Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Cells ; 10(4)2021 04 07.
Article in English | MEDLINE | ID: mdl-33917019

ABSTRACT

CD40-activated CD40L-mediated reverse signalling is a major physiological regulator of neurite growth from excitatory and inhibitory neurons in the developing central nervous system (CNS). Whereas in excitatory pyramidal neurons, CD40L reverse signalling promotes the growth and elaboration of dendrites and axons, in inhibitory GABAergic striatal medium spiny neurons (MSNs), it restricts neurite growth and branching. In pyramidal neurons, we previously reported that CD40L reverse signalling activates an interconnected and interdependent signalling network involving protein kinase C (PKC), extracellular regulated kinases 1 and 2 (ERK1/2), and c-Jun N-terminal kinase (JNK) signalling pathways that regulates dendrite and axon growth. Here, we have studied whether these signalling pathways also influence neurite growth from striatal inhibitory MSNs. To unequivocally activate CD40L reverse signalling, we treated MSN cultures from CD40-deficient mice with CD40-Fc. Here, we report that activation of CD40L reverse signalling in these cultures also increased the phosphorylation of PKC, ERK1/2, and JNK. Using pharmacological activators and inhibitors of these signalling pathways singularly and in combination, we have shown that, as in pyramidal neurons, these signalling pathways work in an interconnected and interdependent network to regulate the neurite growth, but their functions, relationships, and interdependencies are different from those observed in pyramidal neurons. Furthermore, immunoprecipitation studies showed that stimulation of CD40L reverse signalling recruits the catalytic fragment of Syk tyrosine kinase, but in contrast to pyramidal neurons, PKC does not participate in this recruitment. Our findings show that distinctive networks of three signalling pathways mediate the opposite effects of CD40L reverse signalling on neurite growth in excitatory and inhibitory neurons.


Subject(s)
CD40 Antigens/metabolism , CD40 Ligand/metabolism , Neurites/metabolism , Signal Transduction , Animals , CD40 Antigens/deficiency , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , GABAergic Neurons/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , Models, Biological , Phosphorylation , Protein Interaction Maps , Protein Kinase C/metabolism , Syk Kinase/metabolism
2.
Thromb Haemost ; 121(11): 1530-1540, 2021 11.
Article in English | MEDLINE | ID: mdl-33618394

ABSTRACT

OBJECTIVES: The co-stimulatory CD40L-CD40 dyad exerts a critical role in atherosclerosis by modulating leukocyte accumulation into developing atherosclerotic plaques. The requirement for cell-type specific expression of both molecules, however, remains elusive. Here, we evaluate the contribution of CD40 expressed on endothelial cells (ECs) in a mouse model of atherosclerosis. METHODS AND RESULTS: Atherosclerotic plaques of apolipoprotein E-deficient (Apoe -/- ) mice and humans displayed increased expression of CD40 on ECs compared with controls. To interrogate the role of CD40 on ECs in atherosclerosis, we induced EC-specific (BmxCreERT2-driven) deficiency of CD40 in Apoe -/- mice. After feeding a chow diet for 25 weeks, EC-specific deletion of CD40 (iEC-CD40) ameliorated plaque lipid deposition and lesional macrophage accumulation but increased intimal smooth muscle cell and collagen content, while atherosclerotic lesion size did not change. Leukocyte adhesion to the vessel wall was impaired in iEC-CD40-deficient mice as demonstrated by intravital microscopy. In accord, expression of vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) in the vascular endothelium declined after deletion of CD40. In vitro, antibody-mediated inhibition of human endothelial CD40 significantly abated monocyte adhesion on ECs. CONCLUSION: Endothelial deficiency of CD40 in mice promotes structural features associated with a stable plaque phenotype in humans and decreases leukocyte adhesion. These results suggest that endothelial-expressed CD40 contributes to inflammatory cell migration and consecutive plaque formation in atherogenesis.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , CD40 Antigens/deficiency , Chemotaxis, Leukocyte , Endothelial Cells/metabolism , Macrophages/metabolism , Monocytes/metabolism , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , CD40 Antigens/genetics , Cell Adhesion , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/pathology , Humans , Intercellular Adhesion Molecule-1/metabolism , Macrophages/immunology , Male , Mice, Knockout, ApoE , Monocytes/immunology , Plaque, Atherosclerotic , Signal Transduction , Vascular Cell Adhesion Molecule-1/metabolism
3.
Cell Mol Life Sci ; 78(3): 1065-1083, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32506167

ABSTRACT

CD40-activated CD40L reverse signaling is a major physiological regulator of axon and dendrite growth from developing hippocampal pyramidal neurons. Here we have studied how CD40L-mediated reverse signaling promotes the growth of these processes. Cultures of hippocampal pyramidal neurons were established from Cd40-/- mouse embryos to eliminate endogenous CD40/CD40L signaling, and CD40L reverse signaling was stimulated by a CD40-Fc chimera. CD40L reverse signaling increased phosphorylation and hence activation of proteins in the PKC, ERK, and JNK signaling pathways. Pharmacological activators and inhibitors of these pathways revealed that whereas activation of JNK inhibited growth, activation of PKC and ERK1/ERK2 enhanced growth. Experiments using combinations of pharmacological reagents revealed that these signaling pathways regulate growth by functioning as an interconnected and interdependent network rather than acting in a simple linear sequence. Immunoprecipitation studies suggested that stimulation of CD40L reverse signaling generated a receptor complex comprising CD40L, PKCß, and the Syk tyrosine kinase. Our studies have begun to elucidate the molecular network and interactions that promote axon and dendrite growth from developing hippocampal neurons following activation of CD40L reverse signaling.


Subject(s)
Axons/metabolism , CD40 Ligand/metabolism , Dendrites/physiology , Signal Transduction , Animals , Butadienes/pharmacology , CD40 Antigens/deficiency , CD40 Antigens/genetics , Cells, Cultured , Dendrites/drug effects , Hippocampus/cytology , Hippocampus/metabolism , JNK Mitogen-Activated Protein Kinases/chemistry , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/chemistry , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Phosphorylation , Protein Interaction Domains and Motifs , Protein Kinase C/metabolism , Signal Transduction/drug effects , Syk Kinase/metabolism
4.
Biomed Pharmacother ; 129: 110450, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32768945

ABSTRACT

OBJECTIVES: Astilbin exerts immunoregulatory activities and plays anti-inflammatory effects in inflammation-associated diseases. IL-10-producing B cells are the major subset of regulatory B cells (Bregs) and inhibit inflammation and autoimmune diseases. This study aimed to analyse the inducing effect of astilbin on Bregs and investigate the involved molecular mechanisms. METHODS: The frequencies and activities of IL-10-producing Bregs were observed using the co-treatment of astilbin and lipopolysaccharide (LPS) ex vivo. The protective effect of astilbin/LPS-induced Bregs on dextran sulphate sodium (DSS)-induced colitis was confirmed in vivo. The molecular signalling events of Breg induction were checked via Western blot. CD40-/- and toll-like receptor (TLR) 4-/- B cells were treated with astilbin/LPS to determine the modulatory role of CD40 or TLR4 on astilbin/LPS-induced Bregs. RESULTS: Although astilbin alone could not affect Bregs, the co-treatment of astilbin and LPS remarkably induced CD19+ CD1dhi and CD19+ TIM-1+ cells which produced IL-10 ex vivo. Colonic CD19+ CD1dhi and CD19+ TIM-1+ cells were also increased in astilbin-treated mice with DSS-induced colitis. The adoptive transfer of CD19+ TIM-1+ cells pre-induced by astilbin/LPS directly suppressed the progression of DSS-induced colitis. Combined astilbin and LPS stimulated the STAT3 activation of CD19+ TIM-1+ cells but had no effects on SOCS3, AKT, NF-κB, Erk, JNK nor P38. Inhibiting the STAT3 phosphorylation of CD19+ TIM-1+ cells abolished Breg induction by astilbin/LPS. Furthermore, Breg induction was weakened in CD40-/- B cells with the decrease in STAT3 activation, but had disappeared in TLR4-/- B cells with no STAT3 activation, thereby confirming the indispensable role of TLR4 signalling in the induction of IL-10-producing Bregs. CONCLUSIONS: This study reports the new immunoregulatory role of astilbin for promoting IL-10-producing B cells and suggests the possible use of astilbin in the therapy of inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , B-Lymphocytes, Regulatory/drug effects , Colitis/drug therapy , Colon/drug effects , Flavonols/pharmacology , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , STAT3 Transcription Factor/metabolism , Adoptive Transfer , Animals , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , B-Lymphocytes, Regulatory/transplantation , CD40 Antigens/deficiency , CD40 Antigens/genetics , Cells, Cultured , Coculture Techniques , Colitis/chemically induced , Colitis/immunology , Colitis/metabolism , Colon/immunology , Colon/metabolism , Dextran Sulfate , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
5.
Sci Rep ; 9(1): 15932, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31685838

ABSTRACT

In advanced inflammatory disease, microvascular thrombosis leads to the interruption of blood supply and provokes ischemic tissue injury. Recently, intravascularly adherent leukocytes have been reported to shape the blood flow in their immediate vascular environment. Whether these rheological effects are relevant for microvascular thrombogenesis remains elusive. Employing multi-channel in vivo microscopy, analyses in microfluidic devices, and computational modeling, we identified a previously unanticipated role of leukocytes for microvascular clot formation in inflamed tissue. For this purpose, neutrophils adhere at distinct sites in the microvasculature where these immune cells effectively promote thrombosis by shaping the rheological environment for platelet aggregation. In contrast to larger (lower-shear) vessels, this process in high-shear microvessels does not require fibrin generation or extracellular trap formation, but involves GPIbα-vWF and CD40-CD40L-dependent platelet interactions. Conversely, interference with these cellular interactions substantially compromises microvascular clotting. Thus, leukocytes shape the rheological environment in the inflamed venular microvasculature for platelet aggregation thereby effectively promoting the formation of blood clots. Targeting this specific crosstalk between the immune system and the hemostatic system might be instrumental for the prevention and treatment of microvascular thromboembolic pathologies, which are inaccessible to invasive revascularization strategies.


Subject(s)
Blood Platelets/physiology , Neutrophils/physiology , Platelet Aggregation/physiology , Thrombosis/pathology , Animals , Blood Platelets/metabolism , CD40 Antigens/deficiency , CD40 Antigens/genetics , CD40 Ligand/deficiency , CD40 Ligand/genetics , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Microfluidics/instrumentation , Microfluidics/methods , Microscopy, Fluorescence , Microvessels/drug effects , Microvessels/pathology , Neutrophils/immunology , Platelet Adhesiveness/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Rheology , Thrombosis/metabolism , von Willebrand Factor/metabolism
7.
Bone Marrow Transplant ; 54(1): 63-67, 2019 01.
Article in English | MEDLINE | ID: mdl-29884852

ABSTRACT

Hyper-IgM syndrome due to CD40 deficiency (HIGM3) is a rare disease with only a few reported cases of haematopoietic stem cell transplantation (HSCT). In retrospective study, we reviewed all patients with HIGM3 who underwent HSCT at King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia, between 2008 and 2013. Six patients were identified. Three male and three female patients from three families. The median age of diagnosis was 13 months (range, 1-28 months). All lacked CD40 expression on B cells by flow cytometry. The median time from diagnosis to transplantation was 8.5 months (range, 1-17 months). For all patients, the donors were HLA-identical siblings, with the exception of one patient for whom the donor was a sibling with one antigen mismatch. The conditioning regimen was busulfan and cyclophosphamide in five patients and busulfan, cyclophosphamide and antithymocyte globulin in one patient. For GVHD prophylaxis, cyclosporine and methotrexate was used. All patients engrafted. The survival rate was 100%, with a median follow-up of 54 months (range, 30-116 months). One patient developed acute GVHD. All patients showed complete immune recovery with positive CD40 expression on B cells and discontinued IVIG replacement. Our study shows that HSCT is potentially effective at curing the disease.


Subject(s)
CD40 Antigens/deficiency , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Hyper-IgM Immunodeficiency Syndrome , Siblings , Transplantation Conditioning , Antilymphocyte Serum/administration & dosage , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Busulfan/administration & dosage , Child, Preschool , Cyclophosphamide/administration & dosage , Disease-Free Survival , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Graft vs Host Disease/pathology , Graft vs Host Disease/prevention & control , Humans , Hyper-IgM Immunodeficiency Syndrome/immunology , Hyper-IgM Immunodeficiency Syndrome/mortality , Hyper-IgM Immunodeficiency Syndrome/pathology , Hyper-IgM Immunodeficiency Syndrome/therapy , Infant , Infant, Newborn , Male , Retrospective Studies , Survival Rate
8.
J Pathol ; 247(4): 471-480, 2019 04.
Article in English | MEDLINE | ID: mdl-30471110

ABSTRACT

The costimulatory CD40L-CD40 dyad plays a major role in multiple sclerosis (MS). CD40 is highly expressed on MHCII+ B cells, dendritic cells and macrophages in human MS lesions. Here we investigated the role of the CD40 downstream signaling intermediates TNF receptor-associated factor 2 (TRAF2) and TRAF6 in MHCII+ cells in experimental autoimmune encephalomyelitis (EAE). Both MHCII-CD40-Traf2-/- and MHCII-CD40-Traf6-/- mice showed a reduction in clinical signs of EAE and prevented demyelination. However, only MHCII-CD40-Traf6-/- mice displayed a decrease in myeloid and lymphoid cell infiltration into the CNS that was accompanied by reduced levels of TNF-α, IL-6 and IFN-γ. As CD40-TRAF6 interactions predominantly occur in macrophages, we subjected CD40flfl LysMcre mice to EAE. This myeloid-specific deletion of CD40 resulted in a significant reduction in EAE severity, reduced CNS inflammation and demyelination. In conclusion, the CD40-TRAF6 signaling pathway in MHCII+ cells plays a key role in neuroinflammation and demyelination during EAE. Concomitant with the fact that CD40-TRAF6 interactions are predominant in macrophages, depletion of myeloid CD40 also reduces neuroinflammation. CD40-TRAF6 interactions thus represent a promising therapeutic target for MS. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
CD40 Antigens/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Macrophages/immunology , Signal Transduction/immunology , TNF Receptor-Associated Factor 6/physiology , Animals , Autoantibodies/metabolism , CD40 Antigens/deficiency , CD40 Ligand/physiology , Cytokines/metabolism , Female , Immunoglobulin G/immunology , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Neuritis/immunology
9.
Front Immunol ; 9: 1761, 2018.
Article in English | MEDLINE | ID: mdl-30131802

ABSTRACT

Mutations in genes that control class switch recombination and somatic hypermutation during the germinal center (GC) response can cause diverse immune dysfunctions. In particular, mutations in CD40LG, CD40, AICDA, or UNG cause hyper-IgM (HIGM) syndrome, a heterogeneous group of primary immunodeficiencies. Follicular helper (Tfh) and follicular regulatory (Tfr) T cells play a key role in the formation and regulation of GCs, but their role in HIGM pathogenesis is still limited. Here, we found that compared to CD40 ligand (CD40L)- and activation-induced cytidine deaminase (AICDA)-deficient patients, circulating Tfh and Tfr cells were severely compromised in terms of frequency and activation phenotype in a child with CD40 deficiency. These findings offer useful insight for human Tfh biology, with potential implications for understanding the molecular basis of HIGM syndrome caused by mutations in CD40.


Subject(s)
CD40 Antigens/deficiency , Hyper-IgM Immunodeficiency Syndrome/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , CD40 Antigens/genetics , CD40 Ligand/genetics , Child, Preschool , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Female , Humans , Hyper-IgM Immunodeficiency Syndrome/genetics , Immunophenotyping , Lymphocyte Activation , Male , Mutation , Young Adult
10.
J Immunol ; 199(12): 4165-4179, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29084838

ABSTRACT

We describe an MHC class II (I-Ab)-restricted TCR transgenic mouse line that produces CD4+ T cells specific for Plasmodium species. This line, termed PbT-II, was derived from a CD4+ T cell hybridoma generated to blood-stage Plasmodium berghei ANKA (PbA). PbT-II cells responded to all Plasmodium species and stages tested so far, including rodent (PbA, P. berghei NK65, Plasmodium chabaudi AS, and Plasmodium yoelii 17XNL) and human (Plasmodium falciparum) blood-stage parasites as well as irradiated PbA sporozoites. PbT-II cells can provide help for generation of Ab to P. chabaudi infection and can control this otherwise lethal infection in CD40L-deficient mice. PbT-II cells can also provide help for development of CD8+ T cell-mediated experimental cerebral malaria (ECM) during PbA infection. Using PbT-II CD4+ T cells and the previously described PbT-I CD8+ T cells, we determined the dendritic cell (DC) subsets responsible for immunity to PbA blood-stage infection. CD8+ DC (a subset of XCR1+ DC) were the major APC responsible for activation of both T cell subsets, although other DC also contributed to CD4+ T cell responses. Depletion of CD8+ DC at the beginning of infection prevented ECM development and impaired both Th1 and follicular Th cell responses; in contrast, late depletion did not affect ECM. This study describes a novel and versatile tool for examining CD4+ T cell immunity during malaria and provides evidence that CD4+ T cell help, acting via CD40L signaling, can promote immunity or pathology to blood-stage malaria largely through Ag presentation by CD8+ DC.


Subject(s)
Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , Dendritic Cells/immunology , Malaria/immunology , Mice, Transgenic/immunology , Parasitemia/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Protozoan/immunology , CD40 Antigens/deficiency , CD40 Ligand/immunology , Cells, Cultured , Crosses, Genetic , Hybridomas , Lymphocyte Activation , Malaria, Cerebral/immunology , Malaria, Cerebral/prevention & control , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic/genetics , Plasmodium berghei/immunology , Radiation Chimera
11.
Elife ; 62017 11 07.
Article in English | MEDLINE | ID: mdl-29111976

ABSTRACT

Dendrite size and morphology are key determinants of the functional properties of neurons and neural circuits. Here we show that CD40, a member of the TNF receptor superfamily, is a major regulator of dendrite growth and elaboration in the developing brain. The dendrites of hippocampal excitatory neurons were markedly stunted in Cd40-/- mice, whereas those of striatal inhibitory neurons were much more exuberant. These striking and opposite phenotypic changes were also observed in excitatory and inhibitory neurons cultured from Cd40-/- mice and were rescued by soluble CD40. The changes in excitatory and inhibitory neurons cultured from Cd40-/- mice were mimicked in neurons of Cd40+/+ mice by treatment with soluble CD40L and were dependent on PKC-ß and PKC-γ, respectively. These results suggest that CD40-activated CD40L reverse signalling has striking and opposite effects on the growth and elaboration of dendrites among major classes of brain neurons by PKC-dependent mechanisms.


Subject(s)
CD40 Antigens/metabolism , Dendrites/physiology , Hippocampus/cytology , Neurons/cytology , Animals , CD40 Antigens/deficiency , CD40 Ligand/metabolism , Mice , Mice, Knockout , Protein Kinase C/metabolism
12.
PLoS One ; 12(10): e0186573, 2017.
Article in English | MEDLINE | ID: mdl-29023539

ABSTRACT

Upon recognition of peptide displayed on MHC molecules, Th1 and Th2 cells form distinct immunological synapse structures. Th1 cells have a bull's eye synapse structure with TCR/ MHC-peptide interactions occurring central to a ring of adhesion molecules, while Th2 cells have a multifocal synapse with small clusters of TCR/MHC interactions throughout the area of T cell/antigen-presenting cell interaction. In this study, we investigated whether this structural difference in the immunological synapse affects delivery of T cell help. The immunological synapse is thought to ensure antigen-specific delivery of cytolytic granules and killing of target cells by NK cells and cytolytic T cells. In helper T cells, it has been proposed that the immunological synapse may direct delivery of other effector molecules including cytokines. CD40 ligand (CD40L) is a membrane-bound cytokine essential for antigen-specific T cell help for B cells in the antibody response. We incubated Th1 and Th2 cells overnight with a mixture of antigen-presenting and bystander B cells, and the delivery of CD40L to B cells and subsequent B cell responses were compared. Despite distinct immunological synapse structures, Th1 and Th2 cell do not differ in their ability to deliver CD40L and T cell help in an antigen-specific fashion, or in their susceptibility to inhibition of help by a blocking anti-CD40L antibody.


Subject(s)
Antigen-Presenting Cells/metabolism , B-Lymphocytes/metabolism , CD40 Ligand/metabolism , Synapses/chemistry , Th2 Cells/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Antigen-Presenting Cells/cytology , B-Lymphocytes/immunology , CD40 Antigens/deficiency , CD40 Antigens/genetics , CD40 Ligand/genetics , CD40 Ligand/immunology , Cyclosporine/pharmacology , Female , Immunological Synapses/chemistry , Immunological Synapses/drug effects , Immunological Synapses/immunology , Interleukin-4/immunology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Synapses/metabolism , Th1 Cells/cytology , Th1 Cells/metabolism , Th2 Cells/cytology
13.
J Immunol ; 198(11): 4490-4501, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28455435

ABSTRACT

The inflammatory CD40-CD40L pathway is implicated in various autoimmune diseases, but the activity status of this pathway in various stages of rheumatoid arthritis (RA) progression is unknown. In this study, we used gene signatures of CD40L stimulation derived from human immature dendritic cells and naive B cells to assess the expression of CD40-downstream genes in synovial tissues from anti-citrullinated protein Ab-positive arthralgia, undifferentiated arthritis (UA), early RA, and established RA cohorts in comparison with healthy donors. Interestingly, the expression of CD40LG and active full-length CD40 was increased in the disease tissues, whereas that of a dominant-negative CD40 isoform was decreased. Gene set variation analysis revealed that CD40L-responsive genes in immature dendritic cells and naive B cells were significantly enriched in synovial tissues from UA, early RA, and established RA patients. Additionally, CD40L-induced naive B cell genes were also significantly enriched in synovial tissues from arthralgia patients. In our efforts to characterize downstream mediators of CD40L signaling, we have identified GPR120 and KDM6B as novel components of the pathway. In conclusion, our data suggest that therapeutic CD40-CD40L blocking agents may prove efficacious not only in early and established RA, but also in inhibiting the progression of the disease from arthralgia or UA to RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis/immunology , CD40 Ligand/immunology , CD40 Ligand/metabolism , Disease Progression , Signal Transduction , Adult , Aged , Arthralgia/immunology , Arthralgia/physiopathology , Arthritis, Rheumatoid/physiopathology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Biopsy , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/deficiency , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/genetics , CD40 Ligand/pharmacology , Dendritic Cells/immunology , Dendritic Cells/physiology , Female , Healthy Volunteers , Humans , Lymphocyte Activation , Male , Middle Aged , Synovial Fluid/cytology , Synovial Fluid/immunology , Transcriptome
14.
Int Immunol ; 29(3): 121-131, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28369442

ABSTRACT

Fascin is an actin-bundling protein that, among immune cells, is restricted to expression in dendritic cells (DCs). Previous reports have suggested that fascin plays an important role in governing DC antigen presentation to CD4+ T cells. However, no report has clearly linked the receptor-ligand engagement that can direct downstream regulation of fascin expression. In this study, bone marrow-derived DCs from wild-type versus CD40-knockout C57BL/6 mice were used to elucidate the mechanisms of fascin expression and activity upon CD40-CD40 ligand (CD40L) engagement. These investigations now show that CD40 engagement governs fascin expression in DCs to promote CD4+ T-cell cytokine production. Absence of CD40 signaling resulted in diminished fascin expression in DCs and was associated with impaired CD4+ T-cell responses. Furthermore, the study found that loss of CD40-CD40L engagement resulted in reduced DC-T-cell contacts. Rescue by ectopic fascin expression in CD40-deficient DCs was able to re-establish sustained contacts with T cells and restore cytokine production. Taken together, these results show that cross-talk through CD40-CD40L signaling drives elevated fascin expression in DCs to support acquisition of full T-cell responses.


Subject(s)
Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , CD40 Ligand/immunology , Carrier Proteins/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/metabolism , Microfilament Proteins/biosynthesis , Animals , CD40 Antigens/deficiency , Carrier Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/immunology
15.
J Immunol ; 198(5): 1921-1927, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28093524

ABSTRACT

The origin and nature of age-associated B cells (ABCs) in mice are poorly understood. In this article, we show that their emergence required MHC class II and CD40/CD40L interactions. Young donor B cells were adoptively transferred into congenic recipients and allowed to remain for 1 mo in the absence of external Ag. B cells expressing the T-bet transcription factor, a marker for ABCs, were generated after multiple cell divisions from C57BL/6 donors but not from MHC class II- or CD40-deficient donors. Furthermore, old CD154 (CD40L)-deficient mice did not accrue ABCs, confirming that they arise primarily through T-dependent interactions. To determine what Igs ABCs express, we sequenced VH and Vκ rearranged genes from unimmunized 22-mo-old C57BL/6 mice and showed that they had a heterogeneous repertoire, which was comparable to that seen in old follicular and marginal zone B cell subsets. However, in contrast to the follicular and marginal zone cells, ABCs displayed significant somatic hypermutation. The mutation frequency was lower than found in germinal center cells after deliberate immunization, suggesting that ABCs have undergone mild stimulation from endogenous Ags over time. These observations show that quiescent ABCs are Ag-experienced cells that accumulate during T cell-dependent responses to diverse Ags during the life of an individual.


Subject(s)
Aging/immunology , B-Lymphocyte Subsets/immunology , Single-Domain Antibodies/genetics , Somatic Hypermutation, Immunoglobulin , Animals , B-Lymphocyte Subsets/metabolism , CD40 Antigens/deficiency , CD40 Antigens/immunology , Gene Rearrangement , Genes, MHC Class II , Germinal Center/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Sequence Analysis, DNA
16.
Atherosclerosis ; 255: 80-89, 2016 12.
Article in English | MEDLINE | ID: mdl-27835742

ABSTRACT

BACKGROUND AND AIMS: CD40/CD40L signaling exerts a critical role in the development of atherosclerosis, and microRNAs (miRNAs) are key regulators in vascular inflammation and plaque formation. In this work, we investigated mRNA/miRNA expression during progression of atherosclerotic lesions through CD40 silencing. METHODS: We silenced CD40 with a specific siRNA in ApoE-/- mice and compared expression of mRNA/miRNA in ascending aorta with scrambled treated mice. RESULTS: siRNA-CD40 treated mice significantly reduced the extension and severity of atherosclerotic lesions, as well as the number of F4/80+, galectin-3+ macrophages and NF-κB+ cells in the intima. Genome-wide mRNA/miRNA profiling allowed the identification of transcripts, which were significantly upregulated during atherosclerosis; among them, miR-125b and miR-30a, Xpr1, a regulator of macrophage differentiation, Taf3, a core transcription factor and the NF-κB activator Ikkß, whereas, the NF-κB inhibitor Ikbα was downregulated during disease progression. All those changes were reversed upon CD40 silencing. Interestingly, TAF3, XPR1 and miR-125b were also overexpressed in human atherosclerotic plaques. Murine Taf3 and Xpr1 were detected in the perivascular adipose tissue (PVAT), and Taf3 also in intimal foam cells. Finally, expression of miR-125b was regulated by the CD40-NF-κB signaling axis in RAW264.7 macrophages. CONCLUSIONS: CD40 silencing with a specific siRNA ameliorates progression of experimental atherosclerosis in ApoE-/- mice, and evidences a role for NF-κB, Taf3, Xpr1, and miR-125b in the pathogenesis of atherosclerosis.


Subject(s)
Aorta/metabolism , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , CD40 Antigens/deficiency , MicroRNAs/metabolism , NF-kappa B/metabolism , RNA Interference , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , CD40 Antigens/genetics , Disease Models, Animal , Disease Progression , Female , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Macrophages/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , NF-kappa B/genetics , Phenotype , Plaque, Atherosclerotic , RAW 264.7 Cells , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Signal Transduction , TATA-Binding Protein Associated Factors , Time Factors , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcriptome , Xenotropic and Polytropic Retrovirus Receptor
17.
Arterioscler Thromb Vasc Biol ; 36(3): 482-90, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26821950

ABSTRACT

OBJECTIVE: Beyond their eminent role in hemostasis and thrombosis, platelets are recognized as mediators of inflammation. Platelet cluster of differentiation 40 (CD40) ligand (CD40L and CD154) plays a key role in mediating platelet-induced inflammation in atherosclerosis. CD40, the receptor for CD40L, is present on platelets; however, the role of CD40 on this cell type is until now undefined. APPROACH AND RESULTS: We found that in both mice and humans, platelet CD40 mediates the formation of platelet-leukocyte aggregates and the release of chemokine (C-X-C motif) ligand 4. Leukocytes were also less prone to adhere to CD40-deficient thrombi. However, platelet CD40 was not involved in platelet aggregation. Activated platelets isolated from Cd40(-/-)Apoe(-/-) mice adhered less to the endothelium upon injection into Apoe(-/-) mice when compared with CD40-sufficient platelets. Furthermore, lack of CD40 on injected platelets led to reduced leukocyte recruitment to the carotid artery as assayed by intravital microscopy. This was accompanied by a decrease in endothelial vascular cell adhesion molecule-1, platelet endothelial cell adhesion molecule, VE-cadherin, and P-selectin expression. To investigate the effect of platelet CD40 in atherosclerosis, Apoe(-/-) mice received thrombin-activated Apoe(-/-) or Cd40(-/-)Apoe(-/-) platelets every 5 days for 12 weeks, starting at the age of 17 weeks, when atherosclerotic plaques had already formed. When compared with mice that received Apoe(-/-) platelets, those receiving Cd40(-/-)Apoe(-/-) platelets exhibited a >2-fold reduction in atherosclerosis. Plaques of mice receiving CD40-deficient platelets were less advanced, contained less macrophages, neutrophils, and collagen, and displayed smaller lipid cores. CONCLUSIONS: Platelet CD40 plays a crucial role in inflammation by stimulating leukocyte activation and recruitment and activation of endothelial cells, thereby promoting atherosclerosis.


Subject(s)
Atherosclerosis/blood , Blood Platelets/metabolism , CD40 Antigens/blood , Endothelial Cells/metabolism , Inflammation/blood , Leukocytes/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , CD40 Antigens/deficiency , CD40 Antigens/genetics , CD40 Ligand/blood , Cells, Cultured , Chemotaxis , Coculture Techniques , Diet, High-Fat , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/prevention & control , Male , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/blood , P-Selectin/genetics , Plaque, Atherosclerotic , Platelet Adhesiveness , Platelet Aggregation , Platelet Transfusion , Signal Transduction , Time Factors
18.
J Immunol ; 195(1): 194-202, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26026056

ABSTRACT

We previously showed that LIGHT and its receptor herpes virus entry mediator (HVEM) are important for development of optimal CD4(+) Th1 cell immunity and resistance to primary Leishmania major infection in mice. In this study, we further characterized the contributions of this molecule in dendritic cell (DC) maturation, initiation, and maintenance of primary immunity and secondary anti-Leishmania immunity. Flow-cytometric studies showed that CD8α(+) DC subset was mostly affected by HVEM-Ig and lymphotoxin ß receptor-Ig treatment. LIGHT signaling is required at both the priming and the maintenance stages of primary anti-Leishmania immunity but is completely dispensable during secondary immunity in wild type mice. However, LIGHT blockade led to impaired IL-12 and IFN-γ responses and loss of resistance in healed CD40-deficient mice after L. major challenge. The protective effect of LIGHT was mediated primarily via its interaction with lymphotoxin ß receptor on CD8α(+) DCs. Collectively, our results show that although LIGHT is critical for maintenance of primary Th1 response, it is dispensable during secondary anti-Leishmania immunity in the presence of functional CD40 signaling as seen in wild type mice.


Subject(s)
CD40 Antigens/deficiency , Host-Pathogen Interactions/immunology , Leishmaniasis, Cutaneous/immunology , Th1 Cells/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/parasitology , Bone Marrow Cells/pathology , CD40 Antigens/genetics , CD40 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/parasitology , Dendritic Cells/pathology , Female , Gene Expression Regulation , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Leishmania major/immunology , Leishmaniasis, Cutaneous/genetics , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/pathology , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Mice , Signal Transduction , Th1 Cells/parasitology , Th1 Cells/pathology , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
19.
J Immunol ; 194(5): 2268-79, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25646303

ABSTRACT

The costimulatory molecule CD40 enhances immunity through several distinct roles in T cell activation and T cell interaction with other immune cells. In a mouse model of immunity to liver stage Plasmodium infection, CD40 was critical for the full maturation of liver dendritic cells, accumulation of CD8(+) T cells in the liver, and protective immunity induced by immunization with the Plasmodium yoelii fabb/f(-) genetically attenuated parasite. Using mixed adoptive transfers of polyclonal wild-type and CD40-deficient CD8(+) T cells into wild-type and CD40-deficient hosts, we evaluated the contributions to CD8(+) T cell immunity of CD40 expressed on host tissues including APC, compared with CD40 expressed on the CD8(+) T cells themselves. Most of the effects of CD40 could be accounted for by expression in the T cells' environment, including the accumulation of large numbers of CD8(+) T cells in the livers of immunized mice. Thus, protective immunity generated during immunization with fabb/f(-) was largely dependent on effective APC licensing via CD40 signaling.


Subject(s)
CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Liver/immunology , Malaria Vaccines/administration & dosage , Malaria/prevention & control , Plasmodium yoelii/immunology , Sporozoites/immunology , Adoptive Transfer , Animals , CD40 Antigens/deficiency , CD40 Antigens/genetics , CD8-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Dendritic Cells/immunology , Dendritic Cells/parasitology , Dendritic Cells/pathology , Female , Gene Deletion , Gene Expression , Hepatocytes/immunology , Hepatocytes/parasitology , Hepatocytes/pathology , Immunity, Innate , Liver/parasitology , Liver/pathology , Lymphocyte Activation , Malaria/immunology , Malaria/parasitology , Malaria/pathology , Mice , Mice, Inbred C57BL , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Signal Transduction , Sporozoites/chemistry , Vaccines, Attenuated
20.
Eur J Immunol ; 45(2): 574-83, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25521433

ABSTRACT

Thymus colonisation and thymocyte positioning are regulated by interactions between CCR7 and CCR9, and their respective ligands, CCL19/CCL21 and CCL25. The ligands of CCR7 and CCR9 also interact with the atypical receptor CCRL1 (also known as ACKR4), which is expressed in the thymus and has recently been reported to play an important role in normal αßT-cell development. Here, we show that CCRL1 is expressed within the thymic cortex, predominantly by MHC-II(low) CD40(-) cortical thymic epithelial cells and at the subcapsular zone by a population of podoplanin(+) thymic epithelial cells in mice. Interestingly, CCRL1 is also expressed by stromal cells which surround the pericytes of vessels at the corticomedullary junction, the site for progenitor cell entry and mature thymocyte egress from the thymus. We show that CCRL1 suppresses thymocyte progenitor entry into the thymus, however, the thymus size and cellularity are the same in adult WT and CCRL1(-/-) mice. Moreover, CCRL1(-/-) mice have no major perturbations in T-cell populations at different stages of thymic differentiation and development, and have a similar rate of thymocyte migration into the blood. Collectively, our findings argue against a major role for CCRL1 in normal thymus development and function.


Subject(s)
Epithelial Cells/metabolism , Lymphopoiesis/genetics , Receptors, CCR/genetics , Stromal Cells/metabolism , Thymocytes/metabolism , Thymus Gland/metabolism , Animals , CD40 Antigens/deficiency , CD40 Antigens/genetics , CD40 Antigens/immunology , Cell Differentiation , Cell Movement , Cellular Microenvironment , Epithelial Cells/cytology , Epithelial Cells/immunology , Female , Gene Expression Regulation, Developmental/immunology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Pericytes/cytology , Pericytes/immunology , Receptors, CCR/deficiency , Receptors, CCR/immunology , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Signal Transduction , Stem Cells/cytology , Stem Cells/immunology , Stromal Cells/cytology , Stromal Cells/immunology , Thymocytes/cytology , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/growth & development , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...