Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Front Immunol ; 12: 763065, 2021.
Article in English | MEDLINE | ID: mdl-34868008

ABSTRACT

Neuropsychiatric lupus (NPSLE), the nervous system presentation of systemic lupus erythematosus (SLE), remains challenging to treat due to its unclear pathogenesis and lack of available targeted therapies. A potential contributor to disease progression is brain tertiary lymphoid structures (TLS); these ectopic lymphoid follicles that can develop tissue-targeted antibodies have recently been described in the MRL/lpr lupus mouse strain, a classic model for studying NPSLE. The brains of MRL/lpr mice show a significant increase of CXCL13, an important chemokine in lymphoid follicle formation and retention that may also play a role in the disease progression of NPSLE. The aim of the present study was to inhibit CXCL13 and examine the effect of this intervention on lymphoid formation and the development of neurobehavioral manifestations in lupus mice. Female MRL/lpr mice were injected with an anti-CXCL13 antibody, an IgG1 isotype-matched antibody, or PBS either three times a week for 12 weeks intraperitoneally (IP) starting at 6-8 weeks of age, or continuously intracerebroventricularly (ICV) with an osmotic pump over a two-week period starting at 15 weeks of age. Cognitive dysfunction and depression-like behavior were assessed at the end of treatment. When treatment was delivered IP, anti-CXCL13 treated mice showed significant improvement in cognitive function when compared to control treated mice. Depression-like behavior was attenuated as well. Furthermore, mice that received anti-CXCL13 by the ICV route showed similar beneficial effects. However, the extent of lymphocyte infiltration into the brain and the general composition of the aggregates were not substantively changed by anti-CXCL13 irrespective of the mode of administration. Nevertheless, analysis of brain gene expression in anti-CXCL13 treated mice showed significant differences in key immunological and neuro-inflammatory pathways that most likely explained the improvement in the behavioral phenotype. Our results indicate that CXCL13 affects the behavioral manifestations in the MRL/lpr strain and is important to the pathogenesis of murine NPSLE, suggesting it as a potential therapeutic target.


Subject(s)
Chemokine CXCL13/antagonists & inhibitors , Lupus Vasculitis, Central Nervous System/drug therapy , Animals , Brain/pathology , Chemokine CXCL13/physiology , Cognition/drug effects , Female , Injections, Intraventricular , Lupus Vasculitis, Central Nervous System/pathology , Lupus Vasculitis, Central Nervous System/psychology , Mice , Mice, Inbred MRL lpr , STAT3 Transcription Factor/physiology
2.
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
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
4.
J Neuroimmunol ; 338: 577087, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31715459

ABSTRACT

To evaluate occurrence and extent of CSF CXCL13 elevations beyond Lyme neuroborreliosis, we investigated CXCL13 in an unselected patient cohort with neuroinflammatory disease. From March 2016 to March 2017, 180 in-patients with CSF pleocytosis were categorized into following groups: pyogenic CNS infections, aseptic meningoencephalitis, neuroimmunological diseases, and reactive pleocytosis. We provide evidence that CXCL13 elevation occurs at variable extent in the majority of neuroinflammatory diseases. The exact role of CXCL13 in CSF is elusive, but the broad occurrence in neuroinflammation points at CNS immune activation, which is not exclusive for LNB.


Subject(s)
Chemokine CXCL13/cerebrospinal fluid , Lyme Neuroborreliosis/cerebrospinal fluid , Adolescent , Adult , Aged , Aged, 80 and over , Chemokine CXCL13/physiology , Female , Humans , Leukocytosis/cerebrospinal fluid , Leukocytosis/immunology , Lyme Neuroborreliosis/immunology , Male , Meningoencephalitis/cerebrospinal fluid , Meningoencephalitis/immunology , Middle Aged , Young Adult
6.
J Immunol ; 203(9): 2415-2424, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31570507

ABSTRACT

Bacterial, parasitic, and viral infections are well-known causes of lymphoid tissue disorganization, although the factors, both host and/or pathogen derived, that mediate these changes are largely unknown. Ehrlichia muris infection in mice causes a loss of germinal center (GC) B cells that is accompanied by the generation of extrafollicular T-bet+ CD11c+ plasmablasts and IgM memory B cells. We addressed a possible role for TNF-α in this process because this cytokine has been shown to regulate GC development. Ablation of TNF-α during infection resulted in an 8-fold expansion of GL7+ CD38lo CD95+ GC B cells, and a 2.5- and 5-fold expansion of CD138+ plasmablasts and T-bet+ memory cells, respectively. These changes were accompanied by a reduction in splenomegaly, more organized T and B cell zones, and an improved response to Ag challenge. CXCL13, the ligand for CXCR5, was detected at 6-fold higher levels following infection but was much reduced following TNF-α ablation, suggesting that CXCL13 dysregulation also contributes to loss of lymphoid tissue organization. T follicular helper cells, which also underwent expansion in infected TNF-α--deficient mice, may also have contributed to the expansion of T-bet+ B cells, as the latter are known to require T cell help. Our findings contrast with previously described roles for TNF-α in GCs and reveal how host-pathogen interactions can induce profound changes in cytokine and chemokine production that can alter lymphoid tissue organization, GC B cell development, and extrafollicular T-bet+ B cell generation.


Subject(s)
B-Lymphocytes/immunology , Bacterial Infections/immunology , Germinal Center/immunology , Lymphoid Tissue/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Chemokine CXCL13/physiology , Immune Tolerance , Immunologic Memory , Lymphoid Tissue/immunology , Mice , Mice, Inbred C57BL
7.
J Immunol ; 203(9): 2401-2414, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31548329

ABSTRACT

Ligand-engaged chemoattractant receptors trigger Gαi subunit nucleotide exchange, stimulating the activation of downstream effector molecules. Activated chemoattractant receptors also dock G protein-coupled receptor kinases (GRKs) that help mediate receptor desensitization. In this study, we show that the B cell-specific loss of GRK2 severely disrupts B cell trafficking and immune cell homeostasis. The GRK2 deficiency in developing murine B cells leads to a severe immune phenotype, including a major reduction of bone marrow IgD+ cells, splenomegaly with a loss of white pulp and grossly expanded red pulp, a deficit of Peyer patches, and small lymph nodes with marked reductions in B cell numbers. The major phenotypes in these mice arise from excessive S1PR1 signaling combined with inadequate homeostatic chemokine receptor signaling. CXCL13 signaling is the most severely compromised. In B cells, our data also indicate that S1PR1 signals constitutively, as blocking S1PR1 signaling with an S1PR1 antagonist enhanced CXCL13-triggered wild-type B cell migration. Furthermore, blocking S1PR1 signaling in the GRK2-deficient B cells partially corrected their poor response to chemokines. Treating mice lacking GRK2 expression in their B cells with an S1PR1 antagonist partially normalized B cell trafficking into lymph node and splenic follicles. These findings reveal the critical interdependence of Gαi-linked signaling pathways in controlling B lymphocyte trafficking.


Subject(s)
B-Lymphocytes/physiology , Homeostasis , Lymphoid Tissue/physiology , Receptors, Chemokine/physiology , Sphingosine-1-Phosphate Receptors/physiology , Animals , Calcium/metabolism , Cell Movement , Chemokine CXCL13/physiology , G-Protein-Coupled Receptor Kinase 2/physiology , Leukocytosis/immunology , Lysophospholipids/pharmacology , Mice , Mice, Inbred C57BL , Receptors, CXCR4/physiology , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
8.
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
9.
F1000Res ; 7: 514, 2018.
Article in English | MEDLINE | ID: mdl-30345018

ABSTRACT

Immunomodulatory therapies available for the treatment of patients with multiple sclerosis (MS) accomplish control and neutralization of peripheral immune cells involved in the activity of the disease cascade but their spectrum of action in the intrathecal space and brain tissue is limited, taking into consideration the persistence of oligoclonal bands and the variation of clones of lymphoid cells throughout the disease span. In animal models of experimental autoimmune encephalomyelitis (EAE), the presence of CXCL13 has been associated with disease activity and the blockade of this chemokine could work as a potential complementary therapeutic strategy in patients with MS in order to postpone disease progression. The development of therapeutic alternatives with ability to modify the intrathecal inflammatory activity of the meningeal tertiary lymphoid organ to ameliorate neurodegeneration is mandatory.


Subject(s)
Chemokine CXCL13/physiology , Immunomodulation , Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Animals , Chemokine CXCL13/genetics , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Humans , Lymphoid Tissue/immunology , Meninges/immunology
10.
Arthritis Res Ther ; 20(1): 114, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29880013

ABSTRACT

BACKGROUND: B cells exert their pathogenic action in rheumatoid arthritis (RA) locally in the synovium. This study was undertaken to elucidate the chemokines responsible for the recruitment of B cells in the inflamed synovium, taking into account that the rich chemokine milieu present in the synovial tissue can fine-tune modulate discrete chemokine receptors. METHODS: Expression levels of chemokine receptors from the CC and CXC family, as well as CD27, were assessed by flow cytometry in CD20+ mononuclear cells isolated from the peripheral blood (PB) and synovial fluid (SF) of RA and psoriatic arthritis patients. Transwell experiments were used to study migration of B cells in response to a chemokine or in the presence of multiple chemokines. RESULTS: B cells from the SF of arthritis patients showed a significant increase in the surface expression of CCR1, CCR2, CCR4, CCR5 and CXCR4 with respect to PB. Conversely, SF B cells expressed consistently lower amounts of CXCR5, CXCR7 and CCR6, independent of CD27 expression. Analysis of permeabilized B cells suggested internalization of CXCR5 and CCR6 in SF B cells. In Transwell experiments, CCL20 and CXCL13, ligands of CCR6 and CXCR5, respectively, caused a significantly higher migration of B cells from PB than of those from SF of RA patients. Together, these two chemokines synergistically increased B-cell migration from PB, but not from SF. CONCLUSIONS: These results suggest that CXCL13 and CCL20 might play major roles in RA pathogenesis by acting singly on their selective receptors and synergistically in the accumulation of B cells within the inflamed synovium.


Subject(s)
Arthritis, Rheumatoid/metabolism , B-Lymphocytes/metabolism , Cell Movement/physiology , Chemokine CCL20/physiology , Chemokine CXCL13/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , B-Lymphocytes/immunology , Cells, Cultured , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Male , Middle Aged , Synovial Fluid/cytology , Synovial Fluid/immunology , Synovial Fluid/metabolism , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology
11.
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
12.
Ocul Immunol Inflamm ; 25(6): 811-819, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27380422

ABSTRACT

PURPOSE: B cells participate in diverse retinal immunopathologies. Endothelial adhesion molecules and chemokines direct leukocyte trafficking. We examined the involvement of three molecular signals in retinal transendothelial migration of human B cells: ICAM-1, VCAM-1, and CXCL13. METHODS: Peripheral blood B cells were isolated by negative selection. Migration was studied in transwells populated with human retinal endothelial monolayers, using antibody to block ICAM-1 or VCAM-1. Retinal expression of CXCL13 was investigated. RESULTS: B cells crossed retinal endothelium. ICAM-1 blockade significantly reduced migration when results for all subjects were combined, and for a majority when results were analyzed by individual. This effect was irrespective of the presence or absence of CXCL13, although CXCL13 increased migration. CXCL13 was detected in neural retina and retinal pigment epithelium. Endothelial cells of some retinal vessels presented CXCL13 protein. CONCLUSION: ICAM-1 blockade may be an effective treatment in some patients with retinal diseases that involve B cells.


Subject(s)
B-Lymphocytes/physiology , Cell Movement/physiology , Chemokine CXCL13/physiology , Intercellular Adhesion Molecule-1/physiology , Retina/physiology , Vascular Cell Adhesion Molecule-1/physiology , Endothelium, Vascular/metabolism , Humans , Immunohistochemistry , Immunophenotyping , Retinal Pigment Epithelium/physiology , Retinal Vessels , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transendothelial and Transepithelial Migration/physiology
13.
Brain Dev ; 38(5): 439-48, 2016 May.
Article in English | MEDLINE | ID: mdl-26786246

ABSTRACT

Despite advances in inducing remission in pediatric opsoclonus-myoclonus syndrome (OMS), relapse remains a challenge. By definition, relapse is not a characteristic of monophasic OMS, but occurs at any time in the course of multiphasic OMS. Due to variability and heterogeneity, patients are best approached and treated on a case-by-case basis, using precepts derived from clinical and scientific studies. Treatment of provocations, such as infection or immunotherapy tapering, is the short-term goal, but discovering unresolved neuroinflammation and re-configuring disease-modifying agents is crucial in the long-term. The working hypothesis is that much of the injury in OMS results from neuroinflammation involving dysregulated B cells, which may cause loss of tolerance and autoantibody production. Biomarkers of disease activity include cerebrospinal fluid (CSF) B cell frequency, oligoclonal bands (OCB), B cell attractants (CXCL13) and activating factors (BAFF). Measuring these markers comprises modern detection and characterization of neuroinflammation or verifies 'no evidence of disease activity'. The decision making process is three-tiered: deciding if the relapse is bone fide, identifying its etiology, and formulating a therapeutic plan. Relapsing-remitting OMS is treatable, and combination multimodal/multi-mechanistic immunotherapy is improving the outcome. However, some patients progress to a refractory state with cognitive impairment and disability from failure to go into remission, multiple relapses, or more aggressive disease. This report provides new insights on underappreciated risks and pitfalls inherent in relapse, pro-active efforts to avoid progression, the need for early and sufficient treatment beyond corticosteroids and immunoglobulins, and utilization of disease activity biomarkers to identify high-risk patients and safely withdraw immunotherapy.


Subject(s)
Opsoclonus-Myoclonus Syndrome/drug therapy , Secondary Prevention , Adolescent , Adrenal Cortex Hormones/therapeutic use , Autoantibodies , B-Lymphocytes/pathology , Biomarkers/cerebrospinal fluid , Chemokine CXCL13/physiology , Child , Child, Preschool , Cross-Sectional Studies , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunotherapy/trends , Neoplasm Recurrence, Local/complications , Neoplasm Recurrence, Local/therapy , Opsoclonus-Myoclonus Syndrome/cerebrospinal fluid , Opsoclonus-Myoclonus Syndrome/rehabilitation , Secondary Prevention/trends
14.
Oncotarget ; 7(7): 7550-62, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26771137

ABSTRACT

Abnormal overexpression of CXCL13 is observed in many inflamed tissues and in particular in autoimmune diseases. Myasthenia gravis (MG) is a neuromuscular disease mainly mediated by anti-acetylcholine receptor autoantibodies. Thymic hyperplasia characterized by ectopic germinal centers (GCs) is a common feature in MG and is correlated with high levels of anti-AChR antibodies. We previously showed that the B-cell chemoattractant, CXCL13 is overexpressed by thymic epithelial cells in MG patients. We hypothesized that abnormal CXCL13 expression by the thymic epithelium triggered B-cell recruitment in MG. We therefore created a novel transgenic (Tg) mouse with a keratin 5 driven CXCL13 expression. The thymus of Tg mice overexpressed CXCL13 but did not trigger B-cell recruitment. However, in inflammatory conditions, induced by Poly(I:C), B cells strongly migrated to the thymus. Tg mice were also more susceptible to experimental autoimmune MG (EAMG) with stronger clinical signs, higher titers of anti-AChR antibodies, increased thymic B cells, and the development of germinal center-like structures. Consequently, this mouse model finally mimics the thymic pathology observed in human MG. Our data also demonstrated that inflammation is mandatory to reveal CXCL13 ability to recruit B cells and to induce tertiary lymphoid organ development.


Subject(s)
B-Lymphocytes/pathology , Chemokine CXCL13/physiology , Inflammation/complications , Myasthenia Gravis, Autoimmune, Experimental/pathology , Thymus Hyperplasia/physiopathology , Animals , B-Lymphocytes/metabolism , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Flow Cytometry , Germinal Center/metabolism , Germinal Center/pathology , Humans , Immunoenzyme Techniques , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myasthenia Gravis, Autoimmune, Experimental/etiology , Myasthenia Gravis, Autoimmune, Experimental/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
15.
Acta Histochem ; 117(8): 732-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26456520

ABSTRACT

The chemokine CXC ligand 13 protein (CXCL13) is reported to closely related to the disease activity and severity of systemic lupus erythematosus (SLE), moreover, the level of CXCL13 was markedly raised in kidney tissues of lupus nephritis (LN) patients. The aim of the present study was to explore whether the blockade of CXCL13 has therapeutic effects on murine LN. MRL/lpr mice received 50µg anti-CXCL13 neutralizing antibody or isotype IgG by intraperitoneal injection everyday for six weeks, and renal damage of each group was determined. Our results showed that the blockade of CXCL13 significantly reduced urine protein, serum creatinine, and dramatically attenuated renal pathology injury. Treatment with anti-CXCL13Ab also reduced serum anti-dsDNA level, renal immune complex deposition as well as inflammatory cytokines secretion. Meanwhile, Th17/Treg ratio in spleens of MRL/lpr mice was significantly decreased by the blocking of CXCL13. These findings suggested that CXCL13 may be a promising target for the therapy of LN.


Subject(s)
Chemokine CXCL13/antagonists & inhibitors , Lupus Nephritis/drug therapy , Animals , Autoantibodies/blood , CD4 Lymphocyte Count , Chemokine CXCL13/physiology , Drug Evaluation, Preclinical , Female , Kidney/drug effects , Kidney/metabolism , Lupus Nephritis/blood , Mice, Inbred MRL lpr , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
16.
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
17.
Brain Tumor Pathol ; 32(1): 41-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25433721

ABSTRACT

Chemokines are peptides that function as chemoattractant cytokines in cell activation, differentiation and trafficking. Endothelin B receptor (ETBR) is a receptor for endothelin, which is known to function as a vasoconstrictor. In the present study, to clarify the immune escape mechanism of primary central nervous system lymphomas (PCNSLs), the expression of ETBR and of subsets of chemokines (CXCL12, 13) in 24 PCNSLs was investigated. CXCL12 was expressed by lymphoma cells in different resident brain cell populations in 22/24 cases. CXCL13 expression was identified in tumor cells in 19/24 cases, but was only expressed by tumor cells and by proliferating vascular endothelial cells. In addition, tumor-infiltrated lymphocytes (TILs) accumulated in areas with expression of chemokines, particularly of CXCL13. ETBR expression was detected in 12/24 cases. Positive ETBR cases were associated with a paucity of TILs, particularly of cytotoxic T cells, whereas negative ETBR cases were associated with an abundance of TILs. The combined data indicate that CXCL12 and CXCL13 up-regulation may be differently linked to the development of PCNSLs and to the accumulation of TILs. In addition, ETBR expression by lymphoma and endothelial cells may mediate trafficking of TILs, which may explain the immune escape processes of PCNSLs.


Subject(s)
Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/immunology , Chemokine CXCL12/physiology , Chemokine CXCL13/physiology , Lymphoma/genetics , Lymphoma/immunology , Receptor, Endothelin B/physiology , Tumor Escape/genetics , Tumor Microenvironment/genetics , Aged , Aged, 80 and over , Central Nervous System Neoplasms/blood supply , Central Nervous System Neoplasms/pathology , Chemokine CXCL12/genetics , Chemokine CXCL13/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphoma/pathology , Male , Middle Aged , Receptor, Endothelin B/genetics , Up-Regulation
18.
J Leukoc Biol ; 94(5): 1079-89, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23904442

ABSTRACT

SS is an autoimmune disease. pSS affects exocrine glands predominantly, whereas sSS occurs with other autoimmune connective tissue disorders. Currently, care for patients with SS is palliative, as no established therapeutics target the disease directly, and its pathogenetic mechanisms remain uncertain. B-cell abnormalities have been identified in SS. CXCL13 directs B-cell chemotaxis and is elevated in several autoimmune diseases. In this study, we tested the hypothesis that CXCL13 is elevated in SS in mice and humans and that neutralization of the chemokine ameliorates disease in a murine model. We assayed CXCL13 in mouse models and human subjects with SS to determine whether CXCL13 is elevated both locally and systemically during SS progression and whether CXCL13 may play a role in and be a biomarker for the disease. Cxcl13 expression in salivary tissue increases with disease progression, and its blockade resulted in a modest reduction in glandular inflammation in an SS model. We demonstrate that in humans CXCL13 is elevated in serum and saliva, and an elevated salivary CXCL13 level distinguishes patients with xerostomia. These data suggest a role for CXCL13 as a valuable biomarker in SS, as 74% of patients with SS displayed elevated CXCL13 in sera, saliva, or both. Thus, CXCL13 may be pathogenically involved in SS and may serve as a new marker and a potential therapeutic target.


Subject(s)
Chemokine CXCL13/physiology , Sjogren's Syndrome/immunology , Animals , Chemokine CXCL13/analysis , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, CXCR5/analysis , Salivary Glands/chemistry , Sjogren's Syndrome/etiology , Xerostomia/immunology
19.
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
20.
Am J Transplant ; 12(6): 1610-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22335599

ABSTRACT

Posttransplant lymphoproliferative disease (PTLD) is a severe complication of immunosuppressive treatment in organ-grafted children. Early diagnosis of PTLD is hampered by both unspecific clinical symptoms and lack of easy accessible markers. The homeostatic chemokine CXCL13, which plays a crucial role in B-cell homing and lymphoid organ development, is expressed in some lymphomatous diseases. This study aims to investigate whether serum CXCL13 (sCXCL13) levels correlate with occurrence and regression of PTLD in pediatric solid-organ graft recipients. Serum samples from PTLD patients (n = 21), patients with Epstein-Barr virus (EBV) reactivation (n = 18), and healthy age-matched controls (n = 19) were tested for CXCL13 using a commercially available ELISA kit. sCXCL13 levels were significantly higher in PTLD patients than in healthy children. PTLD patients had also higher sCXCL13 values than pediatric solid-organ recipients with EBV reactivation. An increase in sCXCL13 levels was observed from EBV reactivation to PTLD diagnosis in most cases. Elevated sCXCL13 levels were detected up to 2 years prior to PTLD diagnosis and correlated well with response to cytoreductive treatment in individual patients. sCXCL13, thus, may be a readily available surrogate marker for the diagnosis of PTLD and for monitoring of response to treatment in patients with initially elevated sCXCL13 levels.


Subject(s)
Chemokine CXCL13/physiology , Lymphoproliferative Disorders/diagnosis , Adolescent , Base Sequence , Child , Child, Preschool , DNA Primers , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lymphoproliferative Disorders/physiopathology , Male , Monitoring, Physiologic
SELECTION OF CITATIONS
SEARCH DETAIL
...