Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
J Immunol ; 212(7): 1142-1149, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38372645

ABSTRACT

How reparative processes are coordinated following injury is incompletely understood. In recent studies, we showed that autocrine C3a and C5a receptor (C3ar1 and C5ar1) G protein-coupled receptor signaling plays an obligate role in vascular endothelial growth factor receptor 2 growth signaling in vascular endothelial cells. We documented the same interconnection for platelet-derived growth factor receptor growth signaling in smooth muscle cells, epidermal growth factor receptor growth signaling in epidermal cells, and fibroblast growth factor receptor signaling in fibroblasts, indicative of a generalized cell growth regulatory mechanism. In this study, we examined one physiological consequence of this signaling circuit. We found that disabling CD55 (also known as decay accelerating factor), which lifts restraint on autocrine C3ar1/C5ar1 signaling, concomitantly augments the growth of each cell type. The mechanism is heightened C3ar1/C5ar1 signaling resulting from the loss of CD55's restraint jointly potentiating growth factor production by each cell type. Examination of the effect of lifted CD55 restraint in four types of injury (burn, corneal denudation, ear lobe puncture, and reengraftment of autologous skin) showed that disabled CD55 function robustly accelerated healing in all cases, whereas disabled C3ar1/C5ar1 signaling universally retarded it. In wild-type mice with burns or injured corneas, applying a mouse anti-mouse CD55 blocking Ab (against CD55's active site) to wounds accelerated the healing rate by 40-70%. To our knowledge, these results provide new insights into mechanisms that underlie wound repair and open up a new tool for accelerating healing.


Subject(s)
CD55 Antigens , Endothelial Cells , Vascular Endothelial Growth Factor A , Wound Healing , Animals , Mice , Endothelial Cells/metabolism , Signal Transduction , Skin , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/physiology , CD55 Antigens/antagonists & inhibitors , CD55 Antigens/metabolism
2.
J Immunol ; 211(9): 1359-1366, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37756526

ABSTRACT

Demethylation of the T regulatory cell (Treg)-specific demethylation region (TSDR) of the Foxp3 gene is the hallmark of Foxp3+ Treg stability, but the cellular signaling that programs this epigenetic state remains undefined. In this article, we show that suppressed C3a and C5a receptor (C3ar1/C5ar1) signaling in murine Tregs plays an obligate role. Murine C3ar1-/-C5ar1-/- Foxp3+ cells showed increased suppressor of cytokine signaling 1/2/3 expression, vitamin C stabilization, and ten-eleven translocation (TET) 1, TET2, and TET3 expression, all of which are linked to Treg stability. C3ar1-/-C5ar1-/- Foxp3+ cells additionally were devoid of BRD4 signaling that primes Th17 cell lineage commitment. Orally induced OVA-specific C3ar1-/-C5ar1-/- Foxp3+ OT-II Tregs transferred to OVA-immunized wild-type recipients remained >90% Foxp3+ out to 4 mo, whereas identically generated CD55-/- (DAF-/-) Foxp3+ OT-II Tregs (in which C3ar1/C5ar1 signaling is potentiated) lost >75% of Foxp3 expression by 14 d. After 4 mo in vivo, the C3ar1-/-C5ar1-/- Foxp3+ OT-II Tregs fully retained Foxp3 expression even with OVA challenge and produced copious TGF-ß and IL-10. Their TSDR was demethylated comparably with that of thymic Tregs. They exhibited nuclear translocation of NFAT and NF-κB reported to stabilize thymic Tregs by inducing hairpin looping of the TSDR to the Foxp3 promoter. Thus, disabled CD4+ cell C3ar1/C5ar1 signaling triggers the sequential cellular events that lead to demethylation of the Foxp3 TSDR.


Subject(s)
DNA Methylation , T-Lymphocytes, Regulatory , Mice , Animals , Transcription Factors/metabolism , Gene Expression Regulation , Receptor, Anaphylatoxin C5a/metabolism , Nuclear Proteins/genetics , Demethylation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism
3.
Front Immunol ; 14: 1290684, 2023.
Article in English | MEDLINE | ID: mdl-38406578

ABSTRACT

The transcription factor Kruppel-like factor 4 (KLF4) regulates the expression of immunosuppressive and anti-thrombotic proteins. Despite its importance in maintaining homeostasis, the signals that control its expression and the mechanism of its transactivation remain unclarified. CD55 [aka decay accelerating factor (DAF)], now known to be a regulator of T and B cell responses, biases between pro- and anti-inflammatory processes by controlling autocrine C3a and C5a receptor (C3ar1/C5ar1) signaling in cells. The similarity in CD55's and KLF4's regulatory effects prompted analyses of their functional relationship. In vascular endothelial cells (ECs), CD55 upregulation accompanied KLF4 expression via a p-CREB and CREB Binding Protein (CBP) mechanism. In both ECs and macrophages, CD55 expression was essential for KLF4's downregulation of pro-inflammatory/pro-coagulant proteins and upregulation of homeostatic proteins. Mechanistic studies showed that upregulation of KLF4 upregulated CD55. The upregulated CD55 in turn enabled the recruitment of p-CREB and CBP to KLF4 needed for its transcription. Activation of adenylyl cyclase resulting from repression of autocrine C3ar1/C5ar1 signaling by upregulated CD55 concurrently led to p-CREB and CBP recruitment to KLF4-regulated genes, thereby conferring KLF4's transactivation. Accordingly, silencing CD55 in statin-treated HUVEC disabled CBP transfer from the E-selectin to the eNOS promoter. Importantly, silencing CD55 downregulated KLF4's expression. It did the same in untreated HUVEC transitioning from KLF4low growth to KLF4hi contact inhibition. KLF4's and CD55's function in ECs and macrophages thus are linked via a novel mechanism of gene transactivation. Because the two proteins are co-expressed in many cell types, CD55's activity may be broadly tied to KLF4's immunosuppressive and antithrombotic activities.


Subject(s)
Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Endothelial Cells/metabolism , Up-Regulation , Promoter Regions, Genetic
4.
Am J Pathol ; 192(2): 361-378, 2022 02.
Article in English | MEDLINE | ID: mdl-35144762

ABSTRACT

As per the classical view of the coagulation system, it functions solely in plasma to maintain hemostasis. An experimental approach modeling vascular reconstitution was used to show that vascular endothelial cells (ECs) endogenously synthesize coagulation factors during angiogenesis. Intracellular thrombin generated from this synthesis promotes the mitotic function of vascular endothelial cell growth factor A (VEGF-A). The thrombin concurrently cleaves C5a from EC-synthesized complement component C5 and unmasks the tethered ligand for EC-expressed protease-activated receptor 4 (PAR4). The two ligands jointly trigger EC C5a receptor-1 (C5ar1) and PAR4 signaling, which together promote VEGF receptor 2 growth signaling. C5ar1 is functionally associated with PAR4, enabling C5a or thrombin to elicit Gαi and/or Gαq signaling. EC coagulation factor and EC complement component synthesis concurrently down-regulate with contact inhibition. The connection of these processes with VEGF receptor 2 signaling provides new insights into mechanisms underlying angiogenesis. Knowledge of endogenous coagulation factor/complement component synthesis and joint PAR4/C5ar1 signaling could be applied to other cell types.


Subject(s)
Blood Coagulation Factors/biosynthesis , Endothelial Cells/metabolism , Neovascularization, Physiologic , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Proteinase-Activated/metabolism , Signal Transduction , Animals , Blood Coagulation Factors/genetics , Female , Male , Mice , Mice, Knockout , Receptor, Anaphylatoxin C5a/genetics , Receptors, Proteinase-Activated/genetics
5.
FASEB J ; 34(2): 2105-2125, 2020 02.
Article in English | MEDLINE | ID: mdl-31908021

ABSTRACT

How receptor tyrosine kinase (RTK) growth signaling is controlled physiologically is incompletely understood. We have previously provided evidence that the survival and mitotic activities of vascular endothelial cell growth factor receptor-2 (VEGFR2) signaling are dependent on C3a/C5a receptor (C3ar1/C5ar1) and IL-6 receptor (IL-6R)-gp130 joint signaling in a physically interactive platform. Herein, we document that the platelet derived and epidermal growth factor receptors (PDGFR and EGFR) are regulated by the same interconnection and clarify the mechanism underlying the dependence. We show that the joint signaling is required to overcome dominant restraint on RTK function by the combined repression of tonically activated PHLPP, SOCS1/SOCS3, and CK2/Fyn dependent PTEN. Signaling studies showed that augmented PI-3KÉ£ activation is the process that overcomes the multilevel growth restraint. Live-cell flow cytometry and single-particle tracking indicated that blockade of C3ar1/C5ar1 or IL-6R signaling suppresses RTK growth factor binding and RTK complex formation. C3ar1/C5ar1 blockade abrogated growth signaling of four additional RTKs. Active relief of dominant growth repression via joint C3ar1/C5ar1 and IL-6R joint signaling thus enables RTK mitotic/survival signaling.


Subject(s)
Endothelial Cells/metabolism , PTEN Phosphohydrolase/metabolism , Phosphoprotein Phosphatases/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Complement/metabolism , Receptors, Interleukin-6/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Line , Endothelial Cells/cytology , Genes, Dominant , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , Phosphoprotein Phosphatases/genetics , Receptor, Anaphylatoxin C5a/genetics , Receptors, Complement/genetics , Receptors, Interleukin-6/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
6.
J Immunol ; 203(2): 379-388, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31217324

ABSTRACT

The involvement of complement in B2 cell responses has been regarded as occurring strictly via complement components in plasma. In this study, we show that Ab production and class switch recombination (CSR) depend on autocrine C3a and C5a receptor (C3ar1/C5ar1) signaling in B2 cells. CD40 upregulation, IL-6 production, growth in response to BAFF or APRIL, and AID/Bcl-6 expression, as well as follicular CD4+ cell CD21 production, all depended on this signal transduction. OVA immunization of C3ar1-/-C5ar1-/- mice elicited IgM Ab but no other isotypes, whereas decay accelerating factor (Daf1)-/- mice elicited more robust Ab production and CSR than wild-type (WT) mice. Comparable differences occurred in OVA-immunized µMT recipients of WT, C3ar1-/-C5ar1-/- , and Daf1-/- B2 cells and in hen egg lysozyme-immunized µMT recipients of MD4 B2 cells on each genetic background. B2 cells produced factor I and C3 and autophosphorylated CD19. Immunized C3-/-C5-/- recipients of WT MD4 bone marrow efficiently produced Ab. Thus, B2 cell-produced complement participates in B2 cell activation.


Subject(s)
Autocrine Communication/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Complement/immunology , Animals , Antigens, CD19/immunology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Complement System Proteins/immunology , Interleukin-6/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology
7.
Am J Pathol ; 189(7): 1386-1401, 2019 07.
Article in English | MEDLINE | ID: mdl-31103439

ABSTRACT

Recent studies traced inflammatory bowel disease in some patients to deficiency of CD55 [decay-accelerating factor (DAF)], but the mechanism underlying the linkage remained unclear. Herein, we studied the importance of DAF in enabling processes that program tolerance in the gut and the eye, two immune-privileged sites where immunosuppressive responses are continuously elicited. Unlike oral feeding or ocular injection of ovalbumin in wild-type (WT) mice, which induced dominant immune tolerance, identical treatment of DAF-/- mice or DAF-/- to WT bone marrow chimeras did not. While 10% to 30% of mesenteric and submandibular lymph node CD4+ cells became robust T-regulatory cells (Tregs) in WT forkhead box P3 (Foxp3)-green fluorescent protein mice, few in either site became Tregs with little suppressor activity in DAF-/- Foxp3-green fluorescent protein mice. Phenotyping of CD103+ dendritic cells (DCs) from the ovalbumin-fed DAF-/- mice showed impaired expression of inducer of costimulation (ICOS) ligand, programmed death receptor 1-ligand 1 (PD1-L1), CxxxC chemokine receptor 1 (Cx3CR1), CCR7, and CCR9. Analyses of elicited DAF-/- Foxp3+ Tregs showed reduced expression of interferon regulatory factor 8 (IRF-8)/aldehyde dehydrogenase 1 family member A2 (Aldh1a2) and glycoprotein A repetitions predominant/latency-associated protein associated with Treg transforming growth factor-ß production and presentation, as well as integrin ß6/integrin ß8 associated with Treg and CD103+ DC transforming growth factor-ß release. Thus, DAF is required for the properties of CD103+ DCs and their naïve CD4+ cell partners that together program tolerance.


Subject(s)
Antigens, CD/immunology , Autoimmune Diseases/immunology , CD55 Antigens/immunology , Dendritic Cells/immunology , Immune Tolerance , Integrin alpha Chains/immunology , Animals , Antigens, CD/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , CD55 Antigens/genetics , Dendritic Cells/pathology , Integrin alpha Chains/genetics , Integrin beta Chains/genetics , Integrin beta Chains/immunology , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
8.
J Cell Sci ; 132(6)2019 03 28.
Article in English | MEDLINE | ID: mdl-30765465

ABSTRACT

Purified vascular endothelial cell (EC) growth factor receptor-2 (VEGFR2) auto-phosphorylates upon VEGF-A occupation in vitro, arguing that VEGR2 confers its mitotic and viability signaling in and of itself. Herein, we show that, in ECs, VEGFR2 function requires concurrent C3a/C5a receptor (C3ar1/C5ar1) and IL-6 receptor (IL-6R)-gp130 co-signaling. C3ar1/C5ar1 or IL-6R blockade totally abolished VEGFR2 auto-phosphorylation, downstream Src, ERK, AKT, mTOR and STAT3 activation, and EC cell cycle entry. VEGF-A augmented production of C3a/C5a/IL-6 and their receptors via a two-step p-Tyk2/p-STAT3 process. Co-immunoprecipitation analyses, confocal microscopy, ligand pulldown and bioluminescence resonance energy transfer assays all indicated that the four receptors are physically interactive. Angiogenesis in murine day 5 retinas and in adult tissues was accelerated when C3ar1/C5ar1 signaling was potentiated, but repressed when it was disabled. Thus, C3ar1/C5ar1 and IL-6R-gp130 joint activation is needed to enable physiological VEGFR2 function.


Subject(s)
Cytokine Receptor gp130/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Complement/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Proliferation , Endothelial Cells/metabolism , Interleukin-6/metabolism , Mice , Neovascularization, Physiologic , Signal Transduction , Vascular Endothelial Growth Factors/metabolism
9.
Am J Transplant ; 19(3): 633-645, 2019 03.
Article in English | MEDLINE | ID: mdl-30106232

ABSTRACT

Costimulatory blockade-induced murine cardiac allograft survival requires intragraft accumulation of CD11b+ Ly6Clo Ly6G- regulatory myeloid cells (Mregs) that expand regulatory T cells (Tregs) and suppress effector T cells (Teffs). We previously showed that C5a receptor (C5aR1) signaling on T cells activates Teffs and inhibits Tregs, but whether and/or how C5aR1 affects Mregs required for transplant survival is unknown. Although BALB/c hearts survived >60 days in anti-CD154 (MR1)-treated or cytotoxic T-lymphocyte associated protein 4 (CTLA4)-Ig-treated wild-type (WT) recipients, they were rejected at ~30 days in MR1-treated or CTLA4-Ig-treated recipients selectively deficient in C5aR1 restricted to myeloid cells (C5ar1fl/fl xLysM-Cre). This accelerated rejection was associated with ~2-fold more donor-reactive T cells and ~40% less expansion of donor-reactive Tregs. Analysis of graft-infiltrating mononuclear cells on posttransplant day 6 revealed fewer Ly6Clo monocytes in C5ar1fl/fl xLysM-Cre recipients. Expression profiling of intragraft Ly6Clo monocytes showed that C5aR1 deficiency downregulated genes related to migration/locomotion without changes in genes associated with suppressive function. Cotransfer of C5ar1fl/fl and C5ar1fl/fl xLysM-Cre myeloid cells into MR1-treated allograft recipients resulted in less accumulation of C5ar1-/- cells within the allografts, and in vitro assays confirmed that Ly6Chi myeloid cells migrate to C5a/C5aR1-initiated signals. Together, our results newly link myeloid cell-expressed C5aR1 to intragraft accumulation of myeloid cells required for prolongation of heart transplant survival induced by costimulatory blockade.


Subject(s)
Abatacept/immunology , CTLA-4 Antigen/immunology , Cell Movement , Graft Survival , Heart Transplantation/methods , Myeloid-Derived Suppressor Cells/immunology , Receptor, Anaphylatoxin C5a/metabolism , Abatacept/chemistry , Abatacept/metabolism , Allografts , Animals , CTLA-4 Antigen/metabolism , Graft Rejection , Heart Diseases/immunology , Heart Diseases/therapy , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Minor Histocompatibility Antigens/immunology , Minor Histocompatibility Antigens/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Receptor, Anaphylatoxin C5a/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
10.
Front Immunol ; 9: 2133, 2018.
Article in English | MEDLINE | ID: mdl-30294325

ABSTRACT

Background and aims: Chronic ethanol exposure results in inflammation in adipose tissue; this response is associated with activation of complement as well as the development of alcohol-related liver disease (ALD). Adipose communicates with other organs, including liver, via the release of soluble mediators, such as adipokines and cytokines, characterized as the "adipose secretome." Here we investigated the role of the anaphaylatoxin receptors C3aR and C5aR1 in the development of adipose tissue inflammation and regulation of the adipose secretome in murine ALD (mALD). Methods: Wild-type C57BL/6 (WT), C3aR-/-, and C5aR1-/- mice were fed Lieber-DeCarli ethanol diet for 25 days (6% v/v, 32% kcal) or isocaloric control diets; indicators of inflammation and injury were assessed in gonadal adipose tissue. The adipose secretome was characterized in isolated adipocytes and stromal vascular cells. Results: Ethanol feeding increased the expression of adipokines, chemokines and leukocyte markers in gonadal adipose tissue from WT mice; C3aR-/- were partially protected while C5aR1-/- mice were completely protected. In contrast, induction of CYP2E1 and accumulation of TUNEL-positive cells in adipose in response to ethanol feeding was independent of genotype. Bone marrow chimeras, generated with WT and C5aR1-/- mice, revealed C5aR1 expression on non-myeloid cells, likely to be adipocytes, contributed to ethanol-induced adipose inflammation. Chronic ethanol feeding regulated both the quantity and distribution of adipokines secreted from adipocytes in a C5aR1-dependent mechanism. In WT mice, chronic ethanol feeding induced a predominant release of pro-inflammatory adipokines from adipocytes, while the adipose secretome from C5aR1-/- mice was characterized by an anti-inflammatory/protective profile. Further, the cargo of adipocyte-derived extracellular vesicles (EVs) was distinct from the soluble secretome; in WT EVs, ethanol increased the abundance of pro-inflammatory mediators while EV cargo from C5aR1-/- adipocytes contained a greater diversity and more robust expression of adipokines. Conclusions: C3aR and C5aR1 are potent regulators of ethanol-induced adipose inflammation in mALD. C5aR1 modulated the impact of chronic ethanol on the content of the adipose secretome, as well as influencing the cargo of an extensive array of adipokines from adipocyte-derived EVs. Taken together, our data demonstrate that C5aR1 contributes to ethanol-mediated changes in the adipose secretome, likely contributing to intra-organ injury in ALD.


Subject(s)
Adipose Tissue/metabolism , Ethanol/adverse effects , Liver Diseases, Alcoholic/immunology , Receptor, Anaphylatoxin C5a/metabolism , Receptors, G-Protein-Coupled/metabolism , Adipocytes , Adipokines/immunology , Adipokines/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adipose Tissue/immunology , Anaphylatoxins/immunology , Anaphylatoxins/metabolism , Animals , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Ethanol/administration & dosage , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Female , Liver/immunology , Liver/metabolism , Liver Diseases, Alcoholic/etiology , Liver Diseases, Alcoholic/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology
11.
J Immunol ; 200(6): 2186-2198, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29436411

ABSTRACT

C5aR2 (C5L2/gp77) is a seven-transmembrane spanning receptor that binds to C5a but lacks motifs essential for G protein coupling and associated signal transduction. C5aR2 is expressed on immune cells, modulates various inflammatory diseases in mice, and has been shown to facilitate murine and human regulatory T cell (TREG) generation in vitro. Whether and how C5aR2 impacts in vivo TREG generation and pathogenic T cell-dependent disease models have not been established. In this article, we show that murine T cells express and upregulate C5aR2 during induced TREG (iTREG) generation and that the absence of T cell-expressed C5aR2 limits in vivo iTREG generation following adoptive transfer of naive CD4+ T cells into Rag1-/- recipients. Using newly generated C5aR2-transgenic mice, we show that overexpression of C5aR2 in naive CD4+ T cells augments in vivo iTREG generation. In a model of TREG-dependent cardiac allograft survival, recipient C5aR2 deficiency accelerates graft rejection associated with lower TREG/effector T cell ratios, whereas overexpression of C5aR2 in immune cells prolongs graft survival associated with an increase in TREG/effector T cell ratios. T cell-expressed C5aR2 modulates TREG induction without altering effector T cell proliferation or cytokine production. Distinct from reported findings in neutrophils and macrophages, TREG-expressed C5aR2 does not interact with ß-arrestin or inhibit ERK1/2 signaling. Rather, cumulative evidence supports the conclusion that C5aR2 limits C5aR1-initiated signals known to inhibit TREG induction. Together, the data expand the role of C5aR2 in adaptive immunity by providing in vivo evidence that T cell-expressed C5aR2 physiologically modulates iTREG generation and iTREG-dependent allograft survival.


Subject(s)
Allografts/immunology , Graft Survival/immunology , Receptor, Anaphylatoxin C5a/immunology , T-Lymphocytes, Regulatory/immunology , Adaptive Immunity/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/physiology , Graft Rejection/immunology , Lymphocyte Activation/immunology , MAP Kinase Signaling System/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophils/immunology , Signal Transduction/immunology
12.
Mol Immunol ; 93: 246-252, 2018 01.
Article in English | MEDLINE | ID: mdl-28886871

ABSTRACT

Decay accelerating factor (DAF or CD55) is a cell associated C3 and C5 convertase regulator originally described in terms of protection of self-cells from systemic complement but now known to modulate adaptive T cell responses. It is expressed on all cell types. We investigated whether nonenzymatic glycation could impair its function and potentially be relevant to complications of diabetes mellitus and other conditions that result in nonenzymatic glycation including cancer, Alzheimer's disease, and aging. Immunoblots of affinity-purified DAF from erythrocytes of patients with diabetes showed pentosidine, glyoxal-AGEs, carboxymethyllysine, and argpyrimidine. HPLC/MS analyses of glucose modified DAF localized the sites of AGE modifications to K125 adjacent to K126, K127 at the junction of CCPs2-3 and spatially near R96, and R100, all identified as being critical for DAF's function. Functional analyses of glucose or ribose treated DAF protein showed profound loss of its regulatory activity. The data argue that de-regulated activation of systemic complement and de-regulated activation of T cells and leukocytes could result from non-enzymatic glycation of DAF.


Subject(s)
CD55 Antigens/chemistry , Diabetes Mellitus/blood , Glycation End Products, Advanced/chemistry , Amino Acids/chemistry , Arginine/analogs & derivatives , Arginine/analysis , CD55 Antigens/blood , CD55 Antigens/drug effects , Catalytic Domain/drug effects , Complement Activation , Erythrocytes/chemistry , Glucose/pharmacology , Glycation End Products, Advanced/blood , Humans , Lymphocyte Activation , Lysine/analogs & derivatives , Lysine/analysis , Models, Molecular , Ornithine/analogs & derivatives , Ornithine/analysis , Protein Conformation , Pyrimidines/analysis , Ribose/pharmacology
13.
J Immunol ; 199(1): 278-291, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28539427

ABSTRACT

Induction of proinflammatory T cell immunity is augmented by innate dendritic cell (DC) maturation commonly initiated by TLR signaling. We demonstrate that ligation of TLR3, TLR4, and TLR9 induces murine DC production of complement components and local production of the anaphylatoxin C5a. In vitro, ex vivo, and in vivo analyses show that TLR-induced DC maturation, as assessed by surface phenotype, expression profiling by gene array, and functional ability to stimulate T cell responses, requires autocrine C3a receptor and C5a receptor (C3ar1/C5ar1) signaling. Studies using bone marrow chimeric animals and Foxp3-GFP/ERT2-Cre/dTomato fate-mapping mice show that TLR-initiated DC autocrine C3ar1/C5ar1 signaling causes expansion of effector T cells and instability of regulatory T cells and contributes to T cell-dependent transplant rejection. Together, our data position immune cell-derived complement production and autocrine/paracrine C3ar1/C5ar1 signaling as crucial intermediary processes that link TLR stimulation to DC maturation and the subsequent development of effector T cell responses.


Subject(s)
Complement C5a/immunology , Dendritic Cells/immunology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism , Animals , Cell Differentiation , Cells, Cultured , Complement C3a/immunology , Complement C3a/metabolism , Complement C5a/biosynthesis , Complement C5a/metabolism , Dendritic Cells/physiology , Mice , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/physiology , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 9/immunology
14.
Mol Immunol ; 75: 122-32, 2016 07.
Article in English | MEDLINE | ID: mdl-27280845

ABSTRACT

BACKGROUND: Complement is implicated in the development of alcoholic liver disease. C3 and C5 contribute to ethanol-induced liver injury; however, the role of C5a receptor (C5aR) on myeloid and non-myeloid cells to progression of injury is not known. METHODS: C57BL/6 (WT), global C5aR-/-, myeloid-specific C5aR-/-, and non-myeloid-specific C5aR-/- mice were fed a Lieber-DeCarli diet (32%kcal EtOH) for 25 days. Cultured hepatocytes were challenged with ethanol, TNFα, and C5a. RESULTS: Chronic ethanol feeding increased expression of pro-inflammatory mediators in livers of WT mice; this response was completely blunted in C5aR-/- mice. However, C5aR-/- mice were not protected from other measures of hepatocellular damage, including ethanol-induced increases in hepatic triglycerides, plasma alanine aminotransferase and hepatocyte apoptosis. CYP2E1 and 4-hydroxynonenal protein adducts were induced in WT and C5aR-/- mice. Myeloid-specific C5aR-/- mice were protected from ethanol-induced increases in hepatic TNFα, whereas non-myeloid-specific C5aR-/- displayed increased hepatocyte apoptosis and inflammation after chronic ethanol feeding. In cultured hepatocytes, cytotoxicity induced by challenge with ethanol and TNFα was completely eliminated by treatment with C5a in cells from WT, but not C5aR-/- mice. Further, treatment with C5a enhanced activation of pro-survival signal AKT in hepatocytes challenged with ethanol and TNFα. CONCLUSION: Taken together, these data reveal a differential role for C5aR during ethanol-induced liver inflammation and injury, with C5aR on myeloid cells contributing to ethanol-induced inflammatory cytokine expression, while non-myeloid C5aR protects hepatocytes from death after chronic ethanol feeding.


Subject(s)
Hepatocytes/immunology , Liver Diseases, Alcoholic/immunology , Myeloid Cells/immunology , Receptor, Anaphylatoxin C5a/immunology , Animals , Blotting, Western , Disease Models, Animal , Ethanol/toxicity , Female , Immunohistochemistry , Liver Diseases, Alcoholic/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction
15.
J Immunol ; 190(7): 3740-8, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23447688

ABSTRACT

Adhesion G protein-coupled receptors (aGPCRs) are two-subunit molecules, consisting of an adhesive extracellular α subunit that couples noncovalently to a seven-transmembrane ß subunit. The cooperation between the two subunits and the effect of endogenous ligands on the functioning of aGPCRs is poorly understood. In this study, we investigated the interaction between the pan-leukocyte aGPCR CD97 and its ligand CD55. We found that leukocytes from CD55-deficient mice express significantly increased levels of cell surface CD97 that normalized after transfer into wild-type mice because of contact with CD55 on both leukocytes and stromal cells. Downregulation of both CD97 subunits occurred within minutes after first contact with CD55 in vivo, which correlated with an increase in plasma levels of soluble CD97. In vitro, downregulation of CD97 on CD55-deficient leukocytes cocultured with wild-type blood cells was strictly dependent on shear stress. In vivo, CD55-mediated downregulation of CD97 required an intact circulation and was not observed on cells that lack contact with the blood stream, such as microglia. Notably, de novo ligation of CD97 did not activate signaling molecules constitutively engaged by CD97 in cancer cells, such as ERK and protein kinase B/Akt. We conclude that CD55 downregulates CD97 surface expression on circulating leukocytes by a process that requires physical forces, but based on current evidence does not induce receptor signaling. This regulation can restrict CD97-CD55-mediated cell adhesion to tissue sites.


Subject(s)
CD55 Antigens/metabolism , Leukocytes/metabolism , Membrane Glycoproteins/metabolism , Animals , CD55 Antigens/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Protein Binding , Protein Subunits/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled , Signal Transduction , Stromal Cells/metabolism
16.
Nat Immunol ; 14(2): 162-71, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23263555

ABSTRACT

Signaling through the G protein-coupled receptors for the complement fragments C3a and C5a (C3aR and C5aR, respectively) by dendritic cells and CD4(+) cells provides costimulatory and survival signals to effector T cells. Here we found that when signals from C3aR and C5aR were not transduced into CD4(+) cells, signaling via the kinases PI(3)Kγ, Akt and mTOR ceased, activation of the kinase PKA increased, autoinductive signaling by transforming growth factor-ß1 (TGF-ß1) initiated and CD4(+) T cells became Foxp3(+) induced regulatory T cells (iT(reg) cells). Endogenous TGF-ß1 suppressed signaling through C3aR and C5aR by preventing the production of C3a and C5a and upregulating C5L2, an alternative receptor for C5a. The absence of signaling via C3aR and C5aR resulted in lower expression of costimulatory molecules and interleukin 6 (IL-6) and more production of IL-10. The resulting iT(reg) cells exerted robust suppression, had enhanced stability and suppressed ongoing autoimmune disease. Antagonism of C3aR and C5aR can also induce functional human iT(reg) cells.


Subject(s)
Forkhead Transcription Factors/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Complement/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Communication/immunology , Cell Differentiation , Class Ib Phosphatidylinositol 3-Kinase/immunology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Complement C3a/immunology , Complement C3a/metabolism , Complement C5a/immunology , Complement C5a/metabolism , Cyclic AMP-Dependent Protein Kinases/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptor Cross-Talk/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Receptors, Complement/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta1/immunology
17.
J Clin Invest ; 122(6): 2234-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22585573

ABSTRACT

Acute graft-versus-host disease (GvHD) is a serious complication of allogeneic hematopoietic cell transplantation (allo-HCT) that results from donor allogeneic T cell attack on host tissues. Based on previous work implicating immune cell-derived C3a and C5a as regulators of T cell immunity, we examined the effects of locally produced C3a and C5a on murine T cell-mediated GvHD. We found that total body irradiation, a conditioning regimen required to permit engraftment of allo-HCT, caused upregulation and activation of alternative pathway complement components by recipient APCs. Allo-HCT with decay accelerating factor-null (Daf1(-/-)) host BM and Daf1(-/-) donor lymphocytes led to exacerbated GvHD outcome and resulted in splenic and organ-infiltrating T cell expansion. T cells deficient in C3a receptor (C3aR) and/or C5a receptor (C5aR) responded weakly in allogeneic hosts and exhibited limited ability to induce GvHD. Using a clinically relevant treatment strategy, we showed that pharmacological C5aR blockade reduced GvHD morbidity. Our data mechanistically link APC-derived complement to T cell-mediated GvHD and support complement inhibition as a therapeutic strategy for GvHD in humans.


Subject(s)
Antigen-Presenting Cells/immunology , Complement C3a/immunology , Complement C5a/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , T-Lymphocytes/immunology , Transplantation Conditioning , Acute Disease , Animals , Antigen-Presenting Cells/pathology , Complement C3a/genetics , Complement C5a/genetics , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Humans , Mice , Mice, Knockout , Receptors, Complement/genetics , Receptors, Complement/immunology , T-Lymphocytes/pathology , Transplantation, Homologous , Whole-Body Irradiation
18.
PLoS One ; 6(10): e24431, 2011.
Article in English | MEDLINE | ID: mdl-21984892

ABSTRACT

BACKGROUND: In addition to its complement-regulating activity, CD55 is a ligand of the adhesion class G protein-coupled receptor CD97; however, the relevance of this interaction has remained elusive. We previously showed that mice lacking a functional CD97 gene have increased numbers of granulocytes. METHODOLOGY/RESULTS: Here, we demonstrate that CD55-deficient mice display a comparable phenotype with about two-fold more circulating granulocytes in the blood stream, the marginated pool, and the spleen. This granulocytosis was independent of increased complement activity. Augmented numbers of Gr-1-positive cells in cell cycle in the bone marrow indicated a higher granulopoietic activity in mice lacking either CD55 or CD97. Concomitant with the increase in blood granulocyte numbers, Cd55⁻/⁻ mice challenged with the respiratory pathogen Streptococcus pneumoniae developed less bacteremia and died later after infection. CONCLUSIONS: Collectively, these data suggest that complement-independent interaction of CD55 with CD97 is functionally relevant and involved in granulocyte homeostasis and host defense.


Subject(s)
CD55 Antigens/metabolism , Granulocytes/immunology , Homeostasis/immunology , Host-Pathogen Interactions/immunology , Streptococcus pneumoniae/immunology , Animals , Cell Movement/immunology , Complement System Proteins/immunology , Disease Resistance/immunology , Granulocytes/cytology , Leukocyte Count , Membrane Glycoproteins/metabolism , Mice , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/microbiology , Receptors, G-Protein-Coupled
19.
Am J Pathol ; 179(2): 766-74, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21704012

ABSTRACT

Although induction of CD8 T-cell responses to transplants requires CD4-cell help, how this help is transmitted remains incompletely characterized. In vitro, cognate interactions between CD4 T cells and dendritic cells (DCs) induce C3a and C5a production. CD8(+) T cells lacking C3a receptor (C3aR) and C5a receptor (C5aR) proliferate weakly to allogeneic DCs despite CD4 help, indicating that CD4-cell help is mediated, in part, through DC-derived C3a/C5a acting on CD8(+) T cell-expressed C3aR/C5aR. In support of this concept, augmenting DC C5a/C3a production bypasses the requirement for CD4- and CD40-dependent help to wild-type CD8(+) T cells. CD4-deficient recipients of allogeneic heart transplants prime weak CD8 responses and do not acutely reject their grafts. In contrast, CD4-deficient chimeric mice possessing decay accelerating factor deficient (Daf1(-/-)) bone marrow, in which DC C3a/C5a production is potentiated, acutely reject transplants through a CD8 cell-dependent mechanism. Furthermore, hearts transplanted into CD40(-/-) mice prime weak CD8-cell responses and survive indefinitely, but hearts transplanted into Daf1(-/-)CD40(-/-) recipients undergo CD8 cell-dependent rejection. Together, the data indicate that heightened production and activation of immune cell-derived complement bypasses the need for CD40/CD154 interactions and implicate antigen-presenting cell-produced C5a and C3a as molecular bridges linking CD4 help to CD8(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Complement System Proteins/metabolism , Animals , Bone Marrow Cells/cytology , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Cells, Cultured , Complement C3a/metabolism , Complement C5a/metabolism , Crosses, Genetic , Dendritic Cells/cytology , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Graft Rejection , Heart Transplantation/methods , Immunohistochemistry/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction/methods
20.
J Virol ; 84(24): 12665-74, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20881044

ABSTRACT

Echovirus 7 (EV7) belongs to the Enterovirus genus within the family Picornaviridae. Many picornaviruses use IgG-like receptors that bind in the viral canyon and are required to initiate viral uncoating during infection. However, in addition, some of the enteroviruses use an alternative or additional receptor that binds outside the canyon. Decay-accelerating factor (DAF) has been identified as a cellular receptor for EV7. The crystal structure of EV7 has been determined to 3.1-Å resolution and used to interpret the 7.2-Å-resolution cryo-electron microscopy reconstruction of EV7 complexed with DAF. Each DAF binding site on EV7 is near a 2-fold icosahedral symmetry axis, which differs from the binding site of DAF on the surface of coxsackievirus B3, indicating that there are independent evolutionary processes by which DAF was selected as a picornavirus accessory receptor. This suggests that there is an advantage for these viruses to recognize DAF during the initial process of infection.


Subject(s)
CD55 Antigens/chemistry , CD55 Antigens/metabolism , Echovirus Infections/metabolism , Enterovirus B, Human/chemistry , Enterovirus B, Human/metabolism , Amino Acid Sequence , Binding Sites , Cryoelectron Microscopy , Crystallization , Crystallography, X-Ray , Echovirus Infections/virology , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...