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










Publication year range
1.
Arthritis Res Ther ; 24(1): 284, 2022 12 28.
Article in English | MEDLINE | ID: mdl-36578056

ABSTRACT

BACKGROUND: Lupus nephritis (LN) is the most common and serious complication of systemic lupus erythematosus (SLE). LN pathogenesis is not fully understood. Axl receptor tyrosine kinase is upregulated and contributes to the pathogenic progress in LN. We have reported that Axl disruption attenuates nephritis development in mice. METHODS: In this study, we analyzed the gene expression profiles with RNA-seq using renal cortical samples from nephritic mice. Axl-KO mice were bred onto a B6.lpr spontaneous lupus background, and renal disease development was followed and compared to the Axl-sufficient B6.lpr mice. Finally, anti-glomerular basement membrane (anti-GBM) Ab-induced nephritic mice were treated with Axl small molecule inhibitor, R428, at different stages of nephritis development. Blood urine nitrogen levels and renal pathologies were evaluated. RESULTS: Transcriptome analysis revealed that renal Axl activation contributed to cell proliferation, survival, and motility through regulation of the Akt, c-Jun, and actin pathways. Spontaneous lupus-prone B6.lpr mice with Axl deficiency showed significantly reduced kidney damages and decreased T cell infiltration compared to the renal damage and T cell infiltration in Axl-sufficient B6.lpr mice. The improved kidney function was independent of autoAb production. Moreover, R428 significantly reduced anti-GBM glomerulonephritis at different stages of GN development compared to the untreated nephritic control mice. R428 administration reduced inflammatory cytokine (IL-6) production, T cell infiltration, and nephritis disease activity. CONCLUSIONS: Results from this study emphasize the important role of Axl signaling in LN and highlight Axl as an attractive target in LN.


Subject(s)
Glomerulonephritis , Lupus Erythematosus, Systemic , Lupus Nephritis , Animals , Mice , Glomerulonephritis/drug therapy , Glomerulonephritis/complications , Glomerulonephritis/metabolism , Lupus Nephritis/pathology , Kidney/pathology , Lupus Erythematosus, Systemic/metabolism , Cell Proliferation , Mice, Inbred MRL lpr
2.
Cells ; 11(12)2022 06 09.
Article in English | MEDLINE | ID: mdl-35740998

ABSTRACT

Axl receptor tyrosine kinase expression in the kidney contributes to a variety of inflammatory renal disease by promoting glomerular proliferation. Axl expression in the kidney is negligible in healthy individuals but upregulated under inflammatory conditions. Little is known about Axl transcriptional regulation. We analyzed the 4.4 kb mouse Axl promoter region and found that many transcription factor (TF)-binding sites and regulatory elements are located within a 600 bp fragment proximal to the translation start site. Among four TFs (Sp1, Ap1, MZF1, and Ep300) identified, Sp1 was the most potent TF that promotes Axl expression. Luciferase assays confirmed the siRNA results and revealed additional mechanisms that regulate Axl expression, including sequences encoding a 5'-UTR mini-intron and potential G-quadruplex forming regions. Deletion of the Axl 5'-UTR mini-intron resulted in a 3.2-fold increases in luciferase activity over the full-length UTR (4.4 kb Axl construct). The addition of TMPyP4, a G-quadruplex stabilizer, resulted in a significantly decreased luciferase activity. Further analysis of the mouse Axl 3'-UTR revealed a miRNA-34a binding site, which inversely regulates Axl expression. The inhibitory role of miRNA-34a in Axl expression was demonstrated in mesangial cells using miRNA-34a mimicry and in primary kidney cells with IL-6 stimulated STAT3 activation. Taken together, Axl expression in mouse kidney is synergistically regulated by multiple factors, including TFs and secondary structures, such as mini-intron and G-quadruplex. A unique IL6/STAT3/miRNA-34a pathway was revealed to be critical in inflammatory renal Axl expression.


Subject(s)
Mesangial Cells , MicroRNAs , Animals , Cell Line, Tumor , Interleukin-6 , Mesangial Cells/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Small Interfering
3.
J Leukoc Biol ; 111(3): 563-571, 2022 03.
Article in English | MEDLINE | ID: mdl-34218441

ABSTRACT

Glomerulonephritis (GN), an important pathologic feature of many renal diseases, is frequently characterized by mesangial cell proliferation. We and others have previously shown that the TAM family receptor tyrosine kinases Axl, Mer, and Tyro-3 contribute to cell survival, proliferation, migration, and clearance of apoptotic cells (ACs); that Axl contributes to GN by promoting mesangial cell proliferation; and that small molecule inhibition of Axl ameliorates nephrotoxic serum-induced GN in mice. We now show that stimulation of renal mesangial cell Axl causes a modest increase in intracellular Ca2+ and activates NF-κB, mTOR, and the mTOR-containing mTORC1 complex, which phosphorylates the ribosomal protein S6. Axl-induction of Akt activation is upstream of NF-κB and mTOR activation, which are mutually codependent. Axl-induced NF-κB activation leads to Bcl-xl up-regulation. Axl is more important than Mer at mediating AC phagocytosis by mesangial cells, but less important than Mer at mediating phagocytosis of ACs by peritoneal macrophages. Taken together, our data suggest the possibility that Axl mediates mesangial cell phagocytosis of ACs and promotes mesangial cell proliferation by activating NF-κB and mTORC1.


Subject(s)
Glomerulonephritis , Proto-Oncogene Proteins c-akt , Animals , Cell Proliferation , Female , Humans , Intercellular Signaling Peptides and Proteins , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , NF-kappa B , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , TOR Serine-Threonine Kinases , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
4.
Arthritis Res Ther ; 22(1): 133, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32503684

ABSTRACT

BACKGROUND: The mechanisms involved in the pathogenesis of autoimmune disorders, including systemic lupus erythematosus (SLE), have not been fully elucidated. Some of these mechanisms involve epigenetic regulation of gene expression. The histone methyltransferase Ezh2 contributes to epigenetic regulation of gene expression, is highly expressed in germinal center (GC) B cells and follicular T helper (TFH) cells, and may be involved in lupus pathogenesis. METHODS: The murine bm12 model of lupus-like chronic graft versus host disease (cGVHD) was induced by intra-peritoneal injection of negatively isolated allogeneic CD4+ T cells. Lupus-like disease development was monitored by ELISA determination of serum anti-dsDNA and anti-chromatin antibody titers. Immune cell activation and Ezh2 expression were evaluated by flow cytometry and Western blotting. RESULTS: Decreased autoantibody production and GC formation are observed when Ezh2-deficient CD4+ T cells are used instead of wild-type (WT) to induce cGVHD and when mice that receive allogeneic WT donor T cells to induce cGVHD are treated with GSK503, an Ezh2-specific inhibitor. In the bm12 cGVHD model, WT donor T cells are normally fully activated 1 week after infusion into an allogeneic host, exhibit a TFH cell (PD-1hi/CXCR5hi) phenotype with upregulated Ezh2, and activate B cells to form germinal centers (GCs). In contrast, Ezh2-deficient donor T cells generate fewer TFH cells that fail to activate B cells or promote GC formation. Despite similar T-independent, LPS-induced B cell responses, OVA-immunized CD4.Ezh2-KO mice had a skewed low-affinity IgM phenotype in comparison to similarly treated WT mice. In addition, early after OVA immunization, more CD4+ T cells from B6.CD4.Ezh2-KO mice had a CD44lo/CD62Llo phenotype, which suggests arrested or delayed activation, than CD4+ T cells from ovalbumin-immunized B6.WT mice. CONCLUSION: Ezh2 gene deletion or pharmacological Ezh2 inhibition suppresses autoantibody production and GC formation in bm12 lupus-like cGVHD and decreases affinity maturation and isotype switching in response to immunization with a T cell-dependent antigen. Ezh2 inhibition may be useful for the treatment of lupus and other autoimmune disorders.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Lupus Erythematosus, Systemic , Animals , Epigenesis, Genetic , Germinal Center , Graft vs Host Disease/genetics , Lupus Erythematosus, Systemic/genetics , Mice , T-Lymphocytes , T-Lymphocytes, Helper-Inducer
5.
J Vis Exp ; (147)2019 05 24.
Article in English | MEDLINE | ID: mdl-31180346

ABSTRACT

Cell apoptosis is a natural process and plays a critical role in embryonic development, homeostatic regulation, immune tolerance induction, and resolution of inflammation. Accumulation of apoptotic debris in the body may trigger chronic inflammatory responses that lead to systemic autoimmune diseases over time. Impaired apoptotic cell clearance has been implicated in a variety of autoimmune diseases. Apoptotic clearance is a complex process rarely detected under physiological conditions. It involves abundant surface receptors and signaling molecules. Studying the process of apoptotic cell clearance provides insightful molecular mechanisms and subsequent biological responses, which may lead to the development of new therapeutics. Here, we describe protocols for the induction of apoptotic thymocytes, the preparation of peritoneal macrophages, and the analysis of apoptotic cell clearance by flow cytometry and microscopy. All cells will undergo apoptosis at a certain stage, and many residential and circulating cells can uptake apoptotic debris. Therefore, the protocol described here can be used in many applications to characterize apoptotic cell binding and ingestion by many other cell types.


Subject(s)
Macrophages/metabolism , Thymocytes/metabolism , Animals , Apoptosis , Humans , Mice
6.
J Autoimmun ; 93: 37-44, 2018 09.
Article in English | MEDLINE | ID: mdl-29895432

ABSTRACT

Glomerulonephritis (GN) is a typical lesion in autoantibody and immune complex disorders, including SLE. Because the Gas6/Axl pathway has been implicated in the pathogenesis of many types of GN, targeting this pathway might ameliorate GN. Consequently, we have studied the efficacy and mechanism of R428, a potent selective Axl inhibitor, in the prevention of experimental anti-GBM nephritis. Axl upregulation was investigated with Sp1/3 siRNA in the SV40-transformed mesangial cells. For Axl inhibition, a daily dose of R428 (125 mg/kg) or vehicle was administered orally. GN was induced with anti-GBM sera. Renal disease development was followed by serial blood urine nitrogen (BUN) determinations and by evaluation of kidney histology at the time of sacrifice. Axl-associated signaling proteins were analyzed by Western blotting and inflammatory cytokine secretion was analyzed by Proteome array. SiRNA data revealed the transcription factor Sp1 to be an important regulator of mesangial Axl expression. Anti-GBM serum induced severe nephritis with azotemia, protein casts and necrotic cell death. R428 treatment diminished renal Axl expression and improved kidney function, with significantly decreased BUN and glomerular proliferation. R428 treatment inhibited Axl and significantly decreased Akt phosphorylation and renal inflammatory cytokine and chemokine expression; similar effects were observed in anti-GBM antiserum-treated Axl-KO mice. These studies support a role for Axl inhibition in glomerulonephritis.


Subject(s)
Benzocycloheptenes/pharmacology , Immunologic Factors/pharmacology , Lupus Nephritis/drug therapy , Mesangial Cells/drug effects , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Triazoles/pharmacology , Administration, Oral , Animals , Antibodies/administration & dosage , Cell Line, Transformed , Drug Administration Schedule , Gene Expression Regulation , Glomerular Basement Membrane/drug effects , Glomerular Basement Membrane/immunology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Lupus Nephritis/chemically induced , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Mesangial Cells/immunology , Mesangial Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/immunology , Signal Transduction , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/immunology , Sp3 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/immunology , Axl Receptor Tyrosine Kinase
7.
J Leukoc Biol ; 103(4): 709-717, 2018 04.
Article in English | MEDLINE | ID: mdl-29350876

ABSTRACT

Endotoxin induces a variety of proinflammatory mediators and plays a crucial role in kidney inflammation. The receptor tyrosine kinase, Mer, diminishes renal inflammation by attenuating inflammatory responses. We previously reported that Mer is predominantly expressed on glomerular endothelial cells (GECs) and that Mer deficiency is associated with increased renal inflammation when mice are challenged with nephrotoxic serum. We consequently hypothesized that Mer signaling down-regulates LPS-driven inflammatory responses in GECs. To test this hypothesis, primary GECs were isolated from the kidneys of Mer-KO and wild-type (WT) control mice. LPS treatment induced Akt and STAT3 activation along with Bcl-xl up-regulation in WT GECs; these responses were all increased in Mer-deficient GECs. In addition, STAT1 and ERK1/2 up-regulation and activation were observed in Mer-KO GECs exposed to LPS. In contrast, expression of the inhibitory signaling molecule, suppressor of cytokine signaling-3 (SOCS-3), was much higher in LPS-stimulated WT than Mer-deficient GECs. Deficiency of Mer was also associated with significantly increased NF-κB expression and activation. These observations indicate that Mer functions as an intrinsic feedback inhibitor of inflammatory mediator-driven immune responses in GECs during kidney injury and suggest a new therapeutic strategy for glomerular diseases.


Subject(s)
Endothelial Cells/immunology , Gene Expression Regulation , Inflammation/prevention & control , Kidney Glomerulus/immunology , c-Mer Tyrosine Kinase/physiology , Animals , Cytokines/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Inflammation/chemically induced , Inflammation/metabolism , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Signal Transduction
8.
J Immunol ; 197(6): 2187-94, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27527599

ABSTRACT

Glomerulonephritis is one of the most severe manifestations of systemic lupus erythematosus, with considerable morbidity and mortality. There remains a major unmet need for successful management of lupus nephritis. TAM family receptor tyrosine kinases (Mer and Axl) play an important role in the maintenance of immune homeostasis in the kidney. Mer is constitutively expressed in the glomeruli; Axl expression is inducible in glomeruli under inflammatory conditions. To investigate the distinct functions of Axl and Mer in lupus nephritis, we compared the severity of nephrotoxic serum glomerulonephritis in wild-type (WT), Axl-knockout (KO), Mer-KO, and Axl/Mer-KO mice. Mer-KO mice developed severe glomerulonephritis, with significantly decreased survival and increased blood urea nitrogen levels compared with WT mice given the same treatment. However, nephrotoxic serum-treated Axl-KO mice had significantly increased survival rates and improved renal function compared with similarly treated WT, Mer-KO, and Axl/Mer-KO mice. Interestingly, mice lacking both Axl and Mer developed kidney inflammation comparable to WT mice. Western blot analysis revealed significantly increased Stat3 phosphorylation and caspase-1 activation in the kidneys of nephritic Mer-KO mice. In contrast, Axl-deficient nephrotoxic serum-injected mice showed decreased Akt phosphorylation and Bcl-xL upregulation. Thus, the reciprocal activation of Axl and Mer receptor tyrosine kinases has a major impact on the outcome of renal inflammation.


Subject(s)
Lupus Nephritis/etiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Caspase 1/physiology , Cell Proliferation , Cytokines/biosynthesis , Intercellular Signaling Peptides and Proteins/physiology , Kidney/metabolism , Kidney Glomerulus/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/antagonists & inhibitors , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
9.
Arthritis Rheumatol ; 68(11): 2697-2707, 2016 11.
Article in English | MEDLINE | ID: mdl-27110677

ABSTRACT

OBJECTIVE: Increased levels of type I interferon (IFN) and type I IFN-regulated genes are found in patients with systemic lupus erythematosus (SLE) and may be central to its pathogenesis. Mitochondrial antiviral signaling protein (MAVS) is a key regulator of type I IFN that undergoes a dramatic prion-like aggregation and self propagates the activation signal from viral RNA to amplify downstream IFN production. We undertook this study to determine whether such MAVS aggregates might play a role in the sustained increased production of type I IFN in SLE. METHODS: Peripheral blood mononuclear cells were isolated and mitochondrial extracts were prepared. MAVS aggregation was detected by semidenatured agarose gel electrophoresis and confirmed by immunofluorescence staining. MAVS-associated signaling proteins were analyzed by Western blotting. MAVS aggregation-associated gene expression signature was analyzed by microarray. RESULTS: In blood cells from 22 of 67 SLE patients, essentially all MAVS was in a high molecular weight aggregated form. None of 6 rheumatoid arthritis patients and only 3 of 33 healthy controls had abnormal MAVS. Compared to MAVS aggregate-negative patients, MAVS aggregate-positive SLE patients had significantly higher serum levels of IFNß and significantly increased levels of autoantibodies against Sm and U1 RNP. Gene array data revealed a characteristic gene expression pattern in these patients, with altered expression of genes involved in IFN signaling and membrane trafficking. CONCLUSION: Persistent MAVS aggregates may lead to increased type I IFN production and result in unmitigated signals leading to autoimmunity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Interferon Type I/metabolism , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/metabolism , Protein Aggregation, Pathological/metabolism , Adult , Antibodies, Antinuclear/immunology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Autoantibodies/immunology , Blotting, Western , Case-Control Studies , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Interferon Type I/immunology , Interferon-beta/immunology , Leukocytes, Mononuclear/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Male , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Middle Aged , Mitochondrial Proteins/genetics , Protein Aggregation, Pathological/immunology , Receptors, Complement/genetics , Ribonucleoprotein, U1 Small Nuclear/immunology , Tissue Array Analysis , Transcriptome , Ubiquitin-Protein Ligases/genetics , snRNP Core Proteins/immunology
10.
J Immunol ; 195(9): 4136-43, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26392462

ABSTRACT

Systemic lupus erythematosus (SLE) is a complex multisystem autoimmune disease, characterized by a spectrum of autoantibodies that target multiple cellular components. Glomerulonephritis is a major cause of morbidity in patients with SLE. Little is known about the pathogenesis of SLE renal damage and compromised renal function. Activation of both Stat1 and Stat3 has been reported in lupus and lupus nephritis. The reciprocal activation of these two transcription factors may have a major impact on renal inflammation. To study the role of Stat1 in a lupus model, we induced lupus-like chronic graft-versus-host disease (cGVHD) in Stat1-knockout (KO) and wild-type (WT) mice by i.p. injection of class II-disparate bm12 splenocytes. WT recipients of these alloreactive cells developed anti-dsDNA autoantibodies starting at week 2 as expected, with a decline after week 4. In contrast, Stat1-KO hosts exhibited a prolonged and significant increase of anti-dsDNA autoantibody responses compared with WT mice (week 4 to week 8). Increased autoantibody titers were accompanied by increased proteinuria and mortality in the cGVHD host mice lacking Stat1. Further analysis revealed expression and activation of Stat3 in the glomeruli of Stat1-KO host mice but not WT mice with cGVHD. Glomerular Stat3 activity in the Stat1-KO mice was associated with increased IL-6 and IFN-γ secretion and macrophage infiltration. Interactions between Stat1 and Stat3 thus appear to be crucial in determining the severity of lupus-like disease in the cGVHD model.


Subject(s)
Graft vs Host Disease/immunology , Lupus Nephritis/etiology , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/physiology , Animals , Autoantibodies/biosynthesis , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Chronic Disease , Interferon-gamma/physiology , Interleukin-6/metabolism , Lupus Nephritis/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL
11.
J Autoimmun ; 53: 78-84, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24768065

ABSTRACT

The Mertk receptor tyrosine kinase facilitates macrophage and DC apoptotic-cell clearance and regulates immune tolerance. Mertk may also contribute to B-cell activation, because Mertk-KO mice fail to develop autoantibodies when allo-activated by T cells. We investigated this possibility with a well-characterized model in which injection of mice with goat anti-IgD antibody causes membrane IgD cross-linking that induces T-independent B cell activation and antigen presentation to T cells. Goat anti-mouse IgD antibody-injected C57BL/6 Mertk-KO mice had normal initial B cell activation and proliferation, but significantly lower T cell activation and proliferation, as well as lower IgE and IgG anti-goat IgG responses, as compared to C57BL/6 WT controls. B cell antigen processing, analyzed by evaluating B cell fluorescence following injection of monoclonal anti-IgD antibody labeled with biotin or FITC, was comparable between Mertk-KO mice and WT mice. IgD Ab primed B cells from Mertk-KO mice exhibited significantly lower ability in activating memory T cells isolated from WT mice injected with the same antigen 10 days before. These observations suggest that Mertk expression is required for optimal B-cell antigen presentation, which is, in turn, required in this model for optimal T cell activation and subsequent T cell-dependent B cell differentiation.


Subject(s)
B-Lymphocytes/immunology , Cell Communication/immunology , Immunoglobulin D/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Antigen, B-Cell/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/cytology , Cell Communication/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , Immunoglobulin D/genetics , Immunologic Capping/genetics , Immunologic Capping/immunology , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Antigen, B-Cell/genetics , T-Lymphocytes/cytology , c-Mer Tyrosine Kinase
12.
J Autoimmun ; 39(4): 412-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22854104

ABSTRACT

Activation and migration of marginal zone B (MZB) cells into follicular (FO) regions of the spleen has been proposed as one of the mechanisms that regulate the development of autoreactive B cells. The mer receptor tyrosine kinase (Mertk) mediates apoptotic cell clearance and regulates activation and cytokine secretion. In the well-studied class II chronic GVH model of bm12 cells into B6 hosts, we observed that Mertk deficient B6 mice did not generate autoantibodies in response to this allogeneic stimulus. We posited that Mertk is important in MHC-II-mediated B cell signaling. In the present study, we show that B cells from Mertk(-/-) mice but not WT B6 mice exhibited decreased calcium mobilization and tyrosine phosphorylation when stimulated by MHC-II cross-linking. The finding that Mertk was important for class II signaling in B cells was further supported by the preponderance of a-allotype autoantibodies in cGVH in RAG-KO mice reconstituted with a mixture of bone marrow from Mertk(-/-) mice (b-allotype) and C20 mice (a-allotype). MZB cells from Mertk(-/-) mice were unable to down regulate surface CD1d expression and subsequent inclusion in the MZ, associated with significantly lower germinal center responses compared to MZB cells from WT. Moreover, Mertk(-/-) mice treated with an anti-CD1d down regulating antibody responded significantly to bm12 cells, while no response was observed in Mertk(-/-) mice treated with control antibodies. Taken together, these findings extend the role of Mertk to include CD1d down regulation on MZB cells, a potential mechanism limiting B cell activation in cGVH.


Subject(s)
Antigens, CD1d/immunology , B-Lymphocytes/immunology , Germinal Center/immunology , Graft vs Host Disease/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Antigens, CD1d/genetics , Autoantibodies/biosynthesis , Autoantibodies/immunology , B-Lymphocytes/pathology , Calcium/immunology , Calcium/metabolism , Chronic Disease , Gene Expression/immunology , Germinal Center/pathology , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Humans , Immunoglobulin Allotypes/genetics , Immunoglobulin Allotypes/immunology , Lymphocyte Activation , Mice , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Spleen/immunology , Spleen/pathology , c-Mer Tyrosine Kinase
13.
J Immunol ; 185(10): 5859-68, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20952679

ABSTRACT

Germinal centers (GCs) are specialized microenvironments that generate high-affinity Ab-forming cells (AFCs) and memory B cells. Many B cells undergo apoptosis during B cell clonal selection in GCs. Although the factors that regulate the AFC and GC responses are not precisely understood, it is widely believed that dysregulated AFCs and GCs contribute to autoimmunity. The Mer receptor tyrosine kinase (Mer) facilitates macrophage clearance of apoptotic cells. The Tyro-3, Axl, and Mer receptors, including Mer, suppress TLRs and cytokine-mediated inflammatory responses. We report in this study that tingible body macrophages (TBMφs) in GCs express Mer. Compared to C57BL/6 (B6) controls, Mer-deficient (Mer(-/-)) mice had significantly higher AFC, GC, and Th1-skewed IgG2 Ab (especially IgG2c) responses against the T cell-dependent Ag (4-hydroxy-3-nitrophenyl) acetyl-chicken γ globulin. Mer(-/-) mice had a significantly higher percentage of GC B cells on days 9, 14, and 21 postimmunization compared with B6 controls. Significantly increased numbers of apoptotic cells accumulated in Mer(-/-) GCs than in B6 GCs, whereas the number of TBMφs remained similar in both strains. Our data are the first, to our knowledge, to demonstrate a critical role for Mer in GC apoptotic cell clearance by TBMφs and have interesting implications for Mer in the regulation of B cell tolerance operative in the AFC and GC pathways.


Subject(s)
Antibody-Producing Cells/immunology , Apoptosis/immunology , Macrophages/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Antibody Formation/immunology , Antibody-Producing Cells/metabolism , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Germinal Center/cytology , Germinal Center/immunology , Immunohistochemistry , In Situ Nick-End Labeling , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Spleen/cytology , Spleen/immunology , c-Mer Tyrosine Kinase
14.
Clin Immunol ; 136(2): 236-44, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20444650

ABSTRACT

The Mer receptor tyrosine kinase is strongly expressed in the glomerulus. We wondered if this molecule might modify immune-mediated glomerular disease through its functions as a receptor for apoptotic cells and immunoregulatory molecule. Mer-knockout (KO) mice showed decreased survival rate and greatly increased proteinuria and serum urea levels compared to wild type (WT) mice by day 3 after injection of NTS. Their glomeruli were hyperplastic and later became necrotic. In the glomerulus of WT mice, a significant increase of Mer expression was observed. Apoptotic bodies were evident in NTS-treated Mer-KO kidneys, but not in normal controls. NTS-treated Mer-KO mice had massive neutrophil infiltration and inflammatory cytokine expression. Mer thus has a critical role in attenuating renal inflammation, both as a receptor for apoptotic cells and as a molecule that downregulates inflammation.


Subject(s)
Kidney Glomerulus/enzymology , Nephritis/enzymology , Nephritis/immunology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis , Chemokine CCL2/metabolism , Cytokines/metabolism , Gene Expression Regulation, Enzymologic , Immune Sera/toxicity , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Mice , Mice, Knockout , Neutrophils , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Sheep , Tumor Necrosis Factor-alpha/metabolism , c-Mer Tyrosine Kinase
15.
Clin Immunol ; 133(1): 138-44, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19631584

ABSTRACT

The Mer receptor tyrosine kinase is both an important mediator of apoptotic cell phagocytosis and a regulator of macrophage and DC cytokine production. Since phenotypically distinguishable macrophages are known to have different functions, we have examined Mer expression of murine splenic macrophages. We also used serum deficient in the Mer ligand, growth arrest-specific protein 6 (Gas6) to define better the role of this Mer ligand in macrophage function. By immunofluorescence staining, we found Mer to be strongly expressed in splenic red pulp, largely on platelets. We also found Mer expression on marginal zone macrophages. Strikingly, all tingible body macrophages bore Mer. In functional phagocytosis assays of apoptotic cells, Gas6 appeared to be the sole ligand for Mer, and this system accounted for about 30% of splenic macrophage phagocytosis of apoptotic cells. Taken together, the expression pattern of Mer on macrophage subpopulations in the spleen and its Gas6-dependent role in macrophage phagocytosis suggest an important role for Mer in the modulation of immune responses.


Subject(s)
Apoptosis/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Blood Platelets/immunology , Blood Platelets/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Intercellular Signaling Peptides and Proteins/immunology , Ligands , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phagocytosis/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , c-Mer Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...