'-Nucleotidase/deficiency , Antigens, CD34/metabolism , Arteries/cytology , Arteries/metabolism , Biomarkers/metabolism , Cell Line , Cell Separation/methods , Coculture Techniques , Core Binding Factor Alpha 2 Subunit/metabolism , Endothelial Progenitor Cells/metabolism , GPI-Linked Proteins/deficiency , Hematopoietic Stem Cells/metabolism , Humans , Microscopy, Video , Multipotent Stem Cells/metabolism , Phenotype , Pluripotent Stem Cells/metabolism , Precursor Cells, T-Lymphoid/physiology , Receptors, CXCR5/deficiency , Receptors, Notch/metabolism , Signal Transduction , Time Factors , Veins/cytology , Veins/physiology
10.
J Neuroinflammation ; 11: 31, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-24528805

ABSTRACT

BACKGROUND: The process of neurogenesis in which new neurons are generated by proliferation and differentiation of neural stem/progenitor cells (NSCs/NPCs) has been a topic of intensive recent investigation. Investigations of the factors which regulate this process have recently begun to include immune factors including immune cells and cytokines, however the class of immune proteins designated as chemokines have been relatively neglected. Increasing evidence for novel brain-specific mechanisms of chemokines beyond their classical chemotactic functions has suggested that they may play a role in the regulation of NSC/NPC biology. METHODS: We have investigated the role of the chemokine receptor CXCR5 (ligand is CXCL13) in the activity of these cells through neurobiological and behavioural analysis of CXCR5-deficient mice (CXCR5-/-). These investigations included: immunohistochemistry for the markers Ki67, nestin, doublecortin, and IBA-1, neurosphere assays, and the baseline behavioural tests: open field test and sucrose preference test. RESULTS: We observed a significant increase in doublecortin and nestin staining in the hippocampal dentate gyrus (P = 0.02 and P = 0.0008, respectively) of CXCR5-/- animals as compared to wild-type controls. This was accompanied by a decrease in Ki67 staining subgranular zone (P = 0.009). Behavioural correlates included a significant increase in baseline locomotor activity in an open field test (P <0.00018) and a decrease in stress reactivity in that test (P = 0.015). Deficiency in CXCR5 was not associated with alterations in hippocampal microglial density, microglial activation or systemic cytokine levels, nor with loss of NSC/NPC populations in the neurosphere assay. CONCLUSIONS: These findings are the first to describe a brain-specific function of CXCR5 under physiological conditions. CXCR5 reduces maintenance of immature neural cell populations and enhances proliferation of subgranular zone cells in the hippocampal dentate gyrus, however the mechanism of these effects remains unclear. Further research into the regulation of NSC/NPC activity should consider investigation of CXCR5 and other chemokines which may be relevant to the pathophysiology of psychiatric disorders including depression, anxiety and cognitive impairment/dementia.


Subject(s)
Cell Proliferation , Dentate Gyrus/cytology , Neural Stem Cells/physiology , Receptors, CXCR5/deficiency , Animals , Calcium-Binding Proteins/metabolism , Cerebral Ventricles/cytology , Cytokines/metabolism , Doublecortin Domain Proteins , Exploratory Behavior/physiology , Female , Food Preferences/physiology , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nestin/metabolism , Neuropeptides/metabolism , Receptors, CXCR5/genetics , Saccharin/administration & dosage , Sweetening Agents/administration & dosage
11.
PLoS Biol ; 11(10): e1001672, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24130458

ABSTRACT

Lymph node (LN) stromal cells provide survival signals and adhesive substrata to lymphocytes. During an immune response, B cell follicles enlarge, questioning how LN stromal cells manage these cellular demands. Herein, we used a murine fate mapping system to describe a new stromal cell type that resides in the T cell zone of resting LNs. We demonstrated that upon inflammation, B cell follicles progressively trespassed into the adjacent T cell zone and surrounded and converted these stromal cells into CXCL13 secreting cells that in return delineated the new boundaries of the growing follicle. Acute B cell ablation in inflamed LNs abolished CXCL13 secretion in these cells, while LT-ß deficiency in B cells drastically affected this conversion. Altogether, we reveal the existence of a dormant stromal cell subset that can be functionally awakened by B cells to delineate the transient boundaries of their expanding territories upon inflammation.


Subject(s)
B-Lymphocytes/pathology , Inflammation/immunology , Inflammation/pathology , Animals , Chemokine CXCL13/metabolism , Dendritic Cells/pathology , Fibroblasts/pathology , Lymph Nodes/pathology , Lymphocyte Depletion , Lymphocytes/pathology , Mice , Receptors, CXCR5/deficiency , Receptors, CXCR5/metabolism , Receptors, Complement 3d/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , T-Lymphocytes
12.
Eur J Immunol ; 43(9): 2349-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23749374

ABSTRACT

Chronic viral infections lead to CD8(+) T cell exhaustion, characterized by impaired cytokine secretion. The presence of the immune-regulatory cytokine IL-10 promotes chronic lymphocytic choriomeningitis virus (LCMV) Clone 13 infection in mice, whereas the absence of IL-10/IL-10R signaling early during infection results in viral clearance and higher percentages and numbers of antiviral, cytokine-producing T cells. However, it is currently unclear which cell populations and effector molecules are crucial to protect against chronic infection. In this study, we demonstrate that antiviral, LCMV-binding, non-neutralizing antibodies are needed, in addition to CD4(+) and CD8(+) T cells, to clear a high-dose LCMV infection in mice, in the absence of IL-10. The interaction between CD4(+) T cells and B cells in B-cell follicles via CD40/CD40L, in addition to class switch and/or somatic hypermutation, is crucial for viral control in the absence of IL-10. Interestingly, transfer of LCMV-binding non-neutralizing antibodies protected recipients from chronic infection. In addition, viral clearance in the absence of IL-10R signaling was independent of activating Fcγ receptors and complement. These data highlight that non-neutralizing antibodies effectively contribute to the control of LCMV infection when present prior to infection, suggesting that the induction of neutralizing antibodies is not implicitly necessary for the generation of successful vaccines.


Subject(s)
Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-10/metabolism , Lymphocytic choriomeningitis virus/immunology , Animals , B-Lymphocytes , Complement System Proteins/immunology , Immunoglobulin Class Switching/immunology , Interleukin-10/deficiency , Interleukin-10/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , Receptors, IgG/deficiency , Receptors, IgG/genetics , Viral Load/immunology
13.
J Immunol ; 187(9): 4621-8, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21948984

ABSTRACT

Migration of B cells within lymphoid follicles is controlled by the chemokine receptors CXCR5 and CCR7 and the G-protein-coupled receptor EBI2 (GPR183). Whereas CXCR5 and CCR7 are known to mediate migration toward their respective chemokine ligands, it is unclear whether EBI2 acts by modulating these processes or by directly mediating chemotaxis toward its own spatially restricted ligand. It is also unknown how signals from these three receptors are integrated to control B cell localization. To answer these questions, we generated compound knockout mice deficient in expression of EBI2, CXCR5, or CCR7. Analysis of these mice revealed that EBI2 mediates B cell migration toward the outer areas of follicles and to bridging channels of the spleen independent of both CXCR5 and CCR7. Migratory signals delivered by EBI2 were shown to control B cell organization within the spleen and to be particularly important for positioning activated B cells in the early stages of Ab responses. An additional minor role for EBI2 was identified in the organization and affinity maturation of B cells in germinal centers. Thus, EBI2-mediated chemotaxis provides a third dimension to B cell migration that balances and integrates with the inputs from CXCR5 and CCR7 to determine B cell positioning.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Communication/immunology , Chemotaxis, Leukocyte/immunology , Germinal Center/immunology , Receptors, CCR7/physiology , Receptors, CXCR5/physiology , Receptors, G-Protein-Coupled/physiology , Animals , B-Lymphocyte Subsets/cytology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Communication/genetics , Chemotaxis, Leukocyte/genetics , Germinal Center/cytology , Germinal Center/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR7/deficiency , Receptors, CXCR5/deficiency , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/deficiency , Signal Transduction/genetics , Signal Transduction/immunology
14.
J Immunol ; 182(5): 2610-9, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234155

ABSTRACT

Solitary intestinal lymphoid tissue (SILT) comprises a spectrum of phenotypically diverse lymphoid aggregates interspersed throughout the small intestinal mucosa. Manifestations of SILT range from tiny lymphoid aggregates almost void of mature lymphocytes to large structures dominated by B cells. Large SILT phenotypically resemble a single Peyer's patch follicle, suggesting that SILT might contribute to intestinal humoral immune responses. In this study, we track the fate of individual SILT in vivo over time and analyze SILT formation and function in chemokine receptor CXCR5-deficient mice. We show that, in analogy to Peyer's patches, formation of SILT is invariantly determined during ontogeny and depends on CXCR5. Young CXCR5-deficient mice completely lack SILT, suggesting that CXCR5 is essential for SILT formation during regular postnatal development. However, microbiota and other external stimuli can induce the formation of aberrant SILT distinguished by impaired development of B cell follicles in CXCR5-deficient mice. Small intestinal transplantation and bone marrow transplantation reveal that defect follicle formation is due to impaired B cell homing. Moreover, oral immunization with cholera toxin or infection with noninvasive Salmonella fail to induce efficient humoral immune responses in CXCR5-deficient mice. Bone marrow transplantation of CXCR5-deficient recipients with wild-type bone marrow rescued B cell follicle formation in SILT but failed to restore full humoral immune responses. These results reveal an essential role of CXCR5 in Peyer's patch and SILT development and function and indicate that SILT do not fully compensate for the lack of Peyer's patches in T cell-dependent humoral immune responses.


Subject(s)
B-Lymphocytes/immunology , Chemotaxis, Leukocyte/immunology , Immunoglobulin A/biosynthesis , Intestinal Mucosa/immunology , Lymphoid Tissue/immunology , Receptors, CXCR5/physiology , Administration, Oral , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Bone Marrow Transplantation/immunology , Cholera Toxin/therapeutic use , Intestinal Mucosa/metabolism , Lymphoid Tissue/growth & development , Lymphoid Tissue/metabolism , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/pathology , Salmonella typhimurium/immunology , T-Lymphocytes/immunology
15.
J Immunol ; 179(8): 5099-108, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17911595

ABSTRACT

Th cell access to primary B cell follicles is dependent on CXCR5. However, whether CXCR5 induction on T cells is sufficient in determining their follicular positioning has been unclear. In this study, we find that transgenic CXCR5 overexpression is not sufficient to promote follicular entry of naive T cells unless the counterbalancing influence of CCR7 ligands is removed. In contrast, the positioning of Ag-engaged T cells at the B/T boundary could occur in the absence of CXCR5. The germinal center (GC) response was 2-fold reduced when T cells lacked CXCR5, although these T cells were able to access the GC. Finally, CXCR5(high)CCR7(low) T cells were found to have elevated IL-4 transcript and programmed cell death gene-1 (PD-1) expression, and PD-1(high) cells were reduced in the absence of T cell CXCR5 or in mice compromised in GC formation. Overall, these findings provide further understanding of how the changes in CXCR5 and CCR7 expression regulate Th cell positioning during Ab responses, and they suggest that development and/or maintenance of a PD-1(high) follicular Th cell subset is dependent on appropriate interaction with GC B cells.


Subject(s)
Antigens, Differentiation/biosynthesis , Chemotaxis, Leukocyte/immunology , Germinal Center/immunology , Lymphoid Tissue/immunology , Receptors, CCR7/physiology , Receptors, CXCR5/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , Chemotaxis, Leukocyte/genetics , Germinal Center/cytology , Germinal Center/metabolism , Lymphocyte Activation/genetics , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Programmed Cell Death 1 Receptor , Receptors, CCR7/biosynthesis , Receptors, CXCR5/biosynthesis , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL