Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 64
1.
Front Immunol ; 12: 768115, 2021.
Article En | MEDLINE | ID: mdl-34966389

Background: It was reported that tumor heterogeneity and the surrounding tumor microenvironment (TME) in ovarian cancer affects immunotherapy efficacy and patient outcomes. And the TME of ovarian cancer is intrinsically heterogeneous. CD47 plays vital roles in cell functional behavior and immune homeostasis relating to cancer prognosis. But how it affects TME and its contribution to heterogeneity in ovarian cancer has not been fully illustrated. Therefore, we aimed to identify a prognostic biomarker which may help explain tumor immune microenvironment heterogeneity of ovarian cancer. Methods: Cancer single-cell state atlas (CancerSEA) was used to evaluate functional role of CD47. Several bioinformatics database including Oncomine, Gene Expression Profiling Interaction Analysis (GEPIA), Tumor Immune Estimation Resource (TIMER), The Human Protein Atlas (HPA), Ualcan and Kaplan-Meier plotter (KM plotter) were applied to illustrate correlation of CD47 with ovarian cancer prognosis and immune infiltration. Tumor Immune Single-cell Hub (TISCH) single cell database was employed to evaluate correlation of CD47 with tumor microenvironment. GeneMANIA was implemented to identify regulation networks of CD47. Differentially expressed genes (DEGs) between CD47 high and low expression groups were analyzed with R package DESeq2. Kyoto encyclopedia of genes and genomes (KEGG) and Gene Set Enrichment Analysis (GSEA) were utilized to explore how CD47 affect the immune related cell signaling pathway. Results: CD47 expression was upregulated and connected to worse OS and PFS in ovarian cancer. Close relation was found between CD47 expression level and immune infiltration in ovarian cancer, especially with Treg cells, Monocytes, Macrophages and T cell exhaustion (P<0.05). The CD47 expression level was relatively low in plasma cells, dendritic cells and Mono/Macro cells of OV_GSE115007, in myofibroblasts, fibroblasts and endothelial cells of OV_GSE118828, compared to malignant cells of OV_GSE118828 dataset. The cell components and distribution in primary and metastatic ovarian cancer are quite distinct, which may lead to TME heterogeneity of ovarian cancer. Conclusion: Our results indicated that CD47 is closely correlated to ovarian cancer immune microenvironment and might induce ovarian cancer heterogeneity. Therefore, CD47 may be used as a candidate prognostic biomarker and provide us with new insights into potential immunotherapy in ovarian cancer patients.


CD47 Antigen/physiology , Ovarian Neoplasms/immunology , CD47 Antigen/analysis , Female , Humans , Ovarian Neoplasms/pathology , Prognosis , Tumor Microenvironment
2.
J Hematol Oncol ; 13(1): 96, 2020 07 16.
Article En | MEDLINE | ID: mdl-32677994

CD47, or integrin-associated protein, is a cell surface ligand expressed in low levels by nearly all cells of the body. It plays an integral role in various immune responses as well as autoimmunity, by sending a potent "don't eat me" signal to prevent phagocytosis. A growing body of evidence demonstrates that CD47 is overexpressed in various hematological malignancies and its interaction with SIRPα on the phagocytic cells prevents phagocytosis of cancer cells. Additionally, it is expressed by different cell types in the tumor microenvironment and is required for establishing tumor metastasis. Overexpression of CD47 is thus often associated with poor clinical outcomes. CD47 has emerged as a potential therapeutic target and is being investigated in various preclinical studies as well as clinical trials to prove its safety and efficacy in treating hematological neoplasms. This review focuses on different therapeutic mechanisms to target CD47, either alone or in combination with other cell surface markers, and its pivotal role in impairing tumor growth and metastatic spread of various types of hematological malignancies.


CD47 Antigen/physiology , Hematologic Neoplasms/physiopathology , Molecular Targeted Therapy , Neoplasm Proteins/physiology , Angiogenic Proteins/metabolism , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Differentiation/metabolism , Antineoplastic Agents, Immunological/therapeutic use , CD47 Antigen/antagonists & inhibitors , Clinical Trials as Topic , Drug Delivery Systems , Drug Design , Drug Screening Assays, Antitumor , Hematologic Neoplasms/therapy , Humans , Integrins/metabolism , Leukemia/metabolism , Leukemia/physiopathology , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/physiopathology , Molecular Mimicry , Myeloid Cells/metabolism , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Oligopeptides/therapeutic use , Protein Binding , Protein Domains , Protein Interaction Mapping , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Signal Transduction/physiology
3.
Theranostics ; 10(9): 4056-4072, 2020.
Article En | MEDLINE | ID: mdl-32226539

Rationale: CD47 plays a vital role in the immune escape of tumor cells, but its role in regulating immune-unrelated biological processes such as proliferation and metastasis remains unclear. We seek to explore the immune-independent functions of CD47 in colorectal cancer (CRC). Methods: The expression of CD47 in CRC was determined by immunohistochemistry. The biological effect of CD47 signaling on tumor cell proliferation and metastasis was evaluated in vitro and in vivo. RNA sequencing analysis was performed to identify pivotal signaling pathways modulated by CD47. The interaction between CD47 and ENO1 was verified by co-immunoprecipitation (co-IP). The effect of CD47 on glycolytic metabolites was analyzed by seahorse XF and targeted metabolomics. Results: The expression of CD47 was upregulated and correlated to poor prognosis in CRC patients. Functional assays revealed that CD47 promoted CRC cell growth and metastasis in vitro and in vivo. Our mechanistic investigations demonstrated that CD47 interacted with ENO1 and protected it from ubiquitin-mediated degradation, subsequently promoting glycolytic activity and phosphorylation of ERK in CRC cells. Inhibition of ENO1 diminished CD47-mediated cell growth and migration. Clinically, the combined expression of CD47 and ENO1 provided reliable predictive biomarkers for the prognosis of CRC patients. Conclusions: CD47 is overexpressed in CRC, and its expression is associated with poor prognosis. Through stabilizing ENO1, CD47 enhances the aerobic glycolysis and ERK activity in CRC cells, thereby promoting the progression of CRC. Our studies reveal an unconventional role of CD47, suggesting that targeting the CD47-ENO1 axis may provide a novel therapeutic avenue for CRC.


Biomarkers, Tumor/metabolism , CD47 Antigen/physiology , Colorectal Neoplasms , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Phosphopyruvate Hydratase/metabolism , Tumor Suppressor Proteins/metabolism , Aged , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Glycolysis , HEK293 Cells , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Metastasis , Severity of Illness Index
5.
Cell Oncol (Dordr) ; 43(1): 19-30, 2020 Feb.
Article En | MEDLINE | ID: mdl-31485984

BACKGROUND: CD47 is a widely expressed cellular receptor well known for its immunoregulatory functions. By interacting with its ligands, including thrombospondin-1 (TSP-1), signal regulatory protein α (SIRPα), integrins, and SH2-domain bearing protein tyrosine phosphatase substrate-1 (SHPS-1), it modulates cellular phagocytosis by macrophages, transmigration of neutrophils and activation of dendritic cells, T cells and B cells. Ample studies have shown that various types of cancer express high levels of CD47 to escape from the immune system. Based on this observation, CD47 is currently considered as a prominent target in cancer therapy. CONCLUSIONS: Here, we review the role of CD47 in the maintenance of immune system homeostasis. We also depict three emerging CD47-targeting strategies for cancer therapy, including the use of mimicry peptides, antibodies, and gene silencing strategies. Among these approaches, the most advanced one is the use of anti-CD47 antibodies, which enhances cancer cell phagocytosis via inhibition of the CD47-SIRPα axis. These antibodies can also achieve higher anti-cancer efficacies when combined with chemotherapy and immunotherapy and hold promise for improving the survival of patients with cancer.


B-Lymphocytes/immunology , CD47 Antigen/physiology , Dendritic Cells/immunology , Macrophages/immunology , Neoplasms/immunology , Phagocytosis/immunology , T-Lymphocytes/immunology , Animals , Antigens, Differentiation/metabolism , CD47 Antigen/metabolism , CD47 Antigen/therapeutic use , Dendritic Cells/metabolism , Humans , Integrins/metabolism , Macrophages/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Neutrophils/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
6.
Oxid Med Cell Longev ; 2019: 7121763, 2019.
Article En | MEDLINE | ID: mdl-31827695

In this study, we investigated whether CD47 deficiency attenuates isoproterenol- (ISO-) induced cardiac remodeling in mice. Cardiac remodeling was induced by intraperitoneal (i.p.) injection of ISO (60 mg·kg-1·d-1 in 100 µl of sterile normal saline) daily for 14 days and was confirmed by increased levels of lactate dehydrogenase (LDH) and creatine kinase MB (CK-MB), increased heart weight to body weight (HW/BW) ratios, and visible cardiac fibrosis. Apoptosis was evaluated by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining. Levels of malondialdehyde (MDA) and reactive oxygen species (ROS) were found to be significantly higher in the ISO group than in the control group, while superoxide dismutase (SOD) levels were suppressed in the ISO group. However, CD47 knockout significantly limited ISO-induced increases in LDH, CK-MB, and HW/BW ratios, cardiac fibrosis, oxidative stress, and apoptosis in the heart. In addition, CD47 deficiency also increased p-AMPK and LAMP2 expression and decreased HDAC3, cleaved Caspase-3, cleaved Caspase-9, LC3II, and p62 expression in cardiac tissues. In conclusion, CD47 deficiency reduced i.p. ISO-induced cardiac remodeling probably by inhibiting the HDAC3 pathway, improving AMPK signaling and autophagy flux, and rescuing autophagic clearance.


CD47 Antigen/physiology , Cardiomegaly/prevention & control , Cardiotonic Agents/toxicity , Isoproterenol/toxicity , Ventricular Remodeling/physiology , Animals , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Ventricular Remodeling/drug effects
7.
Pharmacol Ther ; 188: 12-25, 2018 08.
Article En | MEDLINE | ID: mdl-29444453

Atherosclerosis is a dynamic and progressive inflammatory process in the intimal layer of large and medium-sized arteries, and it is the major contributor to the atherosclerotic cardiovascular disease (ACVD), the leading cause of death worldwide. In an atherosclerotic plaque, phagocytosis of apoptotic cells occurs through an intricate process designated efferocytosis. Defective efferocytosis has emerged as a causal factor in the etiopathogenesis of atherosclerosis and its progression into overt ACVD. Both specialized phagocytes (macrophages and dendritic cells) and non-specialized cells with phagocytic capabilities (smooth muscle and endothelial cells) are involved in the efferocytotic process. Moreover, several signaling and regulatory molecules are involved in the different steps of efferocytosis, and they include "Find-Me" signals (lysophosphatidylcholine), "Eat-Me" signals [phosphatidylserine, Mer tyrosine kinase (MerTK), and milk fat globule-EGF factor 8], and "Don't Eat-Me" signals [cluster of differentiation 47 (CD47)]. Regulation of efferocytosis is in a close nexus with inflammation, the key component in atherosclerosis. The predominance of pro-inflammatory and anti-inflammatory molecules plays a crucial role in lesion progression and regression, respectively. Polarization of macrophages towards the M1 phenotype causes them to secrete proinflammatory cytokines, while polarization towards the M2 phenotype causes them to secrete of anti-inflammatory cytokines, including interleukin-10 and transforming growth factor ß, so tending to shift the balance towards resolution of the inflammation. Dysfunction of any regulatory signal may cause expansion of the necrotic core of an atherosclerotic plaque with ensuing conversion of the plaque into an unstable plaque with an increased susceptibility to rupture and to atherothrombotic complication. In this review we aim at elucidating the determinant factors and pathways of efferocytosis which can be considered as potential novel targets when striving to develop more personalized and efficient treatment regimens for patients with ACVD.


Atherosclerosis/etiology , Phagocytosis/physiology , Antigens, Surface/physiology , Apoptosis , CD47 Antigen/physiology , Calreticulin/physiology , Humans , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Macrophages/physiology , Milk Proteins , c-Mer Tyrosine Kinase/physiology
8.
Mol Cell Biol ; 38(9)2018 05 01.
Article En | MEDLINE | ID: mdl-29463646

A gain-of-function mutation in isocitrate dehydrogenase 1 (IDH1) affects immune surveillance in gliomas. As elevated CD47 levels are associated with immune evasion in cancers, its status in gliomas harboring mutant IDH1 (IDH1-MT cells) was investigated. Decreased CD47 expression in IDH1-R132H-overexpressing cells was accompanied by diminished nuclear ß-catenin, pyruvate kinase isoform M2 (PKM2), and TCF4 levels compared to those in cells harboring wild-type IDH1 (IDH1-WT cells). The inhibition of ß-catenin in IDH1-WT cells abrogated CD47 expression, ß-catenin-TCF4 interaction, and the transactivational activity of ß-catenin/TCF4. The reverse effect was observed in IDH1-MT cells upon the pharmacological elevation of nuclear ß-catenin levels. Genetic and pharmacological manipulation of nuclear PKM2 levels in IDH1-WT and IDH1-MT cells suggested that PKM2 is a positive regulator of the ß-catenin-TCF4 interaction. The Cancer Genome Atlas (TCGA) data sets indicated diminished CD47, PKM2, and ß-catenin levels in IDH1-MT gliomas compared to IDH1-WT gliomas. Also, elevated BRG1 levels with mutations in the ATP-dependent chromatin-remodeling site were observed in IDH1-MT glioma. The ectopic expression of ATPase-deficient BRG1 diminished CD47 expression as well as TCF4 occupancy on its promoter. Sequential chromatin immunoprecipitation (ChIP-re-ChIP) revealed the recruitment of the PKM2-ß-catenin-BRG1-TCF4 complex to the TCF4 site on the CD47 promoter. This occupancy translated into CD47 transcription, as a diminished recruitment of this complex was observed in glioma cells bearing IDH1-R132H. In addition to its involvement in CD47 transcriptional regulation, PKM2-ß-catenin-BRG1 cross talk affected the phagocytosis of IDH1-MT cells by microglia.


CD47 Antigen/physiology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/physiology , CD47 Antigen/genetics , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Line, Tumor , DNA Helicases/genetics , DNA Helicases/physiology , DNA Methylation , Gain of Function Mutation/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Glioma/genetics , Glioma/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/physiology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Promoter Regions, Genetic/genetics , Thyroid Hormones/genetics , Thyroid Hormones/physiology , Transcription Factors/genetics , Transcription Factors/physiology , beta Catenin/genetics , beta Catenin/physiology , Thyroid Hormone-Binding Proteins
9.
Transplantation ; 102(3): 406-416, 2018 03.
Article En | MEDLINE | ID: mdl-28968355

BACKGROUND: Macrophages play important roles in xenograft rejection. Here, we investigated whether overexpression of human CD200 or CD47 in porcine endothelial cells (PEC) can suppress macrophages activation in xenogeneic immune responses. METHODS: PECs and human macrophages were incubated together, harvested, and analyzed for in vitro macrophage phagocytic and cytotoxicity activity, and cytokine release. Next, PECs were injected into renal subcapsular space of humanized mice. On day 10 posttransplantation, we analyzed xenograft survival and perigraft inflammatory cell infiltrations in PEC-to-humanized mouse transplantation. RESULTS: PECs highly expressing human CD200, CD47, or both CD47/CD200 were established by lentiviral vector transduction. Both CD200 and CD47 suppressed in vitro macrophage phagocytic and cytotoxic activity against PECs; decreased TNF-α, IL-1ß, and IL-6 secretion; and increased IL-10 secretion. However, simultaneous overexpression of CD200 and CD47 did not show additive effects. Next, PECs were transplanted into NOD-scid IL-2Rg null mice, and human monocytes and lymphocytes were adoptively transferred 1 day after xenotransplantation. PEC xenograft cell death and apoptosis were decreased in the CD200-PEC and CD47/CD200-PEC groups. Perigraft infiltration of human T cells was suppressed by CD47; CD200 suppressed infiltration of human macrophages to a greater extent than CD47; and the CD47/CD200-PEC group exhibited the lowest level of leukocyte infiltration. In summary, overexpression of CD200 in PECs suppressed xenogeneic activation of human macrophages and improved survival of PEC xenografts in humanized mice; however, coexpression of CD200 and CD47 did not show additive effects. CONCLUSIONS: Therefore, overexpression of human CD200 in donor pigs could constitute a promising strategy for overcoming xenograft rejection.


Antigens, CD/physiology , CD47 Antigen/physiology , Macrophage Activation , Transplantation, Heterologous , Animals , Cytokines/biosynthesis , Graft Survival , Humans , Mice , Swine
10.
Biochim Biophys Acta Rev Cancer ; 1868(2): 571-583, 2017 Dec.
Article En | MEDLINE | ID: mdl-29056539

Inhibitory or stimulatory immune checkpoint molecules are expressed on a sizeable fraction of tumor cells in different tumor types. It was thought that the main function of tumor cell-associated immune checkpoint molecules would be the modulation (down- or upregulation) of antitumor immune responses. In recent years, however, it has become clear that the expression of immune checkpoint molecules on tumor cells has important consequences on the biology of the tumor cells themselves. In particular, a causal relationship between the expression of these molecules and the acquisition of malignant traits has been demonstrated. Thus, immune checkpoint molecules have been shown to promote the epithelial-mesenchymal transition of tumor cells, the acquisition of tumor-initiating potential and resistance to apoptosis and antitumor drugs, as well as the propensity to disseminate and metastasize. Herein, we review this evidence, with a main focus on PD-L1, the most intensively investigated tumor cell-associated immune checkpoint molecule and for which most information is available. Then, we discuss more concisely other tumor cell-associated immune checkpoint molecules that have also been shown to induce the acquisition of malignant traits, such as PD-1, B7-H3, B7-H4, Tim-3, CD70, CD28, CD137, CD40 and CD47. Open questions in this field as well as some therapeutic approaches that can be derived from this knowledge, are also addressed.


B7-H1 Antigen/physiology , Neoplasms/etiology , Animals , B7 Antigens/physiology , CD47 Antigen/physiology , Epithelial-Mesenchymal Transition , Humans , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/physiology , Programmed Cell Death 1 Receptor/physiology , Proto-Oncogene Proteins c-akt/physiology , TOR Serine-Threonine Kinases/physiology , Tumor Microenvironment , V-Set Domain-Containing T-Cell Activation Inhibitor 1/physiology
11.
Nat Commun ; 8: 14802, 2017 04 05.
Article En | MEDLINE | ID: mdl-28378740

CD47 is a cell surface molecule that inhibits phagocytosis of cells that express it by binding to its receptor, SIRPα, on macrophages and other immune cells. CD47 is expressed at different levels by neoplastic and normal cells. Here, to reveal mechanisms by which different neoplastic cells generate this dominant 'don't eat me' signal, we analyse the CD47 regulatory genomic landscape. We identify two distinct super-enhancers (SEs) associated with CD47 in certain cancer cell types. We show that a set of active constituent enhancers, located within the two CD47 SEs, regulate CD47 expression in different cancer cell types and that disruption of CD47 SEs reduces CD47 gene expression. Finally we report that the TNF-NFKB1 signalling pathway directly regulates CD47 by interacting with a constituent enhancer located within a CD47-associated SE specific to breast cancer. These results suggest that cancers can evolve SE to drive CD47 overexpression to escape immune surveillance.


Breast Neoplasms/metabolism , CD47 Antigen/physiology , Enhancer Elements, Genetic , Inflammation Mediators/metabolism , Inflammation/metabolism , Signal Transduction , Up-Regulation , Animals , Breast Neoplasms/pathology , CD47 Antigen/genetics , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , NF-kappa B p50 Subunit/metabolism , Phagocytosis , Protein Binding , Tumor Necrosis Factor-alpha/metabolism
12.
Article En | MEDLINE | ID: mdl-28018860

In the last decade it has become clear that, similarly to nucleated cells, enucleated red blood cells (RBCs) are susceptible to programmed apoptotic cell death. Erythrocytic apoptosis seems to play a role in physiological clearance of aged RBCs, but it may also be implicated in anemia of different etiological sources including drug therapy and infectious diseases. In malaria, severe anemia is a common complication leading to death of children and pregnant women living in malaria-endemic regions of Africa. The pathogenesis of malarial anemia is multifactorial and involves both ineffective production of RBCs by the bone marrow and premature elimination of non-parasitized RBCs, phenomena potentially associated with apoptosis. In the present overview, we discuss evidences associating erythrocytic apoptosis with the pathogenesis of severe malarial anemia, as well as with regulation of parasite clearance in malaria. Efforts to understand the role of erythrocytic apoptosis in malarial anemia can help to identify potential targets for therapeutic intervention based on apoptotic pathways and consequently, mitigate the harmful impact of malaria in global public health.


Anemia/etiology , Anemia/parasitology , Apoptosis , Erythrocytes/parasitology , Malaria/complications , Africa , Anemia/mortality , Bone Marrow , CD47 Antigen/physiology , Child , Erythropoiesis , Female , Humans , Malaria, Falciparum/complications , Phagocytosis , Pregnancy
13.
Cancer Res ; 76(19): 5657-5670, 2016 10 01.
Article En | MEDLINE | ID: mdl-27698188

The unfolded protein response is an endoplasmic reticulum stress pathway mediated by the protein chaperone glucose regulated-protein 78 (GRP78). Metabolic analysis of breast cancer cells shows that GRP78 silencing increases the intracellular concentrations of essential polyunsaturated fats, including linoleic acid. Accumulation of fatty acids is due to an inhibition of mitochondrial fatty acid transport, resulting in a reduction of fatty acid oxidation. These data suggest a novel role of GRP78-mediating cellular metabolism. We validated the effect of GRP78-regulated metabolite changes by treating tumor-bearing mice with tamoxifen and/or linoleic acid. Tumors treated with linoleic acid plus tamoxifen exhibited reduced tumor area and tumor weight. Inhibition of either GRP78 or linoleic acid treatment increased MCP-1 serum levels, decreased CD47 expression, and increased macrophage infiltration, suggesting a novel role for GRP78 in regulating innate immunity. GRP78 control of fatty acid oxidation may represent a new homeostatic function for GRP78. Cancer Res; 76(19); 5657-70. ©2016 AACR.


Breast Neoplasms/metabolism , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/physiology , Lipid Metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , CD47 Antigen/physiology , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Fatty Acids/metabolism , Female , Humans , Immunity, Innate , Metabolomics , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism , Unfolded Protein Response
14.
J Virol ; 90(15): 6746-6758, 2016 08 01.
Article En | MEDLINE | ID: mdl-27194758

UNLABELLED: An integrin-associated protein CD47, which is a ligand for the inhibitory receptor signal regulatory protein α, is expressed on B and T cells, as well as on most innate immune cells. However, the roles of CD47 in the immune responses to viral infection or vaccination remain unknown. We investigated the role of CD47 in inducing humoral immune responses after intranasal infection with virus or immunization with influenza virus-like particles (VLPs). Virus infection or vaccination with VLPs containing hemagglutinin from A/PR8/34 influenza virus induced higher levels of antigen-specific IgG2c isotype dominant antibodies in CD47-deficient (CD47KO) mice than in wild-type (WT) mice. CD47KO mice with vaccination showed greater protective efficacy against lethal challenge, as evidenced by no loss in body weight and reduced lung viral titers compared to WT mice. In addition, inflammatory responses which include cytokine production, leukocyte infiltrates, and gamma interferon-producing CD4(+) T cells, as well as an anti-inflammatory cytokine (interleukin-10), were reduced in the lungs of vaccinated CD47KO mice after challenge with influenza virus. Analysis of lymphocytes indicated that GL7(+) germinal center B cells were induced at higher levels in the draining lymph nodes of CD47KO mice compared to those in WT mice. Notably, CD47KO mice exhibited significant increases in the numbers of antigen-specific memory B cells in spleens and plasma cells in bone marrow despite their lower levels of background IgG antibodies. These results suggest that CD47 plays a role as a negative regulator in inducing protective immune responses to influenza vaccination. IMPORTANCE: Molecular mechanisms that control B cell activation to produce protective antibodies upon viral vaccination remain poorly understood. The CD47 molecule is known to be a ligand for the inhibitory receptor signal regulatory protein α and expressed on the surfaces of most immune cell types. CD47 was previously demonstrated to play an important role in modulating the migration of monocytes, neutrophils, polymorphonuclear neutrophils, and dendritic cells into the inflamed tissues. The results of this study demonstrate new roles of CD47 in negatively regulating the induction of protective IgG antibodies, germinal center B cells, and plasma cells secreting antigen-specific antibodies, as well as macrophages, upon influenza vaccination and challenge. As a consequence, vaccinated CD47-deficient mice demonstrated better control of influenza viral infection and enhanced protection. This study provides insights into understanding the regulatory functions of CD47 in inducing adaptive immunity to vaccination.


Adaptive Immunity/immunology , CD47 Antigen/physiology , Immunity, Humoral/immunology , Influenza Vaccines/administration & dosage , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae/immunology , Vaccination/methods , Animals , Antibodies, Viral/blood , Cells, Cultured , Immunization , Immunologic Memory/immunology , Interleukin-10/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology
15.
Philos Trans R Soc Lond B Biol Sci ; 370(1680): 20140364, 2015 Oct 19.
Article En | MEDLINE | ID: mdl-26416675

The appearance of stem cells coincides with the transition from single-celled organisms to metazoans. Stem cells are capable of self-renewal as well as differentiation. Each tissue is maintained by self-renewing tissue-specific stem cells. The accumulation of mutations that lead to preleukaemia are in the blood-forming stem cell, while the transition to leukaemia stem cells occurs in the clone at a progenitor stage. All leukaemia and cancer cells escape being removed by scavenger macrophages by expressing the 'don't eat me' signal CD47. Blocking antibodies to CD47 are therapeutics for all cancers, and are currently being tested in clinical trials in the US and UK.


Biological Evolution , Neoplastic Stem Cells/pathology , Stem Cells/cytology , Animals , CD47 Antigen/physiology , Cell Differentiation , Cell Movement , Humans , Mutation , Neoplasm Metastasis , Neoplastic Stem Cells/physiology , Stem Cells/physiology
16.
Proc Natl Acad Sci U S A ; 112(10): 3062-7, 2015 Mar 10.
Article En | MEDLINE | ID: mdl-25713361

Several Plasmodium species exhibit a strong age-based preference for the red blood cells (RBC) they infect, which in turn is a major determinant of disease severity and pathogenesis. The molecular basis underlying this age constraint on the use of RBC and its influence on parasite burden is poorly understood. CD47 is a marker of self on most cells, including RBC, which, in conjunction with signal regulatory protein alpha (expressed on macrophages), prevents the clearance of cells by the immune system. In this report, we have investigated the role of CD47 on the growth and survival of nonlethal Plasmodium yoelii 17XNL (PyNL) malaria in C57BL/6 mice. By using a quantitative biotin-labeling procedure and a GFP-expressing parasite, we demonstrate that PyNL parasites preferentially infect high levels of CD47 (CD47(hi))-expressing young RBC. Importantly, C57BL/6 CD47(-/-) mice were highly resistant to PyNL infection and developed a 9.3-fold lower peak parasitemia than their wild-type (WT) counterparts. The enhanced resistance to malaria observed in CD47(-/-) mice was associated with a higher percentage of splenic F4/80(+) cells, and these cells had a higher percentage of phagocytized parasitized RBC than infected WT mice during the acute phase of infection, when parasitemia was rapidly rising. Furthermore, injection of CD47-neutralizing antibody caused a significant reduction in parasite burden in WT C57BL/6 mice. Together, these results strongly suggest that CD47(hi) young RBC may provide a shield to the malaria parasite from clearance by the phagocytic cells, which may be an immune escape mechanism used by Plasmodium parasites that preferentially infect young RBC.


CD47 Antigen/physiology , Phagocytosis/immunology , Plasmodium yoelii/physiology , Animals , Green Fluorescent Proteins/genetics , Host-Parasite Interactions , Macrophages/immunology , Mice, Inbred C57BL , Plasmodium yoelii/immunology
17.
Anticancer Res ; 35(1): 149-57, 2015 Jan.
Article En | MEDLINE | ID: mdl-25550546

CD47, an integrin-associated protein is over-expressed in several tumors including glioblastomas. Activation of CD47 induces proliferation of human astrocytoma cells but not normal astrocytes via an Akt-dependent way. However, the pathways mediating this process are still unknown. The epigenetic integrator UHRF1 (Ubiquitin-like containing PHD and RING Finger 1) is over-expressed in various cancers and plays a vital role in the silencing of numerous tumor suppressor genes including p16(INK4A), thereby promoting cell proliferation. The aim of the present study was to investigate the role of UHRF1 and p16(INK4A) in CD47-induced effects. Herein we showed that activation of CD47 in human astrocytoma cell lines U87 and CCF- STTG1 (Grade IV), up-regulated the expression of UHRF1 with subsequent down-regulation of p16(INK4A), thus promoting cell proliferation. Blockage of CD47 using a blocking antibody down-regulated UHRF1 expression, accompanied by a re-expression of p16(INK4A), conducting to decreased cell proliferation in both cancer cell lines. Neither CD47 activation nor its blocking has any effect on UHRF1/p16(INK4A) expression in normal human astrocytes. Depletion of CD47 in the U87 cell line resulted in down-regulation of UHRF1. We also found that CD47 activated the inflammatory genes IL-6, IL-7 and MCP-1 by a NF-κB-dependent mechanism in human astrocytoma but not in normal astrocytes. In conclusion, the present findings indicate that CD47 activation increases expression of UHRF1 and suggest, for the first time, that CD47 regulates the epigenetic code by targeting UHRF1. This could represent a new pathway towards cell proliferation and metastasis.


CCAAT-Enhancer-Binding Proteins/metabolism , CD47 Antigen/physiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Neoplastic , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation , Gene Silencing , Glioblastoma , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-7/genetics , Interleukin-7/metabolism , NF-kappa B/physiology , Transcriptional Activation , Ubiquitin-Protein Ligases
18.
Transplantation ; 98(4): 394-401, 2014 Aug 27.
Article En | MEDLINE | ID: mdl-24983310

BACKGROUND: Ischemia-reperfusion injury (IRI) significantly contributes to delayed graft function and inflammation, leading to graft loss. Ischemia-reperfusion injury is exacerbated by the thrombospondin-1-CD47 system through inhibition of nitric oxide signaling. We postulate that CD47 blockade and prevention of nitric oxide inhibition reduce IRI in organ transplantation. METHODS: We used a syngeneic rat renal transplantation model of IRI with bilaterally nephrectomized recipients to evaluate the effect of a CD47 monoclonal antibody (CD47mAb) on IRI. Donor kidneys were flushed with CD47mAb OX101 or an isotype-matched control immunoglobulin and stored at 4°C in University of Wisconsin solution for 6 hr before transplantation. RESULTS: CD47mAb perfusion of donor kidneys resulted in marked improvement in posttransplant survival, lower levels of serum creatinine, blood urea nitrogen, phosphorus and magnesium, and less histological evidence of injury. In contrast, control groups did not survive more than 5 days, had increased biochemical indicators of renal injury, and exhibited severe pathological injury with tubular atrophy and necrosis. Recipients of CD47mAb-treated kidneys showed decreased levels of plasma biomarkers of renal injury including Cystatin C, Osteopontin, Tissue Inhibitor of Metalloproteinases-1 (TIMP1), ß2-Microglobulin, Vascular Endothelial Growth Factor A (VEGF-A), and clusterin compared to the control group. Furthermore, laser Doppler assessment showed higher renal blood flow in the CD47mAb-treated kidneys. CONCLUSION: These results provide strong evidence for the use of CD47 antibody-mediated blockade to reduce IRI and improve organ preservation for renal transplantation.


CD47 Antigen/physiology , Kidney Transplantation , Kidney/blood supply , Reperfusion Injury/prevention & control , Animals , Antibodies, Monoclonal/therapeutic use , CD47 Antigen/immunology , Graft Survival , Kidney/pathology , Male , Rats , Rats, Inbred Lew , Treatment Outcome
19.
J Am Heart Assoc ; 3(3): e000670, 2014 Jun 10.
Article En | MEDLINE | ID: mdl-24922625

BACKGROUND: Left ventricular heart failure (LVHF) remains progressive and fatal and is a formidable health problem because ever-larger numbers of people are diagnosed with this disease. Therapeutics, while relieving symptoms and extending life in some cases, cannot resolve this process and transplant remains the option of last resort for many. Our team has described a widely expressed cell surface receptor (CD47) that is activated by its high-affinity secreted ligand, thrombospondin 1 (TSP1), in acute injury and chronic disease; however, a role for activated CD47 in LVHF has not previously been proposed. METHODS AND RESULTS: In experimental LVHF TSP1-CD47 signaling is increased concurrent with up-regulation of cardiac histone deacetylase 3 (HDAC3). Mice mutated to lack CD47 displayed protection from transverse aortic constriction (TAC)-driven LVHF with enhanced cardiac function, decreased cellular hypertrophy and fibrosis, decreased maladaptive autophagy, and decreased expression of HDAC3. In cell culture, treatment of cardiac myocyte CD47 with a TSP1-derived peptide, which binds and activates CD47, increased HDAC3 expression and myocyte hypertrophy in a Ca(2+)/calmodulin protein kinase II (CaMKII)-dependent manner. Conversely, antibody blocking of CD47 activation, or pharmacologic inhibition of CaMKII, suppressed HDAC3 expression, decreased myocyte hypertrophy, and mitigated established LVHF. Downstream gene suppression of HDAC3 mimicked the protective effects of CD47 blockade and decreased hypertrophy in myocytes and mitigated LVHF in animals. CONCLUSIONS: These data identify a proximate role for the TSP1-CD47 axis in promoting LVHF by CaKMII-mediated up-regulation of HDAC3 and suggest novel therapeutic opportunities.


CD47 Antigen/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Heart Failure/physiopathology , Histone Deacetylases/biosynthesis , Animals , Cells, Cultured , Enzyme Induction/physiology , Heart Failure/etiology , Histone Deacetylases/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Myocytes, Cardiac/physiology , Rats, Sprague-Dawley , Signal Transduction/physiology
20.
Scand J Immunol ; 80(1): 22-35, 2014 Jul.
Article En | MEDLINE | ID: mdl-24684700

CD172α and CD47 are members of glycoprotein expressed on macrophages and various immune cells, promote immune recognition and T cell stimulation that priming phagocytosis of pathogens and apoptotic bodies and malignant cell. Tumour-releasing immunosuppressive factor promotes tumour growth and transforms the tumour resident M1 phenotype of macrophage to M2 phenotype (TAMs) that promotes tumour progression by downregulating the expression of different surface receptor including CD172α and CD47. Recent studies have reported that CD172α and CD47 are involved in the pathogenesis and promote malignancies such as lymphoma, leukaemia, melanoma, lung cancer and multiple myeloma, and their expression varies during infection and malignancies. Autologous Hsp70 is well recognized for its role in activating macrophages leading to enhance production of inflammatory cytokines. It has been observed that Hsp70 derived from normal tissues do not elicit tumour immunity, while Hsp70 preparation from tumour cell was able to elicit tumour immunity. However, the role of exogenous autologous hsp70 on the formation of giant cells is completely unknown. Therefore, in the present study, we sought to investigate the effect of Hsp70-peptide complex on the expression of CD172α and CD47 receptors in normal peritoneal macrophages (NMO) and TAMs. Finding shows that the expression of CD172α and CD47 enhances in TAMs and it reverts back the suppressed function of TAMs into M1 state of immunoregulatory phenotype that promotes tumour regression by enhanced multinucleation and phagocytosis of malignant cells and significantly enhances the homotypic fusion of macrophages and polykaryon formation in vitro by enhancing the expression of SIRPα and IAP.


CD47 Antigen/physiology , HSP70 Heat-Shock Proteins/physiology , Macrophages, Peritoneal/physiology , Receptors, Immunologic/physiology , Animals , CD47 Antigen/analysis , CD47 Antigen/genetics , Female , Flow Cytometry , Fluorescent Antibody Technique , Lymphoma , Male , Mice , Mice, Inbred BALB C , Phenotype , Protein Structure, Tertiary , Receptors, Immunologic/analysis , Receptors, Immunologic/genetics
...