Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Autoimmun ; 123: 102684, 2021 09.
Article in English | MEDLINE | ID: mdl-34237649

ABSTRACT

OBJECTIVE: B-cells are present in the inflamed arteries of giant cell arteritis (GCA) patients and a disturbed B-cell homeostasis is reported in peripheral blood of both GCA and the overlapping disease polymyalgia rheumatica (PMR). In this study, we aimed to investigate chemokine-chemokine receptor axes governing the migration of B-cells in GCA and PMR. METHODS: We performed Luminex screening assay for serum levels of B-cell related chemokines in treatment-naïve GCA (n = 41), PMR (n = 31) and age- and sex matched healthy controls (HC, n = 34). Expression of chemokine receptors on circulating B-cell subsets were investigated by flow cytometry. Immunohistochemistry was performed on GCA temporal artery (n = 14) and aorta (n = 10) and on atherosclerosis aorta (n = 10) tissue. RESULTS: The chemokines CXCL9 and CXCL13 were significantly increased in the circulation of treatment-naïve GCA and PMR patients. CXCL13 increased even further after three months of glucocorticoid treatment. At baseline CXCL13 correlated with disease activity markers. Peripheral CXCR3+ and CXCR5+ switched memory B-cells were significantly reduced in both patient groups and correlated inversely with their complementary chemokines CXCL9 and CXCL13. At the arterial lesions in GCA, CXCR3+ and CXCR5+ B-cells were observed in areas with high CXCL9 and CXCL13 expression. CONCLUSION: Changes in systemic and local chemokine and chemokine receptor pathways related to B-cell migration were observed in GCA and PMR mainly in the CXCL9-CXCR3 and CXCL13-CXCR5 axes. These changes can contribute to homing and organization of B-cells in the vessel wall and provide further evidence for an active involvement of B-cells in GCA and PMR.


Subject(s)
B-Lymphocytes/physiology , Chemokines/physiology , Giant Cell Arteritis/immunology , Polymyalgia Rheumatica/immunology , Aged , Aged, 80 and over , Cell Movement , Chemokine CXCL13/blood , Chemokine CXCL13/physiology , Chemokine CXCL9/blood , Chemokine CXCL9/physiology , Female , Giant Cell Arteritis/etiology , Humans , Male , Middle Aged , Polymyalgia Rheumatica/etiology , Receptors, CXCR3/blood , Receptors, CXCR3/physiology , Receptors, CXCR5/blood , Receptors, CXCR5/physiology
2.
Lab Invest ; 101(2): 228-244, 2021 02.
Article in English | MEDLINE | ID: mdl-32994482

ABSTRACT

Homeostasis of the retinal pigment epithelium (RPE) is essential for the health and proper function of the retina. Regulation of RPE homeostasis is, however, largely unexplored, yet dysfunction of this process may lead to retinal degenerative diseases, including age-related macular degeneration (AMD). Here, we report that chemokine receptor CXCR5 regulates RPE homeostasis through PI3K/AKT signaling and by suppression of FOXO1 activation. We used primary RPE cells isolated from CXCR5-deficient mice and wild type controls, as well as ex vivo RPE-choroidal-scleral complexes (RCSC) to investigate the regulation of homeostasis. CXCR5 expression in mouse RPE cells was diminished by treatment with hydrogen peroxide. Lack of CXCR5 expression leads to an abnormal cellular shape, pigmentation, decreased expression of the RPE differentiation marker RPE65, an increase in the undifferentiated progenitor marker MITF, and compromised RPE barrier function, as well as compromised cell-to-cell interaction. An increase in epithelial-mesenchymal transition (EMT) markers (αSMA, N-cadherin, and vimentin) was noted in CXCR5-deficient RPE cells both in vitro and in age-progression specimens of CXCR5-/- mice (6, 12, 24-months old). Deregulated autophagy in CXCR5-deficient RPE cells was observed by decreased LC3B-II, increased p62, abnormal autophagosomes, and impaired lysosome enzymatic activity as shown by GFP-LC3-RFP reporter plasmid. Mechanistically, deficiency in CXCR5 resulted in the downregulation of PI3K and AKT signaling, but upregulation and nuclear localization of FOXO1. Additionally, inhibition of PI3K in RPE cells resulted in an increased expression of FOXO1. Inhibition of FOXO1, however, reverts the degradation of ZO-1 caused by CXCR5 deficiency. Collectively, these findings suggest that CXCR5 maintains PI3K/AKT signaling, which controls FOXO1 activation, thereby regulating the expression of genes involved in RPE EMT and autophagy deregulation.


Subject(s)
Receptors, CXCR5 , Retinal Pigment Epithelium , Animals , Autophagy/genetics , Cells, Cultured , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Inflammation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Receptors, CXCR5/physiology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/physiopathology , Signal Transduction/genetics
3.
Int J Mol Sci ; 21(17)2020 Aug 24.
Article in English | MEDLINE | ID: mdl-32847038

ABSTRACT

Osteosarcoma is the most common primary tumor of the skeletal system and is well-known to have an aggressive clinical outcome and high metastatic potential. The chemokine (C-X-C motif) ligand 13 (CXCL13) plays a vital role in the development of several cancers. However, the effect of CXCL13 in the motility of osteosarcoma cells remains uncertain. Here, we found that CXCL13 increases the migration and invasion potential of three osteosarcoma cell lines. In addition, CXCL13 expression was upregulated in migration-prone MG-63 cells. Vascular cell adhesion molecule 1 (VCAM-1) siRNA and antibody demonstrated that CXCL13 promotes migration via increasing VCAM-1 production. We also show that CXCR5 receptor controls CXCL13-mediated VCAM-1 expression and cell migration. Our study identified that CXCL13/CXCR5 axis facilitate VCAM-1 production and cell migration in human osteosarcoma via the phospholipase C beta (PLCß), protein kinase C α (PKCα), c-Src, and nuclear factor-κB (NF-κB) signaling pathways. CXCL13 and CXCR5 appear to be a novel therapeutic target in metastatic osteosarcoma.


Subject(s)
Bone Neoplasms/pathology , Cell Movement/genetics , Chemokine CXCL13/metabolism , Osteosarcoma/pathology , Receptors, CXCR5/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Chemokine CXCL13/physiology , Humans , Neoplasm Invasiveness/genetics , Osteosarcoma/genetics , Osteosarcoma/metabolism , Protein Binding , Receptors, CXCR5/physiology , Signal Transduction/physiology , Tumor Cells, Cultured , Vascular Cell Adhesion Molecule-1/metabolism
5.
PLoS Pathog ; 15(4): e1007719, 2019 04.
Article in English | MEDLINE | ID: mdl-30973939

ABSTRACT

The regulation of mucosal immune function is critical to host protection from enteric pathogens but is incompletely understood. The nervous system and the neurotransmitter acetylcholine play an integral part in host defense against enteric bacterial pathogens. Here we report that acetylcholine producing-T-cells, as a non-neuronal source of ACh, were recruited to the colon during infection with the mouse pathogen Citrobacter rodentium. These ChAT+ T-cells did not exclusively belong to one Th subset and were able to produce IFNγ, IL-17A and IL-22. To interrogate the possible protective effect of acetylcholine released from these cells during enteric infection, T-cells were rendered deficient in their ability to produce acetylcholine through a conditional gene knockout approach. Significantly increased C. rodentium burden was observed in the colon from conditional KO (cKO) compared to WT mice at 10 days post-infection. This increased bacterial burden in cKO mice was associated with increased expression of the cytokines IL-1ß, IL-6, and TNFα, but without significant changes in T-cell and ILC associated IL-17A, IL-22, and IFNγ, or epithelial expression of antimicrobial peptides, compared to WT mice. Despite the increased expression of pro-inflammatory cytokines during C. rodentium infection, inducible nitric oxide synthase (Nos2) expression was significantly reduced in intestinal epithelial cells of ChAT T-cell cKO mice 10 days post-infection. Additionally, a cholinergic agonist enhanced IFNγ-induced Nos2 expression in intestinal epithelial cell in vitro. These findings demonstrated that acetylcholine, produced by specialized T-cells that are recruited during C. rodentium infection, are a key mediator in host-microbe interactions and mucosal defenses.


Subject(s)
Acetylcholine/metabolism , Citrobacter rodentium/immunology , Colon/immunology , Enterobacteriaceae Infections/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Colon/metabolism , Cytokines/metabolism , Enterobacteriaceae Infections/microbiology , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR5/physiology
6.
Life Sci ; 227: 175-186, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-31026453

ABSTRACT

The tumor microenvironment comprises stromal and tumor cells which interact with each other through complex cross-talks that are mediated by a variety of growth factors, cytokines, and chemokines. The chemokine ligand 13 (CXCL13) and its chemokine receptor 5 (CXCR5) are among the key chemotactic factors which play crucial roles in deriving cancer cell biology. CXCL13/CXCR5 signaling axis makes pivotal contributions to the development and progression of several human cancers. In this review, we discuss how CXCL13/CXCR5 signaling modulates cancer cell ability to grow, proliferate, invade, and metastasize. Furthermore, we also discuss the preliminary evidence on context-dependent functioning of this axis within the tumor-immune microenvironment, thus, highlighting its potential dichotomy with respect to anticancer immunity and cancer immune-evasion mechanisms. At the end, we briefly shed light on the therapeutic potential or implications of targeting CXCL13/CXCR5 axis within the tumor microenvironment.


Subject(s)
Chemokine CXCL13/metabolism , Neoplasms/physiopathology , Receptors, CXCR5/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL13/physiology , Disease Progression , Humans , Neoplasms/metabolism , Receptors, CXCR5/physiology , Signal Transduction , Tumor Microenvironment
7.
Inflammation ; 40(3): 762-769, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28155010

ABSTRACT

Recent data demonstrated that chemokine CXCL13 mediates neuroinflammation and contributes to the maintenance of neuropathic pain after nerve injury in the spinal cord. Pro-nociceptive chemokines activate mitogen-activated protein kinases (MAPKs) which are potential signaling pathways contributing to the nociceptive behavior in inflammatory or neuropathic pain. However, whether activation of p38 and JNK MAPK signaling pathway in the trigeminal ganglion (TG) are involved in CXCL13 and its receptor CXCR5-mediated orofacial pain has not yet been clarified. Here, we show that the unilateral partial infraorbital nerve ligation (pIONL) induced a profound orofacial pain in wild-type (WT) mice. Western blot results showed that pIONL induced p38 but not JNK activation in the TG of WT mice. However, the orofacial pain induced by pIONL was alleviated in Cxcr5 -/- mice, and the activation of p38 was also abrogated in Cxcr5 -/- mice. Furthermore, intra-TG injection of CXCL13 evoked mechanical hypersensitivity and increased p-p38 expression in WT mice. But CXCL13 had no effect on pain behavior or p-p38 expression in Cxcr5 -/- mice. Finally, pretreatment with p38 inhibitor, SB203580, attenuated the pIONL-induced mechanical allodynia and decreased the mRNA expression of pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) in the TG. Taken together, our data suggest that CXCL13 acts on CXCR5 to increase p38 activation and further contributes to the pathogenesis of orofacial neuropathic pain.


Subject(s)
Chemokine CXCL13/physiology , Eye Injuries/metabolism , Facial Pain/etiology , Trigeminal Ganglion/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Eye/innervation , Eye Injuries/pathology , Mice , Neurogenic Inflammation/etiology , Receptors, CXCR5/physiology
8.
Int J Med Sci ; 12(11): 853-61, 2015.
Article in English | MEDLINE | ID: mdl-26640404

ABSTRACT

BACKGROUND: Chemokine and chemokine receptors could have played an important role in tumor angiogenesis and distant metastasis. The mechanism of inflammation, expression and function of chemokines and chemokine receptors in benign prostatic hyperplasia (BPH) and prostate cancer (PCa) remain unclear. The purpose of present study is to detect differential expression and function of chemokines and chemokine receptors (CCRs) in BPH and PCa. METHODS: BPH-1 and peripheral blood mononuclear cells (PBMCs) were co-cultured in Transwell chambers, and human normal prostate (NP) tissues, BPH tissues and PCa tissues were collected. CCR gene-chips were used to analyze and compare the differential expression of CCRs in BPH-1 cells, BPH-1 cells co-cultured with PBMCs, and LNCaP cells. The differential expression of CCRs was detected and validated using real-time PCR, western blotting and immunofluorescence (IF). The proliferation of LNCaP cells was also investigated after the knockdown CXCR5. RESULTS: RESULTS of gene-chips indicated that there was low or no expression of CCR10, CXCR1, CXCR3 and CXCR5 in BPH-1 cells, whereas the expression of these receptors in BPH-1 cells was increased by PBMCs, and the expression was high in LNCaP cells. Furthermore, real-time PCR and western blotting confirmed the above mentioned results. IF verified no or low expression of CXCR1, CXCR3 and CXCR5 in NP tissues, low or moderate expression in BPH and high expression in PCa. However, CCR10 was not expressed at detectable levels in the three groups. The growth and proliferation of LNCaP cells was markedly inhibited after down-regulation of CXCR5. CONCLUSIONS: PCa cells expressed high levels of CCR10, CXCR1, CXCR3 and CXCR5. Although BPH cells did not express these factors, their expression was up-regulated when BPH-1 cells were incubated with inflammatory cells. Finally, down-regulation of CXCR5 inhibited the growth and proliferation of LNCaP cells.


Subject(s)
Prostatic Hyperplasia/immunology , Prostatic Neoplasms/immunology , Receptors, CXCR5/physiology , Cell Line, Tumor , Humans , Male , Prostate/immunology , RNA, Small Interfering/genetics , Receptors, CXCR5/analysis , Receptors, CXCR5/genetics
9.
Trends Immunol ; 36(12): 758-760, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26588880

ABSTRACT

Three recent studies provide new insight into the mechanisms that promote the differentiation of antigen-primed T cells into the follicular helper T (Tfh) lineage, revealing a key role for the transcription factor TCF-1 in the regulation of the Bcl6-Blimp-1 axis that mediates the divergence between the Tfh and the Th1 lineages.


Subject(s)
T Cell Transcription Factor 1/metabolism , Th1 Cells/metabolism , Th1 Cells/physiology , Animals , Cell Differentiation/physiology , Receptors, CXCR5/physiology , Transcription Factors/metabolism
10.
Mediators Inflamm ; 2015: 638968, 2015.
Article in English | MEDLINE | ID: mdl-26300592

ABSTRACT

Follicular helper CD4(+) T (TFH) cells play a fundamental role in humoral immunity deriving from their ability to provide help for germinal center (GC) formation, B cell differentiation into plasma cells and memory cells, and antibody production in secondary lymphoid tissues. TFH cells can be identified by a combination of markers, including the chemokine receptor CXCR5, costimulatory molecules ICOS and PD-1, transcription repressor Bcl-6, and cytokine IL-21. It is difficult and impossible to get access to secondary lymphoid tissues in humans, so studies are usually performed with human peripheral blood samples as circulating counterparts of tissue TFH cells. A balance of TFH cell generation and function is critical for protective antibody response, whereas overactivation of TFH cells or overexpression of TFH-associated molecules may result in autoimmune diseases. Emerging data have shown that TFH cells and TFH-associated molecules may be involved in the pathogenesis of neuroautoimmune diseases including multiple sclerosis (MS), neuromyelitis optica (NMO)/neuromyelitis optica spectrum disorders (NMOSD), and myasthenia gravis (MG). This review summarizes the features of TFH cells, including their development, function, and roles as well as TFH-associated molecules in neuroautoimmune diseases and their animal models.


Subject(s)
Autoimmune Diseases of the Nervous System/etiology , CD4-Positive T-Lymphocytes/physiology , Animals , Autoimmune Diseases of the Nervous System/immunology , DNA-Binding Proteins/physiology , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Inducible T-Cell Co-Stimulator Protein/physiology , Interleukins/physiology , Multiple Sclerosis/etiology , Multiple Sclerosis/immunology , Myasthenia Gravis/etiology , Myasthenia Gravis/immunology , Neuromyelitis Optica/etiology , Neuromyelitis Optica/immunology , Proto-Oncogene Proteins c-bcl-6 , Receptors, CXCR5/physiology
11.
Neurobiol Dis ; 83: 1-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26299391

ABSTRACT

Ischemic stroke triggers neurogenesis from neural stem/progenitor cells (NSPCs) in the subventricular zone (SVZ) and migration of newly formed neuroblasts toward the damaged striatum where they differentiate to mature neurons. Whether it is the injury per se or the associated inflammation that gives rise to this endogenous neurogenic response is unknown. Here we showed that inflammation without corresponding neuronal loss caused by intrastriatal lipopolysaccharide (LPS) injection leads to striatal neurogenesis in rats comparable to that after a 30 min middle cerebral artery occlusion, as characterized by striatal DCX+ neuroblast recruitment and mature NeuN+/BrdU+ neuron formation. Using global gene expression analysis, changes in several factors that could potentially regulate striatal neurogenesis were identified in microglia sorted from SVZ and striatum of LPS-injected and stroke-subjected rats. Among the upregulated factors, one chemokine, CXCL13, was found to promote neuroblast migration from neonatal mouse SVZ explants in vitro. However, neuroblast migration to the striatum was not affected in constitutive CXCL13 receptor CXCR5(-/-) mice subjected to stroke. Infarct volume and pro-inflammatory M1 microglia/macrophage density were increased in CXCR5(-/-) mice, suggesting that microglia-derived CXCL13, acting through CXCR5, might be involved in neuroprotection following stroke. Our findings raise the possibility that the inflammation accompanying an ischemic insult is the major inducer of striatal neurogenesis after stroke.


Subject(s)
Corpus Striatum/physiopathology , Encephalitis/physiopathology , Infarction, Middle Cerebral Artery/physiopathology , Neural Stem Cells/physiology , Neurogenesis , Neurons/physiology , Stroke/physiopathology , Animals , Cell Death , Cell Movement/drug effects , Chemokine CXCL13/pharmacology , Chemokine CXCL13/physiology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Doublecortin Protein , Encephalitis/chemically induced , Encephalitis/metabolism , Gene Expression , Infarction, Middle Cerebral Artery/pathology , Inflammation Mediators/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Lateral Ventricles/physiopathology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/metabolism , Neurons/pathology , Rats , Rats, Wistar , Receptors, CXCR5/genetics , Receptors, CXCR5/physiology , Stroke/pathology
12.
Tumour Biol ; 35(6): 5273-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24519063

ABSTRACT

Immune dysregulation plays a key role in the development of osteosarcoma (OS). Peripheral blood CD4+CXCR5+ T cells can induce B-cell activation and produce various cytokines and therefore may play critical roles in tumorigenesis. The purpose of the study was to investigate changes of peripheral CD4+CXCR5+ T cells in OS. Peripheral CD4+CXCR5+ T cells and its subtypes were determined by measuring CD3, CD4, CXCR5, CXCR3, and CCR6 in 38 OS patients and 42 healthy controls using flow cytometry. Data demonstrated that percentage of peripheral CD4+CXCR5+ T cells was significantly increased in OS patients (13.9 %) than in controls (8.6 %, p<0.001). Further analysis identified a profound skewing of peripheral CD4+CXCR5+ T cell subsets toward Th2 and Th17 cells in OS patients. Investigating clinical status of the patients showed that prevalence of peripheral CD4+CXCR5+ T cells was significantly elevated in cases with metastasis (17.4 %) than those without metastasis (12.7 %). Similarly, patients with high tumor grade revealed increased percentage of CD4+CXCR5+ T cells compared to those with low tumor grade (15.3 versus 11.0 %). Interestingly, the upregulation of peripheral CD4+CXCR5+ T cells in patients with metastasis or high tumor grade was contributed by Th1 and Th17 subtypes. This study suggests the involvement of peripheral CD4+CXCR5+ T cells in the pathogenesis and progression of OS and provides novel knowledge for understanding this disease.


Subject(s)
Bone Neoplasms/immunology , CD4-Positive T-Lymphocytes/immunology , Osteosarcoma/immunology , Receptors, CXCR5/physiology , Adolescent , Adult , Bone Neoplasms/etiology , Bone Neoplasms/pathology , Child , Female , Humans , Male , Neoplasm Grading , Osteosarcoma/etiology , Osteosarcoma/pathology , Receptors, CXCR5/analysis , Up-Regulation
13.
Blood ; 121(20): 4101-9, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23558016

ABSTRACT

It is not known how naive B cells compute divergent chemoattractant signals of the T-cell area and B-cell follicles during in vivo migration. Here, we used two-photon microscopy of peripheral lymph nodes (PLNs) to analyze the prototype G-protein-coupled receptors (GPCRs) CXCR4, CXCR5, and CCR7 during B-cell migration, as well as the integrin LFA-1 for stromal guidance. CXCR4 and CCR7 did not influence parenchymal B-cell motility and distribution, despite their role during B-cell arrest in venules. In contrast, CXCR5 played a nonredundant role in B-cell motility in follicles and in the T-cell area. B-cell migration in the T-cell area followed a random guided walk model, arguing against directed migration in vivo. LFA-1, but not α4 integrins, contributed to B-cell motility in PLNs. However, stromal network guidance was LFA-1 independent, uncoupling integrin-dependent migration from stromal attachment. Finally, we observed that despite a 20-fold reduction of chemokine expression in virus-challenged PLNs, CXCR5 remained essential for B-cell screening of antigen-presenting cells. Our data provide an overview of the contribution of prototype GPCRs and integrins during naive B-cell migration and shed light on the local chemokine availability that these cells compute.


Subject(s)
B-Lymphocytes/physiology , Cell Communication/physiology , Chemokines/physiology , Chemotaxis, Leukocyte/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Receptors, CCR7/physiology , Receptors, CXCR4/physiology , Receptors, CXCR5/physiology , Stromal Cells/physiology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/physiology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Communication/drug effects , Chemokines/metabolism , Chemokines/pharmacology , Chemotaxis, Leukocyte/drug effects , Female , Gene Deletion , Lymphocyte Function-Associated Antigen-1/physiology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Stromal Cells/metabolism
14.
PLoS One ; 7(11): e47487, 2012.
Article in English | MEDLINE | ID: mdl-23189125

ABSTRACT

BACKGROUND: Regulation of immune responses is critical for controlling inflammation and disruption of this process can lead to tissue damage. We reported that CXCL13 was induced in fallopian tube tissue following C. trachomatis infection. Here, we examined the influence of the CXCL13-CXCR5 axis in chlamydial genital infection. METHODOLOGY AND PRINCIPAL FINDINGS: Disruption of the CXCL13-CXCR5 axis by injecting anti-CXCL13 Ab to BALB/c mice or using Cxcr5-/- mice increased chronic inflammation in the upper genital tract (UGT; uterine horns and oviducts) after Chlamydia muridarum genital infection (GT). Further studies in Cxcr5-/- mice showed an elevation in bacterial burden in the GT and increased numbers of neutrophils, activated DCs and activated NKT cells early after infection. After resolution, we noted increased fibrosis and the accumulation of a variety of T cells subsets (CD4-IFNγ, CD4-IL-17, CD4-IL-10 & CD8-TNFα) in the oviducts. NKT cell depletion in vitro reduced IL-17α and various cytokines and chemokines, suggesting that activated NKT cells modulate neutrophils and DCs through cytokine/chemokine secretion. Further, chlamydial glycolipids directly activated two distinct types of NKT cell hybridomas in a cell-free CD1d presentation assay and genital infection of Cd1d-/- mice showed reduced oviduct inflammation compared to WT mice. CXCR5 involvement in pathology was also noted using single-nucleotide polymorphism analysis in C. trachomatis infected women attending a sub-fertility clinic. Women who developed tubal pathology after a C. trachomatis infection had a decrease in the frequency of CXCR5 SNP +10950 T>C (rs3922). CONCLUSIONS/SIGNIFICANCE: These experiments indicate that disruption of the CXCL13-CXCR5 axis permits increased activation of NKT cells by type I and type II glycolipids of Chlamydia muridarum and results in UGT pathology potentially through increased numbers of neutrophils and T cell subsets associated with UGT pathology. In addition, CXCR5 appears to contribute to inter-individual differences in human tubal pathology following C. trachomatis infection.


Subject(s)
Chemokine CXCL13/physiology , Chlamydia Infections/immunology , Chlamydia Infections/pathology , Chlamydia muridarum/immunology , Natural Killer T-Cells/immunology , Receptors, CXCR5/physiology , Reproductive Tract Infections/immunology , Reproductive Tract Infections/pathology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Chemokine CXCL13/metabolism , Chlamydia Infections/genetics , Cohort Studies , Cytokines/biosynthesis , Disease Models, Animal , Female , Humans , Lymphocyte Activation/immunology , Mice , Natural Killer T-Cells/metabolism , Polymorphism, Single Nucleotide , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Reproductive Tract Infections/genetics , Sexually Transmitted Diseases/genetics , Sexually Transmitted Diseases/immunology , Sexually Transmitted Diseases/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , White People
15.
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
16.
J Immunol ; 186(5): 2889-96, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270407

ABSTRACT

CD4 T cell help for B cells is critical for effective Ab responses. Although many of the molecules involved in helper functions of naive CD4 T cells have been characterized, much less is known about the helper capabilities of memory CD4 T cells, an important consideration for the design of vaccines that aim to prime protective memory CD4 T cells. In this study, we demonstrate that memory CD4 T cells enable B cells to expand more rapidly and class switch earlier than do primary responding CD4 T cells. This accelerated response does not require large numbers of memory cells, and similar numbers of primary responding cells provide less effective help than do memory cells. However, only memory CD4 T cells that express the B cell follicle homing molecule, CXCR5, are able to accelerate the response, suggesting that the rapidity of the Ab response depends on the ability of CD4 memory T cells to migrate quickly toward B cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunologic Memory , Receptors, CXCR5/biosynthesis , Amino Acid Sequence , Animals , B-Lymphocyte Subsets/microbiology , B-Lymphocyte Subsets/virology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Movement/immunology , Female , Immunoglobulin G/biosynthesis , Immunoglobulin G/classification , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Receptors, CXCR5/physiology , Receptors, Lymphocyte Homing/administration & dosage , Receptors, Lymphocyte Homing/biosynthesis , Receptors, Lymphocyte Homing/physiology , Resting Phase, Cell Cycle/immunology
17.
Eur J Immunol ; 39(10): 2765-78, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19757439

ABSTRACT

Nose-associated lymphoid tissue (NALT) in the rodent upper respiratory tract develops postnatally and is considered to be independent of several factors known to be involved in the organogenesis of LN and Peyer's patches (PP). In this study we demonstrate that at least two different pathways result in NALT development. Following NALT anlage formation the intrinsic pathway relies on a signaling cascade including those mediated through the chemokine receptor CXCR5 and the lymphotoxin beta receptor (LTbetaR). This allows for the formation of high endothelial venules and thereby the recruitment of lymphocytes into NALT. Alternatively, high endothelial venule formation and lymphocyte recruitment can be induced in the NALT anlage by environmental signals, which are independent of LT-betaR and chemokine receptor CXCR5 signaling but in part rely on CD40 ligand. Thus, our study identifies a novel mechanism that facilitates the rescue of NALT development at late stages in adult life independent of the canonical LTbetaR-CXCR5 signaling axis.


Subject(s)
Antigens/immunology , Lymphoid Tissue/growth & development , Lymphotoxin beta Receptor/physiology , Nasal Mucosa/growth & development , Receptors, CXCR5/physiology , Signal Transduction/immunology , Adoptive Transfer , Aging/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, Surface/metabolism , B-Lymphocytes/immunology , CD40 Ligand/immunology , Cell Adhesion Molecules/metabolism , Cell Count , Cell Movement/immunology , Germ-Free Life/immunology , Lymph Nodes/cytology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocytes/cytology , Lymphoid Tissue/blood supply , Lymphoid Tissue/pathology , Lymphotoxin-alpha/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mucoproteins , Nasal Mucosa/blood supply , Nasal Mucosa/pathology , Propionibacterium acnes/immunology , Spleen/cytology , Tumor Necrosis Factor Ligand Superfamily Member 14/antagonists & inhibitors , Venules/growth & development , Venules/metabolism , Venules/pathology
18.
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
20.
Nat Immunol ; 9(1): 54-62, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18037889

ABSTRACT

The splenic marginal zone is a site of blood flow, and the specialized B cell population that inhabits this compartment has been linked to the capture and follicular delivery of blood-borne antigens. However, the mechanism of this antigen transport has remained unknown. Here we show that marginal zone B cells were not confined to the marginal zone but continuously shuttled between the marginal zone and follicular areas, such that many of the cells visited a follicle every few hours. Migration to the follicle required the chemokine receptor CXCR5, whereas return to the marginal zone was promoted by the sphingosine 1-phosphate receptors S1P1 and S1P3. Treatment with an S1P1 antagonist caused displacement of marginal zone B cells from the marginal zone. Marginal zone-follicle shuttling of marginal zone B cells provides an efficient mechanism for systemic antigen capture and delivery to follicular dendritic cells.


Subject(s)
Antigens/metabolism , B-Lymphocytes/physiology , Animals , Antigens/blood , Biological Transport , Dendritic Cells, Follicular/immunology , Fingolimod Hydrochloride , Mice , Mice, Knockout , Propylene Glycols/pharmacology , Receptors, CXCR5/genetics , Receptors, CXCR5/physiology , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/physiology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , Spleen/cytology , Spleen/immunology
SELECTION OF CITATIONS
SEARCH DETAIL