Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters

Publication year range
1.
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
2.
Circulation ; 137(20): 2152-2165, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29348261

ABSTRACT

BACKGROUND: Defining conserved molecular pathways in animal models of successful cardiac regeneration could yield insight into why adult mammals have inadequate cardiac regeneration after injury. Insight into the transcriptomic landscape of early cardiac regeneration from model organisms will shed light on evolutionarily conserved pathways in successful cardiac regeneration. METHODS: Here we describe a cross-species transcriptomic screen in 3 model organisms for cardiac regeneration: axolotl, neonatal mice, and zebrafish. Apical resection to remove ≈10% to 20% of ventricular mass was carried out in these model organisms. RNA-sequencing analysis was performed on the hearts harvested at 3 time points: 12, 24, and 48 hours after resection. Sham surgery was used as internal control. RESULTS: Genes associated with inflammatory processes were found to be upregulated in a conserved manner. Complement receptors (activated by complement components, part of the innate immune system) were found to be highly upregulated in all 3 species. This approach revealed induction of gene expression for complement 5a receptor 1 in the regenerating hearts of zebrafish, axolotls, and mice. Inhibition of complement 5a receptor 1 significantly attenuated the cardiomyocyte proliferative response to heart injury in all 3 species. Furthermore, after left ventricular apical resection, the cardiomyocyte proliferative response was diminished in mice with genetic deletion of complement 5a receptor 1. CONCLUSIONS: These data reveal that activation of complement 5a receptor 1 mediates an evolutionarily conserved response that promotes cardiomyocyte proliferation after cardiac injury and identify complement pathway activation as a common pathway of successful heart regeneration.


Subject(s)
Evolution, Molecular , Heart/physiology , Receptor, Anaphylatoxin C5a/metabolism , Regeneration/physiology , Ambystoma mexicanum , Animals , Animals, Newborn , Cell Proliferation , Gene Expression Profiling , Gene Ontology , Mice , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Peptides, Cyclic/pharmacology , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/genetics , Sequence Analysis, RNA , Troponin T/analysis , Zebrafish
3.
Respir Res ; 20(1): 133, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31262295

ABSTRACT

BACKGROUND: Cystic fibrosis (CF) is an inherited disorder caused by mutations in the CF transmembrane conductance regulator (CFTR) gene that promotes persistent lung infection and inflammation and progressive loss of lung function. Patients with CF have increased lung lymphoid follicles (LFs) and B cell-activating factor of tumor necrosis factor family (BAFF) that regulates B cell survival and maturation. A direct role for CFTR in B cell activation and disease pathogenesis in CF remains unclear. METHODS: The number of LFs, BAFF+, TLR4+ and proliferation marker Ki67+ B cells in lung explants or resections from subjects with CF and normal controls was quantified by immunostaining. The role of CFTR in B cell activation and LF development was then examined in two independent cohorts of uninfected CFTR-deficient mice (Cftr -/-) and wild type controls. The number of lung LFs, B cells and BAFF+, CXCR4+, immunoglobulin G+ B cells was examined by immunostaining. Lung and splenocyte B cell activation marker and major histocompatibility complex class II (MHC class II) expression was quantified by flow cytometry. Inflammatory cytokine levels were measured in supernatants from isolated B cells from Cftr -/- and wild type mice stimulated in vitro with Pseudomonas aeruginosa lipopolysaccharide (LPS). RESULTS: There was a significant increase in well-formed LFs in subjects with CF compared to normal controls. Increased B cell activation and proliferation was observed in lung LFs from CF subjects as was quantified by a significant increase in B cell BAFF, TLR4 and Ki67 expression. Uninfected Cftr -/- mice had increased lung LFs and BAFF+ and CXCR4+ B cells compared to wild type controls. Lung B cells isolated from uninfected Cftr -/- mice demonstrated increased MHC class II expression. In vitro, isolated B cells from Cftr -/- mice produced increased IL-6 when stimulated with LPS compared to wild type controls. CONCLUSIONS: These data support a direct role for CFTR in B cell activation, proliferation and inflammatory cytokine production that promotes lung LF follicle development in cystic fibrosis.


Subject(s)
B-Lymphocytes/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cystic Fibrosis/metabolism , Tertiary Lymphoid Structures/metabolism , Adolescent , Animals , Cystic Fibrosis/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Am J Pathol ; 187(6): 1368-1379, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28427861

ABSTRACT

Costimulatory molecules, such as the programmed death ligand (PD-L1), might exert differential effects on T-cell function, depending on the clinical setting and/or immunological environment. Given the impact of T cells on bronchiolitis obliterans (BO) in lung transplantation, we used an established tracheal transplant model inducing BO-like lesions to investigate the impact of PD-L1 on alloimmune responses and histopathological outcome in BO. In contrast to other transplant models in which PD-L1 generally shows protective functions, we demonstrated that PD-L1 has divergent effects depending on its location in donor versus recipient tissue. Although PD-L1 deficiency in donor tissue worsened histopathological outcome, and increased systemic inflammatory response, recipient PD-L1 deficiency induced opposite effects. Mechanistic studies revealed PD-L1-deficient recipients were hyporesponsive toward alloantigen, despite increased numbers of CD8+ effector T cells. The function of PD-L1 on T cells after unspecific stimulation was dependent on both cell type and strength of stimulation. This novel function of recipient PD-L1 may result from the high degree of T-cell activation within the highly immunogenic milieu of the transplanted tissue. In this model, both decreased T-cell alloimmune responses and the reduction of BO in PD-L1-deficient recipients suggest a potential therapeutic role of selectively blocking PD-L1 in the recipient. Further investigation is warranted to determine the impact of this finding embedded in the complex pathophysiological context of BO.


Subject(s)
B7-H1 Antigen/immunology , Bronchiolitis Obliterans/immunology , Trachea/transplantation , Transplantation Immunology , Animals , B7-H1 Antigen/deficiency , Bronchiolitis Obliterans/pathology , Bronchiolitis Obliterans/prevention & control , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Epithelial Cells/immunology , Graft Survival/immunology , Immune Tolerance/immunology , Immunity, Cellular , Isoantigens/immunology , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Tissue Donors , Trachea/pathology , Up-Regulation/immunology
5.
Am J Respir Cell Mol Biol ; 57(6): 711-720, 2017 12.
Article in English | MEDLINE | ID: mdl-28708422

ABSTRACT

Cystic fibrosis (CF) remains the most lethal genetic disease in the Caucasian population. However, there is great variability in clinical phenotypes and survival times, even among patients harboring the same genotype. We identified five patients with CF and a homozygous F508del mutation in the CFTR gene who were in their fifth or sixth decade of life and had shown minimal changes in lung function over a longitudinal period of more than 20 years. Because of the rarity of this long-term nonprogressive phenotype, we hypothesized these individuals may carry rare genetic variants in modifier genes that ameliorate disease severity. Individuals at the extremes of survival time and lung-function trajectory underwent whole-exome sequencing, and the sequencing data were filtered to include rare missense, stopgain, indel, and splicing variants present with a mean allele frequency of <0.2% in general population databases. Epithelial sodium channel (ENaC) mutants were generated via site-directed mutagenesis and expressed for Xenopus oocyte assays. Four of the five individuals carried extremely rare or never reported variants in the SCNN1D and SCNN1B genes of the ENaC. Separately, an independently enriched rare variant in SCNN1D was identified in the Exome Variant Server database associated with a milder pulmonary disease phenotype. Functional analysis using Xenopus oocytes revealed that two of the three variants in δ-ENaC encoded by SCNN1D exhibited hypomorphic channel activity. Our data suggest a potential role for δ-ENaC in controlling sodium reabsorption in the airways, and advance the plausibility of ENaC as a therapeutic target in CF.


Subject(s)
Amino Acid Sequence , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Epithelial Sodium Channels/metabolism , Sequence Deletion , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Sodium Channels/genetics , Female , Humans , Male , Xenopus , Xenopus laevis
6.
Infect Immun ; 85(6)2017 06.
Article in English | MEDLINE | ID: mdl-28348057

ABSTRACT

Burkholderia dolosa caused an outbreak in the cystic fibrosis (CF) clinic at Boston Children's Hospital from 1998 to 2005 and led to the infection of over 40 patients, many of whom died due to complications from infection by this organism. To assess whether B. dolosa significantly contributes to disease or is recognized by the host immune response, mice were infected with a sequenced outbreak B. dolosa strain, AU0158, and responses were compared to those to the well-studied CF pathogen Pseudomonas aeruginosa In parallel, mice were also infected with a polar flagellin mutant of B. dolosa to examine the role of flagella in B. dolosa lung colonization. The results showed a higher persistence in the host by B. dolosa strains, and yet, neutrophil recruitment and cytokine production were lower than those with P. aeruginosa The ability of host immune cells to recognize B. dolosa was then assessed, B. dolosa induced a robust cytokine response in cultured cells, and this effect was dependent on the flagella only when bacteria were dead. Together, these results suggest that B. dolosa can be recognized by host cells in vitro but may avoid or suppress the host immune response in vivo through unknown mechanisms. B. dolosa was then compared to other Burkholderia species and found to induce similar levels of cytokine production despite being internalized by macrophages more than Burkholderia cenocepacia strains. These data suggest that B. dolosa AU0158 may act differently with host cells and is recognized differently by immune systems than are other Burkholderia strains or species.


Subject(s)
Burkholderia Infections/immunology , Cystic Fibrosis/complications , Cytokines/immunology , Flagella/immunology , Flagellin/genetics , Animals , Bronchoalveolar Lavage , Burkholderia/genetics , Burkholderia/immunology , Burkholderia Infections/microbiology , Cell Line , Cystic Fibrosis/microbiology , Disease Models, Animal , Epidemics , Female , Flagella/genetics , Humans , Lung/microbiology , Mice , Mice, Inbred C57BL , Mutation , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/immunology
7.
Immunity ; 29(1): 101-13, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18617426

ABSTRACT

Innate recognition of invading pathogens in peripheral tissues results in the recruitment of circulating memory CD8(+) T cells to sites of localized inflammation during the early phase of a recall response. However, the mechanisms that control the rapid recruitment of these cells to peripheral sites are poorly understood, particularly in relation to influenza and parainfluenza infections of the respiratory tract. In this study, we demonstrate a crucial role for C-C chemokine receptor 5 (CCR5) in the accelerated recruitment of memory CD8(+) T cells to the lung airways during virus challenge. Most importantly, CCR5 deficiency resulted in decreased recruitment of memory T cells expressing key effector molecules and impaired control of virus replication during the initial stages of a secondary response. These data highlight the critical importance of early memory T cell recruitment for the efficacy of cellular immunity in the lung.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Receptors, CCR5/immunology , Respiratory Tract Infections/immunology , Virus Diseases/immunology , Animals , Chemotaxis, Leukocyte/immunology , Flow Cytometry , Mice , Orthomyxoviridae/immunology , Receptors, CXCR3/immunology , Respiratory Tract Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , Sendai virus/immunology
8.
J Immunol ; 195(3): 1034-43, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26091719

ABSTRACT

Staphylococcus aureus is well adapted to the human host. Evasion of the host phagocyte response is critical for successful infection. The staphylococcal bicomponent pore-forming toxins Panton-Valentine leukocidin LukSF-PV (PVL) and γ-hemolysin CB (HlgCB) target human phagocytes through interaction with the complement receptors C5aR1 and C5aR2. Currently, the apparent redundancy of both toxins cannot be adequately addressed in experimental models of infection because mice are resistant to PVL and HlgCB. The molecular basis for species specificity of the two toxins in animal models is not completely understood. We show that PVL and HlgCB feature distinct activity toward neutrophils of different mammalian species, where activity of PVL is found to be restricted to fewer species than that of HlgCB. Overexpression of various mammalian C5a receptors in HEK cells confirms that cytotoxicity toward neutrophils is driven by species-specific interactions of the toxins with C5aR1. By taking advantage of the species-specific engagement of the toxins with their receptors, we demonstrate that PVL and HlgCB differentially interact with human C5aR1 and C5aR2. In addition, binding studies illustrate that different parts of the receptor are involved in the initial binding of the toxin and the subsequent formation of lytic pores. These findings allow a better understanding of the molecular mechanism of pore formation. Finally, we show that the toxicity of PVL, but not of HlgCB, is neutralized by various C5aR1 antagonists. This study offers directions for the development of improved preclinical models for infection, as well as for the design of drugs antagonizing leukocidin toxicity.


Subject(s)
Bacterial Proteins/immunology , Bacterial Toxins/immunology , Exotoxins/immunology , Hemolysin Proteins/immunology , Leukocidins/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Chemokine/immunology , Amino Acid Sequence , Animals , Cattle , Cell Line , HEK293 Cells , Humans , Immune Evasion/immunology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neutrophils/immunology , Phagocytes/immunology , Protein Binding , Protein Structure, Tertiary , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptors, Chemokine/antagonists & inhibitors , Staphylococcal Infections/immunology , Staphylococcus aureus/pathogenicity
9.
Am J Respir Cell Mol Biol ; 55(5): 657-666, 2016 11.
Article in English | MEDLINE | ID: mdl-27285858

ABSTRACT

LPS-induced lung injury in the mouse is one of the most robust experimental models used for studies of acute lung injury (ALI) and acute respiratory distress syndrome in humans. Prior clinical and experimental studies support an important role for complement activation, particularly production of C5a, in the pathophysiology of human ALI/acute respiratory distress syndrome. In the mouse model, however, the precise role of C5a and its receptors is unclear. C5L2, an enigmatic second receptor for C5a, has been characterized, and results have generated substantial debate regarding its in vivo function. Our previous work with human neutrophils revealed a unique role for C5L2 in negatively modulating C5a-C5a receptor (C5aR)-mediated cellular activation, in which antibody-mediated blockade of C5L2 resulted in augmented C5a-C5aR responses. Here, we demonstrate that C5L2-/- mice (BALB/c background) administered intranasal LPS exhibit significantly more airway edema and hemorrhage than do wild-type animals. Bronchoalveolar lavage fluid and lung homogenates have significantly more neutrophils and myeloperoxidase activity, as well as proinflammatory cytokines and chemokines. When a blocking antibody against the C5aR was administered before LPS administration, the increased neutrophilic infiltration and cytokine levels were reversed. Thus, our data show not only that C5a contributes significantly to LPS-induced ALI in the mouse, but also that C5L2 plays an important antiinflammatory role in this model through actions resulting at least in part from negative modulation of C5a receptor activation.


Subject(s)
Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Receptors, Chemokine/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/physiopathology , Administration, Intranasal , Animals , Bronchoalveolar Lavage Fluid , Chemokines/genetics , Chemokines/metabolism , Edema/complications , Edema/pathology , Edema/physiopathology , Gene Expression Regulation , Hemorrhage/complications , Hemorrhage/pathology , Hemorrhage/physiopathology , Inflammation Mediators/metabolism , Lipopolysaccharides/administration & dosage , Lung/pathology , Mice, Inbred BALB C , Neutrophils/metabolism , Peroxidase/metabolism , Receptor, Anaphylatoxin C5a , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/deficiency , Respiratory Mechanics , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
10.
J Immunol ; 191(8): 4001-9, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24043888

ABSTRACT

The complement anaphylatoxin C5a is a critical mediator of allergic contact dermatitis, bridging essential aspects of innate and adaptive immunity. This anaphylatoxin functions by interacting with two 7-transmembrane segment receptors, the C5aR and C5L2. The C5aR is a classical G protein coupled receptor, whereas C5L2 is deficient in coupling to G proteins because of variations in the sequence. Our previous work in human neutrophils revealed a unique role for C5L2 in negatively modulating anaphylatoxin receptor mediated cellular activation through interactions with ß-arrestin. When C5L2 is deficient, C5aR-mediated ß-arrestin signaling is greatly enhanced. The work described in this study was undertaken first to determine the effect of C5L2 deficiency in a murine model of contact sensitivity, and second to determine whether the resultant exacerbation of inflammatory parameters reflects a negative modulatory function of C5L2 on the C5aR. First, we find dramatic increases in inflammation in C5L2(-/-) animals compared with wild type mice. Second, these increases are completely reversed following administration of mAb against the C5aR. Thus, in allergic contact sensitivity, as in isolated human neutrophils, C5L2 functions to suppress C5a-C5aR-mediated responses, further underscoring its role as a negative regulator of anaphylatoxin activity.


Subject(s)
Complement C5a/immunology , Dermatitis, Allergic Contact/immunology , Inflammation/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Chemokine/immunology , Animals , Antibodies, Monoclonal/immunology , Arrestins/immunology , Arrestins/metabolism , Female , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Signal Transduction , beta-Arrestins
11.
J Immunol ; 190(9): 4836-47, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23547118

ABSTRACT

According to the current paradigm, lymphocyte homing to the small intestine requires the expression of two tissue-specific homing receptors, the integrin α4ß7 and the CCL25 receptor CCR9. In this study, we investigated the organ distribution and the homing molecule expression of IgA Ab-secreting cells (ASCs) induced by intrarectal immunization with a particulate Ag, in comparison with other mucosal immunization routes. Intrarectal immunization induces gut-homing IgA ASCs that localize not only in the colon but also in the small intestine, although they are not responsive to CCL25, unlike IgA ASCs induced by oral immunization. The mucosal epithelial chemokine CCL28, known to attract all IgA ASCs, does not compensate for the lack of CCL25 responsiveness, because the number of Ag-specific cells is not decreased in the gut of CCR10-deficient mice immunized by the intrarectal route. However, Ag-specific IgA ASCs induced by intrarectal immunization express the integrin α4ß7, and their number is considerably decreased in the gut of ß7-deficient mice immunized by the intrarectal route, indicating that α4ß7 enables these cells to migrate into the small intestine, even without CCL25 responsiveness. In contrast, IgA ASCs induced by intranasal immunization express low α4ß7 levels and are usually excluded from the gut. Paradoxically, after intranasal immunization, Ag-specific IgA ASCs are significantly increased in the small intestine of ß7-deficient mice, demonstrating that lymphocyte homing is a competitive process and that integrin α4ß7 determines not only the intestinal tropism of IgA ASCs elicited in GALTs but also the intestinal exclusion of lymphocytes primed in other inductive sites.


Subject(s)
Antibody-Producing Cells/immunology , Immunoglobulin A/immunology , Intestine, Small/immunology , Administration, Rectal , Animals , Antibody-Producing Cells/metabolism , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Chemokines, CC/immunology , Chemokines, CC/metabolism , Colon/immunology , Colon/metabolism , Female , Immunization/methods , Immunoglobulin A/metabolism , Integrins/immunology , Integrins/metabolism , Intestine, Small/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mucoproteins , Mucous Membrane/immunology , Mucous Membrane/metabolism , Receptors, CCR10/immunology , Receptors, CCR10/metabolism
12.
Nature ; 462(7272): 510-3, 2009 Nov 26.
Article in English | MEDLINE | ID: mdl-19898495

ABSTRACT

CD4(+) T helper cells are well known for their role in providing critical signals during priming of cytotoxic CD8(+) T lymphocyte (CTL) responses in vivo. T-cell help is required for the generation of primary CTL responses as well as in promoting protective CD8(+) memory T-cell development. However, the role of CD4 help in the control of CTL responses at the effector stage is unknown. Here we show that fully helped effector CTLs are themselves not self-sufficient for entry into the infected tissue, but rely on the CD4(+) T cells to provide the necessary cue. CD4(+) T helper cells control the migration of CTL indirectly through the secretion of IFN-gamma and induction of local chemokine secretion in the infected tissue. Our results reveal a previously unappreciated role of CD4 help in mobilizing effector CTL to the peripheral sites of infection where they help to eliminate infected cells.


Subject(s)
Chemotaxis , Herpesvirus 2, Human/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vagina/immunology , Vagina/virology , Adoptive Transfer , Animals , Chemokines/immunology , Chemokines/metabolism , Female , Herpes Simplex/immunology , Herpes Simplex/virology , Immunity, Mucosal/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Models, Immunological , Mucous Membrane/immunology , Mucous Membrane/virology , Receptors, CXCR3/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
13.
Nature ; 460(7252): 225-30, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19525930

ABSTRACT

Age-related macular degeneration (AMD), a leading cause of blindness worldwide, is as prevalent as cancer in industrialized nations. Most blindness in AMD results from invasion of the retina by choroidal neovascularisation (CNV). Here we show that the eosinophil/mast cell chemokine receptor CCR3 is specifically expressed in choroidal neovascular endothelial cells in humans with AMD, and that despite the expression of its ligands eotaxin-1, -2 and -3, neither eosinophils nor mast cells are present in human CNV. Genetic or pharmacological targeting of CCR3 or eotaxins inhibited injury-induced CNV in mice. CNV suppression by CCR3 blockade was due to direct inhibition of endothelial cell proliferation, and was uncoupled from inflammation because it occurred in mice lacking eosinophils or mast cells, and was independent of macrophage and neutrophil recruitment. CCR3 blockade was more effective at reducing CNV than vascular endothelial growth factor A (VEGF-A) neutralization, which is in clinical use at present, and, unlike VEGF-A blockade, is not toxic to the mouse retina. In vivo imaging with CCR3-targeting quantum dots located spontaneous CNV invisible to standard fluorescein angiography in mice before retinal invasion. CCR3 targeting might reduce vision loss due to AMD through early detection and therapeutic angioinhibition.


Subject(s)
Macular Degeneration/diagnosis , Macular Degeneration/therapy , Receptors, CCR3/antagonists & inhibitors , Receptors, CCR3/metabolism , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Chemokine CCL11/antagonists & inhibitors , Chemokine CCL11/metabolism , Chemokine CCL24/antagonists & inhibitors , Chemokine CCL24/metabolism , Chemokine CCL26 , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/metabolism , Choroid/blood supply , Choroid/cytology , Choroid/metabolism , Choroidal Neovascularization/diagnosis , Choroidal Neovascularization/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Inflammation , Leukocytes , Ligands , Macular Degeneration/metabolism , Mice , Mice, Inbred C57BL , Quantum Dots , Receptors, CCR3/analysis , Receptors, CCR3/genetics , Receptors, CCR3/immunology , Retina/drug effects , Retina/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology
14.
J Am Soc Nephrol ; 25(2): 225-31, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24179165

ABSTRACT

Necrotizing and crescentic GN (NCGN) with a paucity of glomerular immunoglobulin deposits is associated with ANCA. The most common ANCA target antigens are myeloperoxidase (MPO) and proteinase 3. In a manner that requires activation of the alternative complement pathway, passive transfer of antibodies to mouse MPO (anti-MPO) induces a mouse model of ANCA NCGN that closely mimics human disease. Here, we confirm the importance of C5aR/CD88 in the mediation of anti-MPO-induced NCGN and report that C6 is not required. We further demonstrate that deficiency of C5a-like receptor (C5L2) has the reverse effect of C5aR/CD88 deficiency and results in more severe disease, indicating that C5aR/CD88 engagement enhances inflammation and C5L2 engagement suppresses inflammation. Oral administration of CCX168, a small molecule antagonist of human C5aR/CD88, ameliorated anti-MPO-induced NCGN in mice expressing human C5aR/CD88. These observations suggest that blockade of C5aR/CD88 might have therapeutic benefit in patients with ANCA-associated vasculitis and GN.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/prevention & control , Autoantigens/immunology , Glomerulonephritis/prevention & control , Peroxidase/immunology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Administration, Oral , Animals , Complement C6/immunology , Complement Pathway, Alternative , Dose-Response Relationship, Drug , Gene Knock-In Techniques , Glomerulonephritis/complications , Glomerulonephritis/immunology , Hematuria/etiology , Hematuria/prevention & control , Humans , Immunization, Passive , Leukocytes , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Peroxidase/deficiency , Proteinuria/etiology , Proteinuria/prevention & control , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/genetics , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Receptors, Chemokine/physiology , Recombinant Fusion Proteins , Urine/cytology
15.
J Biol Chem ; 288(49): 35039-48, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24145037

ABSTRACT

Chemokines display considerable promiscuity with multiple ligands and receptors shared in common, a phenomenon that is thought to underlie their biochemical "redundancy." Their receptors are part of a larger seven-transmembrane receptor superfamily, commonly referred to as G protein-coupled receptors, which have been demonstrated to be able to signal with different efficacies to their multiple downstream signaling pathways, a phenomenon referred to as biased agonism. Biased agonism has been primarily reported as a phenomenon of synthetic ligands, and the biologic prevalence and importance of such signaling are unclear. Here, to assess the presence of biased agonism that may underlie differential signaling by chemokines targeting the same receptor, we performed a detailed pharmacologic analysis of a set of chemokine receptors with multiple endogenous ligands using assays for G protein signaling, ß-arrestin recruitment, and receptor internalization. We found that chemokines targeting the same receptor can display marked differences in their efficacies for G protein- or ß-arrestin-mediated signaling or receptor internalization. This ligand bias correlates with changes in leukocyte migration, consistent with different mechanisms underlying the signaling downstream of these receptors induced by their ligands. These findings demonstrate that biased agonism is a common and likely evolutionarily conserved biological mechanism for generating qualitatively distinct patterns of signaling via the same receptor in response to different endogenous ligands.


Subject(s)
Receptors, Chemokine/agonists , Receptors, Chemokine/metabolism , Arrestins/metabolism , Chemokines/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Kinetics , Ligands , Models, Biological , Signal Transduction , beta-Arrestins
16.
Infect Immun ; 82(1): 371-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24191300

ABSTRACT

The host immune response plays an important role in the onset and progression of cerebral malaria (CM). The complement system is an essential component of the innate immune response to malaria, and its activation generates the anaphylatoxin C5a. To test the hypothesis that C5a signaling contributes to the pathogenesis of CM, we investigated a causal role for the C5a receptors C5aR and C5L2 in a mouse model of experimental CM (ECM) induced by Plasmodium berghei ANKA infection, and using a case-control design, we examined levels of C5a in plasma samples from Ugandan children presenting with CM or uncomplicated malaria (UM). In the ECM model, C5aR(-/-) mice displayed significantly improved survival compared to their wild-type (WT) counterparts (P = 0.004), whereas C5L2(-/-) mice showed no difference in survival from WT mice. Improved survival in C5aR(-/-) mice was associated with reduced levels of the proinflammatory cytokines tumor necrosis factor (TNF) and gamma interferon (IFN-γ) and the chemokine, monocyte chemoattractant protein 1 (MCP-1) (CCL2). Furthermore, endothelial integrity was enhanced, as demonstrated by increased levels of angiopoietin-1, decreased levels of angiopoietin-2 and soluble ICAM-1, and decreased Evans blue extravasation into brain parenchyma. In the case-control study, the median levels of C5a at presentation were significantly higher in children with CM versus those in children with UM (43.7 versus 22.4 ng/ml; P < 0.001). These findings demonstrate that C5a is dysregulated in human CM and contributes to the pathogenesis of ECM via C5aR-dependent inflammation and endothelial dysfunction.


Subject(s)
Complement C5a/immunology , Malaria, Cerebral/immunology , Receptors, Chemokine/immunology , Receptors, Complement/immunology , Animals , Case-Control Studies , Child , Child, Preschool , Complement C5a/deficiency , Disease Models, Animal , Female , Humans , Infant , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , Receptor, Anaphylatoxin C5a , Receptors, Complement/deficiency , Receptors, Concanavalin A
17.
J Allergy Clin Immunol ; 131(2): 541-8.e1-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22728083

ABSTRACT

BACKGROUND: Mast cells express receptors for complement anaphylatoxins C3a and C5a (ie, C3a receptor [C3aR] and C5a receptor [C5aR]), and C3a and C5a are generated during various IgE-dependent immediate hypersensitivity reactions in vivo. However, it is not clear to what extent mast cell expression of C3aR or C5aR influences C3a- or C5a-induced cutaneous responses or IgE-dependent mast cell activation and passive cutaneous anaphylaxis (PCA) in vivo. OBJECTIVE: We sought to assess whether mouse skin mast cell expression of C3aR or C5aR influences (1) the cells' responsiveness to intradermal injections of C3a or C5a or (2) the extent of IgE-dependent mast cell degranulation and PCA in vivo. METHODS: We measured the magnitude of cutaneous responses to intradermal injections of C3a or C5a and the extent of IgE-dependent mast cell degranulation and PCA responses in mice containing mast cells that did or did not express C3aR or C5aR. RESULTS: The majority of the skin swelling induced by means of intradermal injection of C3a or C5a required that mast cells at the site expressed C3aR or C5aR, respectively, and the extent of IgE-dependent degranulation of skin mast cells and IgE-dependent PCA was significantly reduced when mast cells lacked either C3aR or C5aR. IgE-dependent PCA responses associated with local increases in C3a levels occurred in antibody-deficient mice but not in mice deficient in FcɛRIγ. CONCLUSION: Expression of C3aR and C5aR by skin mast cells contributes importantly to the ability of C3a and C5a to induce skin swelling and can enhance mast cell degranulation and inflammation during IgE-dependent PCA in vivo.


Subject(s)
Immunoglobulin E/metabolism , Inflammation/immunology , Mast Cells/immunology , Receptor, Anaphylatoxin C5a/biosynthesis , Receptors, Complement/biosynthesis , Skin/immunology , Anaphylatoxins/genetics , Anaphylatoxins/immunology , Animals , Cells, Cultured , Complement C3a/genetics , Complement C3a/immunology , Complement C3a/metabolism , Complement C5a/genetics , Complement C5a/immunology , Complement C5a/metabolism , Female , Immunoglobulin E/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/immunology , Receptors, Complement/genetics , Receptors, Complement/immunology , Receptors, Complement/metabolism , Skin/metabolism , Skin/pathology
18.
Blood ; 117(2): 480-8, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20980681

ABSTRACT

We investigated possible cellular receptors for the human CXC chemokine platelet factor-4 variant/CXCL4L1, a potent inhibitor of angiogenesis. We found that CXCL4L1 has lower affinity for heparin and chondroitin sulfate-E than platelet factor-4 (CXCL4) and showed that CXCL10 and CXCL4L1 could displace each other on microvascular endothelial cells. Labeled CXCL4L1 also bound to CXCR3A- and CXCR3B-transfectants and was displaced by CXCL4L1, CXCL4, and CXCL10. The CXCL4L1 anti-angiogenic activity was blocked by anti-CXCR3 antibodies (Abs) in the Matrigel and cornea micropocket assays. CXCL4L1 application in CXCR3(-/-) or in wild-type mice treated with neutralizing anti-CXCR3 Abs, resulted in reduced inhibitory activity of CXCL4L1 on tumor growth and vascularization of Lewis lung carcinoma. Furthermore, CXCL4L1 and CXCL4 chemoattracted activated T cells, human natural killer cells, and human immature dendritic cells (DCs). Migration of DCs toward CXCL4 and CXCL4L1 was desensitized by preincubation with CXCL10 and CXCL11, inhibited by pertussis toxin, and neutralized by anti-CXCR3 Abs. Chemotaxis of T cells, natural killer cells, and DCs is likely to contribute to the antitumoral action. However, the in vivo data indicate that the angiostatic property of CXCL4L1 is equally important in retarding tumor growth. Thus, both CXCR3A and CXCR3B are implicated in the chemotactic and vascular effects of CXCL4L1.


Subject(s)
Angiogenesis Inhibitors/metabolism , Chemotactic Factors/metabolism , Platelet Factor 4/metabolism , Receptors, CXCR3/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Chemotactic Factors/pharmacology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Dendritic Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Platelet Factor 4/pharmacology , Rats , Transfection , Xenograft Model Antitumor Assays
19.
J Immunol ; 187(8): 4245-55, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21918196

ABSTRACT

Respiratory syncytial virus (RSV) infection is associated with serious lung disease in infants and immunocompromised individuals and is linked to development of asthma. In mice, acute RSV infection causes airway hyperresponsiveness (AHR), inflammation, and mucus hypersecretion. Infected cells induce complement activation, producing the anaphylatoxin C3a. In this paper, we show RSV-infected wild-type mice produce Th17 cytokines, a response not previously associated with viral infections. Mice deficient in the C3aR fail to develop AHR following acute RSV infection, and production of Th17 cytokines was significantly attenuated. Tachykinin production also has been implicated in RSV pathophysiology, and tachykinin receptor-null mice were similarly protected from developing AHR. These animals were also deficient in production of Th17 cytokines. Tachykinin release was absent in mice deficient in C3aR, whereas C3a levels were unchanged in tachykinin receptor-null animals. Thus, our data reveal a crucial sequence following acute RSV infection where initial C3a production causes tachykinin release, followed by activation of the IL-17A pathway. Deficiency of either receptor affords protection from AHR, identifying two potential therapeutic targets.


Subject(s)
Bronchial Hyperreactivity/immunology , Complement C3a/immunology , Interleukin-17/immunology , Respiratory Syncytial Virus Infections/immunology , Tachykinins/immunology , Animals , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/virology , Cell Separation , Complement C3a/metabolism , Cytokines/immunology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Gene Expression Profiling , Interleukin-17/metabolism , Mice , Mice, Knockout , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/immunology , Reverse Transcriptase Polymerase Chain Reaction , Tachykinins/metabolism
20.
Proc Natl Acad Sci U S A ; 107(2): 628-32, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-20018651

ABSTRACT

Ubiquitously expressed seven-transmembrane receptors (7TMRs) classically signal through heterotrimeric G proteins and are commonly referred to as G protein-coupled receptors. It is now recognized that 7TMRs also signal through beta-arrestins, which act as versatile adapters controlling receptor signaling, desensitization, and trafficking. Most endogenous receptors appear to signal in a balanced fashion using both beta-arrestin and G protein-mediated pathways. Some 7TMRs are thought to be nonsignaling "decoys" because of their inability to activate typical G protein signaling pathways; it has been proposed that these receptors act to scavenge ligands or function as coreceptors. Here we demonstrate that ligand binding to the decoy receptor CXCR7 does not result in activation of signaling pathways typical of G proteins but does activate MAP kinases through beta-arrestins in transiently transfected cells. Furthermore, we observe that vascular smooth muscle cells that endogenously express CXCR7 migrate to its ligand interferon-inducible T-cell alpha chemoattractant (ITAC), an effect that is significantly attenuated by treatment with either a CXCR7 antagonist or beta-arrestin depletion by siRNA. This example of an endogenous "beta-arrestin-biased" 7TMR that signals through beta-arrestin in the absence of G protein activation demonstrates that some 7TMRs encoded in the genome have evolved to signal through beta-arrestin exclusively and suggests that other receptors that are currently thought to be orphans or decoys may also signal through such nonclassical pathways.


Subject(s)
Arrestins/physiology , Receptors, CXCR/physiology , T-Lymphocytes/physiology , Animals , Cell Line , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/physiology , Humans , Mice , Muscle, Smooth, Vascular/physiology , Phosphorylation , RNA, Messenger/genetics , Rats , Receptors, CXCR/genetics , Receptors, Chemokine/genetics , Signal Transduction/physiology , T-Lymphocytes/cytology , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL