Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
Add more filters

Publication year range
1.
Nature ; 588(7836): 146-150, 2020 12.
Article in English | MEDLINE | ID: mdl-32726800

ABSTRACT

Coronavirus disease 2019 (COVID-19) is a disease caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has resulted in a pandemic1. The C5a complement factor and its receptor C5aR1 (also known as CD88) have a key role in the initiation and maintenance of several inflammatory responses by recruiting and activating neutrophils and monocytes1. Here we provide a longitudinal analysis of immune responses, including phenotypic analyses of immune cells and assessments of the soluble factors that are present in the blood and bronchoalveolar lavage fluid of patients at various stages of COVID-19 severity, including those who were paucisymptomatic or had pneumonia or acute respiratory distress syndrome. The levels of soluble C5a were increased in proportion to the severity of COVID-19 and high expression levels of C5aR1 receptors were found in blood and pulmonary myeloid cells, which supports a role for the C5a-C5aR1 axis in the pathophysiology of acute respiratory distress syndrome. Anti-C5aR1 therapeutic monoclonal antibodies prevented the C5a-mediated recruitment and activation of human myeloid cells, and inhibited acute lung injury in human C5aR1 knock-in mice. These results suggest that blockade of the C5a-C5aR1 axis could be used to limit the infiltration of myeloid cells in damaged organs and prevent the excessive lung inflammation and endothelialitis that are associated with acute respiratory distress syndrome in patients with COVID-19.


Subject(s)
COVID-19/complications , COVID-19/immunology , Complement C5a/immunology , Inflammation/complications , Inflammation/immunology , Receptor, Anaphylatoxin C5a/immunology , Acute Lung Injury/drug therapy , Acute Lung Injury/immunology , Acute Lung Injury/prevention & control , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , CD11b Antigen/immunology , CD11b Antigen/metabolism , COVID-19/blood , COVID-19/pathology , Complement C5a/antagonists & inhibitors , Complement C5a/biosynthesis , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/prevention & control , Disease Models, Animal , Female , Humans , Inflammation/drug therapy , Inflammation/pathology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/pathology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/blood , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/prevention & control , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity
2.
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
3.
Trends Immunol ; 41(11): 965-967, 2020 11.
Article in English | MEDLINE | ID: mdl-33023856

ABSTRACT

The current pandemic of coronavirus disease (COVID-19) caused by SARS-CoV-2 is a significant global health challenge. A recent study by Carvelli and colleagues now demonstrates the involvement of complement C5a and its receptor C5aR1 in disease progression and suggests that blockade of the C5a-C5aR1 axis may represent a potential therapeutic strategy against COVID-19.


Subject(s)
Betacoronavirus/immunology , Complement C5a/immunology , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Receptor, Anaphylatoxin C5a/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Betacoronavirus/drug effects , Betacoronavirus/physiology , COVID-19 , Complement C5a/metabolism , Coronavirus Infections/therapy , Coronavirus Infections/virology , Disease Models, Animal , Humans , Pandemics , Pneumonia, Viral/therapy , Pneumonia, Viral/virology , Receptor, Anaphylatoxin C5a/metabolism , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , SARS-CoV-2 , Signal Transduction/drug effects , Signal Transduction/immunology
4.
Nature ; 543(7643): 108-112, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28225753

ABSTRACT

Gaucher disease is caused by mutations in GBA1, which encodes the lysosomal enzyme glucocerebrosidase (GCase). GBA1 mutations drive extensive accumulation of glucosylceramide (GC) in multiple innate and adaptive immune cells in the spleen, liver, lung and bone marrow, often leading to chronic inflammation. The mechanisms that connect excess GC to tissue inflammation remain unknown. Here we show that activation of complement C5a and C5a receptor 1 (C5aR1) controls GC accumulation and the inflammatory response in experimental and clinical Gaucher disease. Marked local and systemic complement activation occurred in GCase-deficient mice or after pharmacological inhibition of GCase and was associated with GC storage, tissue inflammation and proinflammatory cytokine production. Whereas all GCase-inhibited mice died within 4-5 weeks, mice deficient in both GCase and C5aR1, and wild-type mice in which GCase and C5aR were pharmacologically inhibited, were protected from these adverse effects and consequently survived. In mice and humans, GCase deficiency was associated with strong formation of complement-activating GC-specific IgG autoantibodies, leading to complement activation and C5a generation. Subsequent C5aR1 activation controlled UDP-glucose ceramide glucosyltransferase production, thereby tipping the balance between GC formation and degradation. Thus, extensive GC storage induces complement-activating IgG autoantibodies that drive a pathway of C5a generation and C5aR1 activation that fuels a cycle of cellular GC accumulation, innate and adaptive immune cell recruitment and activation in Gaucher disease. As enzyme replacement and substrate reduction therapies are expensive and still associated with inflammation, increased risk of cancer and Parkinson disease, targeting C5aR1 may serve as a treatment option for patients with Gaucher disease and, possibly, other lysosomal storage diseases.


Subject(s)
Complement System Proteins/immunology , Gaucher Disease/immunology , Gaucher Disease/pathology , Glucosylceramides/immunology , Glucosylceramides/metabolism , Inflammation/immunology , Inflammation/pathology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Autoantibodies/immunology , Complement Activation , Complement C5a/biosynthesis , Complement C5a/immunology , Complement System Proteins/biosynthesis , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Female , Gaucher Disease/metabolism , Gaucher Disease/prevention & control , Glucosylceramidase/antagonists & inhibitors , Glucosylceramidase/deficiency , Glucosylceramidase/genetics , Glucosyltransferases/biosynthesis , Glucosyltransferases/metabolism , Humans , Immunoglobulin G/immunology , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology
5.
J Allergy Clin Immunol ; 147(6): 2083-2097.e6, 2021 06.
Article in English | MEDLINE | ID: mdl-33852936

ABSTRACT

BACKGROUND: Excessive inflammation triggered by a hitherto undescribed mechanism is a hallmark of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and is associated with enhanced pathogenicity and mortality. OBJECTIVE: Complement hyperactivation promotes lung injury and was observed in patients suffering from Middle East respiratory syndrome-related coronavirus, SARS-CoV-1, and SARS-CoV-2 infections. Therefore, we investigated the very first interactions of primary human airway epithelial cells on exposure to SARS-CoV-2 in terms of complement component 3 (C3)-mediated effects. METHODS: For this, we used highly differentiated primary human 3-dimensional tissue models infected with SARS-CoV-2 patient isolates. On infection, viral load, viral infectivity, intracellular complement activation, inflammatory mechanisms, and tissue destruction were analyzed by real-time RT-PCR, high content screening, plaque assays, luminex analyses, and transepithelial electrical resistance measurements. RESULTS: Here, we show that primary normal human bronchial and small airway epithelial cells respond to SARS-CoV-2 infection by an inflated local C3 mobilization. SARS-CoV-2 infection resulted in exaggerated intracellular complement activation and destruction of the epithelial integrity in monolayer cultures of primary human airway cells and highly differentiated, pseudostratified, mucus-producing, ciliated respiratory tissue models. SARS-CoV-2-infected 3-dimensional cultures secreted significantly higher levels of C3a and the proinflammatory cytokines IL-6, monocyte chemoattractant protein 1, IL-1α, and RANTES. CONCLUSIONS: Crucially, we illustrate here for the first time that targeting the anaphylotoxin receptors C3a receptor and C5a receptor in nonimmune respiratory cells can prevent intrinsic lung inflammation and tissue damage. This opens up the exciting possibility in the treatment of COVID-19.


Subject(s)
Bronchi/immunology , COVID-19/immunology , Complement Activation , Epithelial Cells/immunology , Receptor, Anaphylatoxin C5a/immunology , Respiratory Mucosa/immunology , SARS-CoV-2/immunology , Bronchi/pathology , Bronchi/virology , COVID-19/pathology , COVID-19/virology , Cell Line , Complement C3/immunology , Cytokines/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , Humans , Inflammation/immunology , Inflammation/pathology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology
6.
FASEB J ; 34(6): 7540-7560, 2020 06.
Article in English | MEDLINE | ID: mdl-32301538

ABSTRACT

The vascular endothelium has been discovered in the past several years to be important in shaping the cellular immune response. During the immune response the vascular endothelium is constantly perturbed by biologically potent molecules, including the complement activation peptides, C3a and C5a. Despite the importance of C3a and C5a in inflammation and immunity, their role in modulating lymphocyte function via activation of vascular endothelial cells is unknown. Accordingly, we investigated the regulated expression of the C3a and C5a receptors (complement anaphylatoxin C3a receptor [C3aR] and complement anaphylatoxin C5a receptor 1 [C5aR1]) on human umbilical vascular endothelial cells (HUVECs) and examined how C3a or C5a activation of HUVECs affects the activation and polarization of lymphatic cells. Our findings demonstrated that C3a and C5a increase C3aR and C5aR1 expression by HUVECs as well as directing their cellular transmigration and spreading through transwell filters. Moreover, C3a- or C5a-stimulated endothelial cells: (1) caused activation of B-lymphoblasts with significant increase in Fas Ligand (CD95L) (FasL), CD69, and IL-R1 expression, and (2) skewed T-lymphoblast cells toward a Th1 subtype, (CD4+ /CCR5+ ) that correlated with significant increase of IFN-γ. Collectively, these data indicate that C3a and C5a signaling is important in the activation and polarization of lymphocytes as they traffic through the vascular endothelium during the immune response.


Subject(s)
Anaphylatoxins/immunology , B-Lymphocytes/immunology , Complement C3a/immunology , Complement C5a/immunology , Peptides/immunology , T-Lymphocytes/immunology , Cells, Cultured , Complement Activation/immunology , Endothelium, Vascular/immunology , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Complement/immunology , Signal Transduction/immunology
7.
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
8.
J Immunol ; 202(10): 2982-2990, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30952817

ABSTRACT

Biliverdin reductase (BVR)-A is a pleotropic enzyme converting biliverdin to bilirubin and a signaling molecule that has cytoprotective and immunomodulatory effects. We recently showed that biliverdin inhibits the expression of complement activation fragment 5a receptor one (C5aR1) in RAW 264.7 macrophages. In this study, we investigated the role of BVR-A in determining macrophage inflammatory phenotype and function via regulation of C5aR1. We assessed expression of C5aR1, M1-like macrophage markers, including chemokines (RANTES, IP-10), as well as chemotaxis in response to LPS and C5a in bone marrow-derived macrophages from BVR fl/fl and LysM-Cre:BVR fl / fl mice (conditional deletion of BVR-A in myeloid cells). In response to LPS, macrophages isolated from LysM-Cre:BVR fl/fl showed significantly elevated levels of C5aR1 as well as chemokines (RANTES, IP10) but not proinflammatory markers, such as iNOS and TNF. An increase in C5aR1 expression was also observed in peritoneal macrophages and several tissues from LysM-Cre:BVR fl/fl mice in a model of endotoxemia. In addition, knockdown of BVR-A resulted in enhanced macrophage chemotaxis toward C5a. Part of the effects of BVR-A deletion on chemotaxis and RANTES expression were blocked in the presence of a C5aR1 neutralizing Ab, confirming the role of C5a-C5aR1 signaling in mediating the effects of BVR. In summary, BVR-A plays an important role in regulating macrophage chemotaxis in response to C5a via modulation of C5aR1 expression. In addition, macrophages lacking BVR-A are characterized by the expression of M1 polarization-associated chemokines, the levels of which depend in part on C5aR1 signaling.


Subject(s)
Chemokines/immunology , Chemotaxis/immunology , Complement C5a/immunology , Macrophages/immunology , Oxidoreductases Acting on CH-CH Group Donors/immunology , Receptor, Anaphylatoxin C5a/immunology , Signal Transduction/immunology , Animals , Chemokines/genetics , Chemotaxis/genetics , Complement C5a/genetics , Gene Deletion , Macrophages/cytology , Mice , Mice, Transgenic , Oxidoreductases Acting on CH-CH Group Donors/genetics , Receptor, Anaphylatoxin C5a/genetics , Signal Transduction/genetics
9.
Nat Immunol ; 9(11): 1225-35, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18820683

ABSTRACT

The involvement of complement-activation products in promoting tumor growth has not yet been recognized. Here we show that the generation of complement C5a in a tumor microenvironment enhanced tumor growth by suppressing the antitumor CD8(+) T cell-mediated response. This suppression was associated with the recruitment of myeloid-derived suppressor cells into tumors and augmentation of their T cell-directed suppressive abilities. Amplification of the suppressive capacity of myeloid-derived suppressor cells by C5a occurred through regulation of the production of reactive oxygen and nitrogen species. Pharmacological blockade of the C5a receptor considerably impaired tumor growth to a degree similar to the effect produced by the anticancer drug paclitaxel. Thus, our study demonstrates a therapeutic function for complement inhibition in the treatment of cancer.


Subject(s)
Complement C5a/immunology , Immunosuppression Therapy , Neoplasms/immunology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/immunology , Animals , Complement Activation , Complement C3-C5 Convertases/genetics , Complement C5a/antagonists & inhibitors , Complement C5a/pharmacology , Down-Regulation , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/therapy , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
10.
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
11.
Kidney Int ; 96(1): 117-128, 2019 07.
Article in English | MEDLINE | ID: mdl-31029505

ABSTRACT

C5a is a potent proinflammatory agonist that mediates renal ischemia reperfusion (IR) injury, but the potential for modulating chronic post-ischemic fibrosis and use of therapeutic antagonist are undefined. Here we determine whether C5a receptor 1 (C5aR1) signaling is essential to the development of post-ischemic fibrosis and if it is a valid target for therapeutic blockade with soluble receptor antagonist. C5aR1 is required for the development of renal tubulointerstitial fibrosis in a murine model of renal ischemia/reperfusion injury. Deficiency of C5aR1 protected mice from the development of the fibrosis. This protection was associated with attenuated deposition of extracellular matrix components (fibronectin, collagen I), reduced cellular infiltrates (CD45, F4/80), and gene expression of proinflammatory and profibrogenic mediators in the kidney. In an in vitro model of hypoxia/reoxygenation, C5a stimulation caused renal fibroblast proliferation and activation, and upregulated gene expression of interleukin-1α (IL-1α), IL-6 and transforming growth factor-α (TGF-α) in renal tubular epithelial cells and monocytes/macrophages. Administration of a C5aR1 antagonist (PMX53) significantly reduced renal injury and tubulointerstitial fibrosis. Thus, our results demonstrate a pathogenic role for C5aR1 in the progression of tubulointerstitial fibrosis following renal IR injury and support that C5aR1-mediated local inflammatory responses to hypoxic renal injury contribute to tubulointerstitial fibrosis through several cellular pathways, namely, promoting tubule injury, interstitial fibroblast proliferation and epithelial-to-mesenchymal transition of renal tubular epithelial cells. Our results also suggest the C5a-C5aR1 interaction is a therapeutic target for chronic post-ischemic fibrosis.


Subject(s)
Kidney Tubules/pathology , Nephritis, Interstitial/immunology , Receptor, Anaphylatoxin C5a/metabolism , Reperfusion Injury/complications , Signal Transduction/immunology , Animals , Cell Proliferation , Complement C5a/metabolism , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/immunology , Fibroblasts , Fibrosis , Humans , Kidney Tubules/cytology , Kidney Tubules/immunology , Male , Mice , Mice, Knockout , Nephritis, Interstitial/pathology , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/immunology , Reperfusion Injury/immunology , Up-Regulation
12.
Kidney Int ; 96(1): 67-79, 2019 07.
Article in English | MEDLINE | ID: mdl-30910380

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a form of thrombotic microangiopathy (TMA) caused by dysregulated complement activation. Clinically, aHUS is effectively treated by an anti-C5 monoclonal antibody (mAb) but whether the disease is mediated by the C5a receptor (C5aR) or C5b-9 pathway, or both, is unknown. Here we address this in a factor H mutant mouse (FHR/R) which developed complement-mediated TMA as well as macrovascular thrombosis caused by an aHUS-related factor H point mutation (mouse W1206R, corresponding to human W1183R). C5 deficiency and anti-C5 mAb treatment blocked all disease manifestations in FHR/R mice. C5aR1 gene deficiency prevented macrovascular thrombosis in various organs but did not improve survival or reduce renal TMA. Conversely, C6 or C9 deficiency significantly improved survival and markedly diminished renal TMA but did not prevent macrovascular thrombosis. Interestingly, as they aged both FHR/R C6-/- and FHR/R C9-/- mice developed glomerular disease reminiscent of C3 glomerulonephritis. Thus, C5aR and C5b-9 pathways drove different aspects of disease in FHR/R mice with the C5aR pathway being responsible for macrovascular thrombosis and chronic inflammatory injury while the C5b-9 pathway caused renal TMA. Our data provide new understanding of the pathogenesis of complement-mediated TMA and macrovascular thrombosis in FHR/R mice and suggest that C5 blockade is more effective for the treatment of aHUS than selectively targeting the C5aR or C5b-9 pathway alone.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Complement Factor H/genetics , Complement Membrane Attack Complex/immunology , Kidney Glomerulus/pathology , Receptor, Anaphylatoxin C5a/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/pathology , Complement Activation/drug effects , Complement Activation/genetics , Complement Activation/immunology , Complement C6/genetics , Complement C6/immunology , Complement C6/metabolism , Complement Factor H/immunology , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/metabolism , Disease Models, Animal , Female , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/ultrastructure , Male , Mice , Mice, Transgenic , Microscopy, Electron , Point Mutation , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism
13.
Am J Hematol ; 94(3): 327-337, 2019 03.
Article in English | MEDLINE | ID: mdl-30569594

ABSTRACT

Innate immune complement activation may contribute to sickle cell disease (SCD) pathogenesis. Ischemia-reperfusion physiology is a key component of the inflammatory and vaso-occlusive milieu in SCD and is associated with complement activation. C5a is an anaphylatoxin, a potent pro-inflammatory mediator that can activate leukocytes, platelets, and endothelial cells, all of which play a role in vaso-occlusion. We hypothesize that hypoxia-reoxygenation (H/R) in SCD mice activates complement, promoting inflammation and vaso-occlusion. At baseline and after H/R, sickle Townes-SS mice had increased C3 activation fragments and C5b-9 deposition in kidneys, livers and lungs and alternative pathway Bb fragments in plasma compared to control AA-mice. Activated complement promoted vaso-occlusion (microvascular stasis) in SS-mice; infusion of zymosan-activated, but not heat-inactivated serum, induced substantial vaso-occlusion in the skin venules of SS-mice. Infusion of recombinant C5a induced stasis in SS, but not AA-mice that was blocked by anti-C5a receptor (C5aR) IgG. C5a-mediated stasis was accompanied by inflammatory responses in SS-mice including NF-κB activation and increased expression of TLR4 and adhesion molecules VCAM-1, ICAM-1, and E-selectin in the liver. Anti-C5aR IgG blocked these inflammatory responses. Also, C5a rapidly up-regulated Weibel-Palade body P-selectin and von Willebrand factor on the surface of human umbilical vein endothelial cells in vitro and on vascular endothelium in vivo. In SS-mice, a blocking antibody to P-selectin inhibited C5a-induced stasis. Similarly, an antibody to C5 that blocks murine C5 cleavage or an antibody that blocks C5aR inhibited H/R-induced stasis in SS-mice. These results suggest that inhibition of C5a may be beneficial in SCD.


Subject(s)
Anemia, Sickle Cell/immunology , Antibodies, Neutralizing/pharmacology , Cerebrovascular Disorders/immunology , Complement C3/immunology , Complement C5a/immunology , Receptor, Anaphylatoxin C5a/immunology , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/pathology , Animals , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/pathology , Complement C3/genetics , Complement C5a/antagonists & inhibitors , Complement C5a/genetics , Complement Membrane Attack Complex/genetics , Complement Membrane Attack Complex/immunology , Disease Models, Animal , E-Selectin/genetics , E-Selectin/immunology , Gene Expression Regulation , Humans , Immunity, Innate , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Kidney/blood supply , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Liver/blood supply , Liver/drug effects , Liver/immunology , Liver/pathology , Lung/blood supply , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , P-Selectin/antagonists & inhibitors , P-Selectin/genetics , P-Selectin/immunology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/genetics , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
14.
J Immunol ; 198(7): 2989-2999, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28228558

ABSTRACT

Relatively little is known about factors that initiate immunosuppression in tumors and act at the interface between tumor cells and host cells. In this article, we report novel immunosuppressive properties of the ribosomal protein S19 (RPS19), which is upregulated in human breast and ovarian cancer cells and released from apoptotic tumor cells, whereupon it interacts with the complement C5a receptor 1 expressed on tumor infiltrating myeloid-derived suppressor cells. This interaction promotes tumor growth by facilitating recruitment of these cells to tumors. RPS19 also induces the production of immunosuppressive cytokines, including TGF-ß, by myeloid-derived suppressor cells in tumor-draining lymph nodes, leading to T cell responses skewed toward Th2 phenotypes. RPS19 promotes generation of regulatory T cells while reducing infiltration of CD8+ T cells into tumors. Reducing RPS19 in tumor cells or blocking the C5a receptor 1-RPS19 interaction decreases RPS19-mediated immunosuppression, impairs tumor growth, and delays the development of tumors in a transgenic model of breast cancer. This work provides initial preclinical evidence for targeting RPS19 for anticancer therapy enhancing antitumor T cell responses.


Subject(s)
Myeloid-Derived Suppressor Cells/immunology , Neoplasms, Experimental/immunology , Receptor, Anaphylatoxin C5a/immunology , Ribosomal Proteins/immunology , Animals , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Humans , Immunoprecipitation , Mice , T-Lymphocytes/immunology
15.
J Immunol ; 199(9): 3234-3248, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28864475

ABSTRACT

The biological significance of C5a receptor [(C5aR)2/C5L2], a seven-transmembrane receptor binding C5a and C5adesArg, remains ill-defined. Specific ligation of C5aR2 inhibits C5a-induced ERK1/2 activation, strengthening the view that C5aR2 regulates C5aR1-mediated effector functions. Although C5aR2 and C5aR1 are often coexpressed, a detailed picture of C5aR2 expression in murine cells and tissues is still lacking. To close this gap, we generated a floxed tandem dye (td)Tomato-C5aR2 knock-in mouse that we used to track C5aR2 expression in tissue-residing and circulating immune cells. We found the strongest C5aR2 expression in the brain, bone marrow, and airways. All myeloid-derived cells expressed C5aR2, although with different intensities. C5aR2 expression in blood and tissue neutrophils was strong and homogeneous. Specific ligation of C5aR2 in neutrophils from tdTomato-C5aR2 mice blocked C5a-driven ERK1/2 phosphorylation, demonstrating functionality of C5aR2 in the reporter mice. In contrast to neutrophils, we found tissue-specific differences in C5aR2 expression in eosinophils, macrophages, and dendritic cell subsets. Naive and activated T cells stained negative for C5aR2, whereas B cells from different tissues homogeneously expressed C5aR2. Also, NK cell subsets in blood and spleen strongly expressed C5aR2. Activation of C5aR2 in NK cells suppressed IL-12/IL-18-induced IFN-γ production. Intratracheal IL-33 challenge resulted in decreased C5aR2 expression in pulmonary eosinophils and monocyte-derived dendritic cells. In summary, we provide a detailed map of murine C5aR2 immune cell expression in different tissues under steady-state conditions and upon pulmonary inflammation. The C5aR2 knock-in mouse will help to reliably track and conditionally delete C5aR2 expression in experimental models of inflammation.


Subject(s)
Gene Expression Regulation/immunology , Leukocytes/immunology , Pneumonia/immunology , Receptor, Anaphylatoxin C5a/immunology , Animals , Gene Knock-In Techniques , Genes, Reporter/immunology , Leukocytes/pathology , Mice , Mice, Transgenic , Organ Specificity/genetics , Organ Specificity/immunology , Pneumonia/genetics , Pneumonia/pathology , Receptor, Anaphylatoxin C5a/genetics
16.
Am J Respir Crit Care Med ; 197(9): 1164-1176, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29327939

ABSTRACT

RATIONALE: C5aR1 (CD88), a receptor for complement anaphylatoxin C5a, is a potent immune mediator. Its impact on malignant growth and dissemination of non-small cell lung cancer cells is poorly understood. OBJECTIVES: To investigate the contribution of the C5a/C5aR1 axis to the malignant phenotype of non-small cell lung cancer cells, particularly in skeletal colonization, a preferential lung metastasis site. METHODS: Association between C5aR1 expression and clinical outcome was assessed in silico and validated by immunohistochemistry. Functional significance was evaluated by lentiviral gene silencing and ligand l-aptamer inhibition in in vivo models of lung cancer bone metastasis. In vitro functional assays for signaling, migration, invasion, metalloprotease activity, and osteoclastogenesis were also performed. MEASUREMENTS AND MAIN RESULTS: High levels of C5aR1 in human lung tumors were significantly associated with shorter recurrence-free survival, overall survival, and bone metastasis. Silencing of C5aR1 in lung cancer cells led to a substantial reduction in skeletal metastatic burden and osteolysis in in vivo models. Furthermore, metalloproteolytic, migratory, and invasive tumor cell activities were modulated in vitro by C5aR1 stimulation or gene silencing. l-Aptamer blockade or C5aR1 silencing significantly reduced the osseous metastatic activity of lung cancer cells in vivo. This effect was associated with decreased osteoclastogenic activity in vitro and was rescued by the exogenous addition of the chemokine CXCL16. CONCLUSIONS: Disruption of C5aR1 signaling in lung cancer cells abrogates their tumor-associated osteoclastogenic activity, impairing osseous colonization. This study unveils the role played by the C5a/C5aR1 axis in lung cancer dissemination and supports its potential use as a novel therapeutic target.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Chemokine CXCL16/immunology , Lung Neoplasms/complications , Lung Neoplasms/immunology , Neoplasm Metastasis/immunology , Receptor, Anaphylatoxin C5a/immunology , Signal Transduction/immunology , Adult , Aged , Aged, 80 and over , Bone Neoplasms/immunology , Female , Humans , Male , Middle Aged
17.
J Dairy Sci ; 102(2): 1457-1472, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30594380

ABSTRACT

In neutrophils, toll-like receptor and complement component 5a (C5a) signaling are critical pathways regulating innate immunity. In cows, not much is known about the second C5a receptor, complement component 5a receptor 2 (C5AR2). It is an interesting player in sepsis treatment because it is considered to have an anti-inflammatory effect during normal inflammation. Periparturient cows are prone to severe infections, and the objectives of this study were to investigate the expression and functionality of C5AR2 during peripartum. We investigated the effect of 2 major inflammatory stimuli, C5a and lipopolysaccharide (LPS), on the expression of a selected number of genes (C5AR1, C5AR2, TLR4, ITGAM, COX2, and CXCL8) and functions linked to these receptors. Overall, TLR4, ITGAM, and C5AR2, all of which are involved in early inflammation, showed a lower expression in periparturient cows. However, an overall lower expression seems not to be the only explanation for the increased risk of sepsis in periparturient cows. Normally, in response to inflammation and as seen in the mid-lactation group, the expression of these genes increases after stimulation with LPS. However, in periparturient cows, stimulation with LPS led to a decrease in expression of these receptors, indicating a different response of neutrophils in response to LPS during this period. A decrease in ITGAM (coding for CD11b) expression complicates correct neutrophil localization and phagocytosis. Its downregulation upon stimulation might be detrimental for adequate eradication of the pathogen and might increase the risk of an imbalanced inflammation; C5AR2 seems to play a central role in this altered response. In addition, myeloperoxidase (MPO) activity in periparturient cows is lower in response to C5a stimulation. It has been suggested that MPO plays an important role in neutrophil shutdown and, thereby, timely resolution of inflammation. A decreased MPO activity might thus prolong the inflammatory reaction of the neutrophils. This finding was supported by the increased viability of the neutrophils obtained from periparturient cows. Even after stimulation, we found a lower caspase-3 activity in this group, indicating that they might be activated for a longer time compared with the neutrophils from mid-lactation cows. Accordingly, these alterations might contribute to a temporal mismatch in inflammatory responses, as often seen in severe periparturient infections.


Subject(s)
Cattle Diseases/immunology , Complement C5a/immunology , Inflammation/immunology , Neutrophils/immunology , Peripartum Period/immunology , Receptor, Anaphylatoxin C5a/immunology , Animals , Biomarkers , Cattle , Female , Gene Expression , Immunity, Innate , Inflammation/metabolism , Lactation/immunology , Lipopolysaccharides/immunology , Parturition/immunology , Phagocytosis , Pregnancy , Sepsis/immunology , Sepsis/veterinary , Signal Transduction
18.
Kidney Int ; 93(3): 615-625, 2018 03.
Article in English | MEDLINE | ID: mdl-29241626

ABSTRACT

The prospects for complement-targeted therapy in ANCA-associated vasculitis have been enhanced by a recent clinical trial in which C5a receptor 1 (C5aR1) inhibition safely replaced glucocorticoids in induction treatment. C5aR1 primes neutrophils for activation by anti-neutrophil cytoplasmic antibody (ANCA) and is therefore required in models of glomerulonephritis induced by anti-myeloperoxidase antibody. Although humoral and cellular autoimmunity play essential roles in ANCA-associated vasculitis, a role for C5aR1 in these responses has not been described. Here, we use murine models to dissect the role of C5aR1 in the generation of anti-myeloperoxidase autoimmunity and the effector responses resulting in renal injury. The genetic absence or pharmacological inhibition of C5aR1 results in reduced autoimmunity to myeloperoxidase with an attenuated Th1 response, increased Foxp3+ regulatory T cells and reduction in generation of myeloperoxidase-ANCA. These changes are mediated by C5aR1 on dendritic cells, which promotes activation, and thus myeloperoxidase autoimmunity and glomerulonephritis. We also use renal intravital microscopy to determine the effect of C5aR1 inhibition on ANCA induced neutrophil dysfunction. We found that myeloperoxidase-ANCA induce neutrophil retention and reactive oxygen species burst within glomerular capillaries. These pathological behaviors are abrogated by C5aR1 inhibition. Thus, C5aR1 inhibition ameliorates both autoimmunity and intra-renal neutrophil activation in ANCA-associated vasculitis.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/immunology , Autoimmunity , Glomerulonephritis/immunology , Kidney Glomerulus/immunology , Neutrophil Activation , Neutrophils/immunology , Peroxidase/immunology , Receptor, Anaphylatoxin C5a/immunology , Animals , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Glomerulonephritis/genetics , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Immunity, Cellular , Immunity, Humoral , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Peroxidase/genetics , Peroxidase/metabolism , Reactive Oxygen Species/metabolism , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Respiratory Burst , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism
19.
J Immunol ; 197(6): 2353-61, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27521340

ABSTRACT

There is accumulating evidence during sepsis that cardiomyocyte (CM) homeostasis is compromised, resulting in cardiac dysfunction. An important role for complement in these outcomes is now demonstrated. Addition of C5a to electrically paced CMs caused prolonged elevations of intracellular Ca(2+) concentrations during diastole, together with the appearance of spontaneous Ca(2+) transients. In polymicrobial sepsis in mice, we found that three key homeostasis-regulating proteins in CMs were reduced: Na(+)/K(+)-ATPase, which is vital for effective action potentials in CMs, and two intracellular Ca(2+) concentration regulatory proteins, that is, sarcoplasmic/endoplasmic reticulum calcium ATPase 2 and the Na(+)/Ca(2+) exchanger. Sepsis caused reduced mRNA levels and reductions in protein concentrations in CMs for all three proteins. The absence of either C5a receptor mitigated sepsis-induced reductions in the three regulatory proteins. Absence of either C5a receptor (C5aR1 or C5aR2) diminished development of defective systolic and diastolic echocardiographic/Doppler parameters developing in the heart (cardiac output, left ventricular stroke volume, isovolumic relaxation, E' septal annulus, E/E' septal annulus, left ventricular diastolic volume). We also found in CMs from septic mice the presence of defective current densities for Ik1, l-type calcium channel, and Na(+)/Ca(2+) exchanger. These defects were accentuated in the copresence of C5a. These data suggest complement-related mechanisms responsible for development of cardiac dysfunction during sepsis.


Subject(s)
Coinfection/immunology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Sepsis/immunology , Sepsis/physiopathology , Animals , Calcium/metabolism , Calcium Channels, L-Type/immunology , Coinfection/microbiology , Coinfection/physiopathology , Complement C5a/immunology , Cytoplasm/chemistry , Cytoplasm/metabolism , Heart/physiopathology , Mice , Myocytes, Cardiac/microbiology , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/immunology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sepsis/complications
20.
Adv Exp Med Biol ; 1074: 45-51, 2018.
Article in English | MEDLINE | ID: mdl-29721926

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of blindness in the USA. Polymorphisms in various complement components are associated with an increased risk for AMD, and it has been hypothesized that an overactive complement system is partially responsible for the pathology of AMD. AMD is classified as early, intermediate, or late AMD, depending on the degree of the associated pathologies. Late AMD can be characterized as either lesions associated with neovascular AMD or geographic atrophy. Both sets of lesions are associated with pathology at the RPE/choroid interface, which include a thickening of Bruch's membrane, presence of drusen, and pigmentary alterations, and deterioration of the blood-retina barrier has been reported. These changes can lead to the slow degeneration and atrophy of the photoreceptors in the macula in dry AMD, or progress to choroidal neovascularization (CNV) and leakage of these new vessels in wet AMD. It has been shown previously that complement anaphylatoxins C3a and C5a, signaling via their respective G-protein-coupled receptors, can alter RPE cell function and promote choroidal neovascularization. However, it is important to note these components also play a role in tissue repair. Here we discuss anaphylatoxin signaling in AMD-related target cells and the potential implications for the design of anti-complement therapeutics.


Subject(s)
Complement Activation , Complement C3/immunology , Complement C5a/immunology , Macular Degeneration/immunology , Receptor, Anaphylatoxin C5a/immunology , Aging/immunology , Animals , Bruch Membrane/pathology , Choroid/pathology , Choroidal Neovascularization/immunology , Cytokines/biosynthesis , Endothelial Cells/pathology , Fibrosis , Forecasting , Humans , Inflammasomes/metabolism , Inflammation , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Mice , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/pathology , Retinal Rod Photoreceptor Cells/immunology , Retinal Rod Photoreceptor Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL