Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 19(3): 835-846, 2020 03.
Article in English | MEDLINE | ID: mdl-31879362

ABSTRACT

Inhibitors of adaptive immune checkpoints have shown promise as cancer treatments. CD47 is an innate immune checkpoint receptor broadly expressed on normal tissues and overexpressed on many tumors. Binding of tumor CD47 to signal regulatory protein alpha (SIRPα) on macrophages and dendritic cells triggers a "don't eat me" signal that inhibits phagocytosis enabling escape of innate immune surveillance. Blocking CD47/SIRPα interaction promotes phagocytosis reducing tumor burden in numerous xenograft and syngeneic animal models. We have developed a next-generation humanized anti-CD47 antibody, AO-176, that not only blocks the CD47/SIRPα interaction to induce tumor cell phagocytosis, but also induces tumor cytotoxicity in hematologic and solid human tumor cell lines, but not normal noncancerous cells, by a cell autonomous mechanism (not ADCC). AO-176 also binds preferentially to tumor versus many normal cell types. In particular, AO-176 binds negligibly to RBCs in contrast to tumor cells, even at high concentrations up to 200 µg/mL and does not agglutinate RBCs up to 1 mg/mL in vitro These properties are expected not only to decrease the antigen sink, but also to minimize on-target clinical adverse effects observed following treatment with other reported RBC-binding anti-CD47 antibodies. When tested in cynomolgus monkeys, AO-176 was well tolerated with no adverse effects. Finally, we show that AO-176 demonstrates dose-dependent antitumor activity in tumor xenograft models. Taken together, the unique properties and antitumor activity of our next-generation anti-CD47 antibody, AO-176, distinguishes it from other CD47/SIRPα axis targeting agents in clinical development.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , CD47 Antigen/antagonists & inhibitors , Erythrocytes/metabolism , Immunity, Innate/immunology , Neoplasms/drug therapy , Phagocytosis , Receptors, Immunologic/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antigens, Differentiation/immunology , Apoptosis , CD47 Antigen/immunology , Cell Proliferation , Female , Humans , Immunity, Innate/drug effects , Macaca fascicularis , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/immunology , Neoplasms/pathology , Receptors, Immunologic/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Transplantation ; 100(7): 1480-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27331362

ABSTRACT

BACKGROUND: Despite the efficacy of orthotopic liver transplantation in the treatment of end-stage liver diseases, its therapeutic utility is severely limited by the availability of donor organs. The ability to rehabilitate marginal organs, such as steatotic allografts, has the potential to address some of the supply limitations of available organs for transplantation. Steatotic livers are more susceptible to ischemia-reperfusion injury (IRI), which is exacerbated by the thrombospondin-1/CD47 pathway through inhibition of nitric oxide signaling. We postulated that CD47 blockade with a monoclonal antibody specific to CD47, clone 400 (CD47mAb400) may reduce the extent of IRI in steatotic liver allografts. METHODS: Orthotopic liver transplantation was performed using steatotic liver grafts from Zucker rats transplanted into lean recipients. Control IgG or the CD47mAb400 was administered to the donor livers at procurement. Serum transaminases, histological changes, and animal survival were assessed. Hepatocellular damage, oxidative and nitrosative stress, and inflammation were also quantified. RESULTS: Administration of CD47mAb400 to donor livers increased recipient survival and resulted in significant reduction of serum transaminases, bilirubin, triphosphate nick-end labeling staining, caspase-3 activity, oxidative and nitrosative stresses, and proinflammatory cytokine expression of TNF-α, IL-6 and IL-1ß. CONCLUSIONS: We conclude that administration of CD47mAb400 to donor grafts may reduce IRI through CD47 blockade to result in improved function of steatotic liver allografts and increased survival of recipients and represent a novel strategy to allow the use of livers with higher levels of steatosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD47 Antigen/immunology , Fatty Liver/immunology , Liver Transplantation , Reperfusion Injury/drug therapy , Allografts , Animals , Cell Movement , Enzyme-Linked Immunosorbent Assay , Fatty Liver/surgery , Immunoglobulin G/immunology , Inflammation , Liver/pathology , Liver/surgery , Nitric Oxide/metabolism , Oxidative Stress , Rats , Rats, Zucker , Reactive Oxygen Species/metabolism , Reperfusion Injury/pathology , Signal Transduction , Transaminases/blood , Treatment Outcome
3.
Nat Med ; 21(10): 1209-15, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26322579

ABSTRACT

Macrophage phagocytosis of tumor cells mediated by CD47-specific blocking antibodies has been proposed to be the major effector mechanism in xenograft models. Here, using syngeneic immunocompetent mouse tumor models, we reveal that the therapeutic effects of CD47 blockade depend on dendritic cell but not macrophage cross-priming of T cell responses. The therapeutic effects of anti-CD47 antibody therapy were abrogated in T cell-deficient mice. In addition, the antitumor effects of CD47 blockade required expression of the cytosolic DNA sensor STING, but neither MyD88 nor TRIF, in CD11c+ cells, suggesting that cytosolic sensing of DNA from tumor cells is enhanced by anti-CD47 treatment, further bridging the innate and adaptive responses. Notably, the timing of administration of standard chemotherapy markedly impacted the induction of antitumor T cell responses by CD47 blockade. Together, our findings indicate that CD47 blockade drives T cell-mediated elimination of immunogenic tumors.


Subject(s)
CD47 Antigen/immunology , Immunotherapy , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Antineoplastic Agents/therapeutic use , DNA/physiology , Humans , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Xenograft Model Antitumor Assays
4.
PLoS One ; 10(6): e0129667, 2015.
Article in English | MEDLINE | ID: mdl-26102349

ABSTRACT

BNIP3 is a dual function protein, able to activate autophagy and induce cell death. Upon expression of BNIP3, which is upregulated by hypoxia, the protein induces mitochondrial dysfunction, often leading to cell death. However, some highly respiring cells and cancer cells tolerate BNIP3 expression, suggesting that a yet unknown mechanism exists to restrain the lethal effects of BNIP3 on mitochondria. Here we present evidence that BNIP3 undergoes several phosphorylation events at its C-terminus, adjacent to the transmembrane domain. Phosphorylation at these residues inhibits BNIP3-induced mitochondrial damage, preventing a loss of mitochondrial mass and mitochondrial membrane potential, as well as preventing an increase in reactive oxygen species. This decrease in mitochondrial damage, as well as the reduction of cell death upon C-terminal BNIP3 phosphorylation, can be explained by a diminished interaction between BNIP3 and OPA1, a key regulator of mitochondrial fusion and mitochondrial inner membrane structure. Importantly, phosphorylation of these C-terminal BNIP3 residues blocks cell death without preventing autophagy, providing evidence that the two functional roles of BNIP3 can be regulated independently. These findings establish phosphorylation as a switch to determine the pro-survival and pro-death effects of the protein. Our findings also suggest a novel target for the regulation of these activities in transformed cells where BNIP3 is often highly expressed.


Subject(s)
Autophagy , Cell Death , Membrane Proteins/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , HEK293 Cells , Humans , Membrane Proteins/chemistry , Molecular Sequence Data , Phosphorylation , Proto-Oncogene Proteins/chemistry
5.
Cancer Lett ; 360(2): 302-9, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25721088

ABSTRACT

Human hepatocellular carcinoma (HCC) has a high rate of tumor recurrence and metastasis, resulting in shortened survival times. The efficacy of current systemic therapies for HCC is limited. In this study, we used xenograft tumor models to investigate the use of antibodies that block CD47 and inhibit HCC tumor growth. Immunostaining of tumor tissue and HCC cell lines demonstrated CD47 over-expression in HCC as compared to normal hepatocytes. Macrophage phagocytosis of HCC cells was increased after treatment with CD47 antibodies (CD47mAbs) that block CD47 binding to SIRPα. Further, CD47 blockade inhibited tumor growth in both heterotopic and orthotopic models of HCC, and promoted the migration of macrophages into the tumor mass. Our results demonstrate that targeting CD47 by specific antibodies has potential immunotherapeutic efficacy in human HCC.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD47 Antigen/immunology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , CD47 Antigen/biosynthesis , Carcinoma, Hepatocellular/immunology , Cell Movement/immunology , Hep G2 Cells , Humans , Liver Neoplasms/immunology , Macrophages/immunology , Male , Mice , Mice, SCID , Phagocytosis/immunology , Xenograft Model Antitumor Assays
6.
Liver Transpl ; 21(4): 468-477, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25482981

ABSTRACT

Orthotopic liver transplantation (OLT) remains the standard treatment option for nonresponsive liver failure. Because ischemia/reperfusion injury (IRI) is an important impediment to the success of OLT, new therapeutic strategies are needed to reduce IRI. We investigated whether blocking the CD47/thrombospondin-1 inhibitory action on nitric oxide signaling with a monoclonal antibody specific to CD47 (CD47mAb400) would reduce IRI in liver grafts. Syngeneic OLT was performed with Lewis rats. Control immunoglobulin G or CD47mAb400 was administered to the donor organ at procurement or to both the organ and the recipient at the time of transplant. Serum transaminases, histological changes of the liver, and animal survival were assessed. Oxidative stress, inflammatory responses, and hepatocellular damage were also quantified. A significant survival benefit was not achieved when CD47mAb400 was administered to the donor alone. However, CD47mAb400 administration to both the donor and the recipient increased animal survival afterward. The CD47mAb400-treated group showed lower serum transaminases, bilirubin, oxidative stress, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling staining, caspase-3 activity, and proinflammatory cytokine expression of tumor necrosis factor α, interleukin-1ß, and interleukin-6. Thus, CD47 blockade with CD47mAb400 administered both to the donor and the recipient reduced liver graft IRI in a rat liver transplantation model. This may translate to decreased liver dysfunction and increased survival of liver transplant recipients.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD47 Antigen/metabolism , Cold Ischemia/adverse effects , Liver Transplantation/adverse effects , Liver/drug effects , Liver/surgery , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Biomarkers/blood , CD47 Antigen/immunology , Cytoprotection , Disease Models, Animal , Inflammation Mediators/blood , Liver/blood supply , Liver/immunology , Liver/metabolism , Liver/pathology , Male , Necrosis , Oxidative Stress/drug effects , Rats, Inbred Lew , Reperfusion Injury/blood , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects , Time Factors
7.
Transplantation ; 98(4): 394-401, 2014 Aug 27.
Article in English | MEDLINE | ID: mdl-24983310

ABSTRACT

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.


Subject(s)
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
8.
Mol Biol Cell ; 24(21): 3358-68, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24006483

ABSTRACT

CD47 plays an important but incompletely understood role in the innate and adaptive immune responses. CD47, also called integrin-associated protein, has been demonstrated to associate in cis with ß1 and ß3 integrins. Here we test the hypothesis that CD47 regulates adhesive functions of T-cell α4ß1 (VLA-4) and αLß2 (LFA-1) in in vivo and in vitro models of inflammation. Intravital microscopy studies reveal that CD47(-/-) Th1 cells exhibit reduced interactions with wild-type (WT) inflamed cremaster muscle microvessels. Similarly, murine CD47(-/-) Th1 cells, as compared with WT, showed defects in adhesion and transmigration across tumor necrosis factor-α (TNF-α)-activated murine endothelium and in adhesion to immobilized intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion protein 1 (VCAM-1) under flow conditions. Human Jurkat T-cells lacking CD47 also showed reduced adhesion to TNF-α-activated endothelium and ICAM-1 and VCAM-1. In cis interactions between Jurkat T-cell ß2 integrins and CD47 were detected by fluorescence lifetime imaging microscopy. Unexpectedly, Jurkat CD47 null cells exhibited a striking defect in ß1 and ß2 integrin activation in response to Mn(2+) or Mg(2+)/ethylene glycol tetraacetic acid treatment. Our results demonstrate that CD47 associates with ß2 integrins and is necessary to induce high-affinity conformations of LFA-1 and VLA-4 that recognize their endothelial cell ligands and support leukocyte adhesion and transendothelial migration.


Subject(s)
CD47 Antigen/immunology , Integrin alpha4beta1/immunology , Lymphocyte Function-Associated Antigen-1/immunology , T-Lymphocytes/immunology , Animals , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cell Adhesion/immunology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/metabolism , Humans , Immunoblotting , Integrin alpha4beta1/metabolism , Jurkat Cells , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Protein Binding/immunology , T-Lymphocytes/metabolism , Transendothelial and Transepithelial Migration/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
9.
Mol Imaging ; 12(8)2013.
Article in English | MEDLINE | ID: mdl-24447619

ABSTRACT

CD47 functions as a marker of "self" by inhibiting phagocytosis of autologous cells. CD47 has been shown to be overexpressed by various tumor types as a means of escaping the antitumor immune response. The goal of this research was to investigate the utility of CD47 imaging using positron emission tomography (PET) in both human xenograft and murine allograft tumor models. Anti-CD47 antibodies were conjugated with p-isothiocyanatobenzyldesferrioxamine (Df-Bz-NCS) and labeled with 89Zr. We employed xenograft and allograft small-animal models of cancer in biodistribution and PET imaging studies to investigate the specificity and PET imaging robustness of CD47. Ab-Df-Bz-NCS conjugates were labeled with 89Zr with specific activity of 0.9 to 1.6 µCi/µg. Biodistribution studies in the xenograft and allograft model showed similar specific tumor uptake of the antihuman and antimouse CD47 antibodies. However, the tracer retention in the liver, spleen, and kidneys was significantly higher in the allograft-bearing animals, suggesting uptake mediated by the CD47 normally expressed throughout the reticular endothelial system. CD47, a marker of "self," was evaluated as a diagnostic PET biomarker in xenograft and allograft cancer animal models. CD47 imaging is feasible, warranting further studies and immunoPET tracer development.


Subject(s)
CD47 Antigen/analysis , Neoplasms, Experimental/diagnostic imaging , Allografts , Animals , Antibodies, Monoclonal/immunology , CD47 Antigen/immunology , Cell Line, Tumor , Female , Heterografts , Humans , Kidney/metabolism , Liver/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Positron-Emission Tomography , Tissue Distribution
11.
Matrix Biol ; 30(2): 154-61, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21256215

ABSTRACT

CD47, a receptor for thrombospondin-1, limits two important regulatory axes: nitric oxide-cGMP signaling and cAMP signaling, both of which can promote mitochondrial biogenesis. Electron microscopy revealed increased mitochondrial densities in skeletal muscle from both CD47 null and thrombospondin-1 null mice. We further assessed the mitochondria status of CD47-null vs WT mice. Quantitative RT-PCR of RNA extracted from tissues of 3 month old mice revealed dramatically elevated expression of mRNAs encoding mitochondrial proteins and PGC-1α in both fast and slow-twitch skeletal muscle from CD47-null mice, but modest to no elevation in other tissues. These observations were confirmed by Western blotting of mitochondrial proteins. Relative amounts of electron transport enzymes and ATP/O(2) ratios of isolated mitochondria were not different between mitochondria from CD47-null and WT cells. Young CD47-null mice displayed enhanced treadmill endurance relative to WTs and CD47-null gastrocnemius had undergone fiber type switching to a slow-twitch pattern of myoglobin and myosin heavy chain expression. In 12 month old mice, both skeletal muscle mitochondrial volume density and endurance had decreased to wild type levels. Expression of myosin heavy chain isoforms and myoglobin also reverted to a fast twitch pattern in gastrocnemius. Both CD47 and TSP1 null mice are leaner than WTs, use less oxygen and produce less heat than WT mice. CD47-null cells produce substantially less reactive oxygen species than WT cells. These data indicate that loss of signaling from the TSP1-CD47 system promotes accumulation of normally functioning mitochondria in a tissue-specific and age-dependent fashion leading to enhanced physical performance, lower reactive oxygen species production and more efficient metabolism.


Subject(s)
Aging/physiology , CD47 Antigen/metabolism , Mitochondria/physiology , Muscle, Skeletal/metabolism , Animals , Body Composition/physiology , Body Weight/physiology , CD47 Antigen/genetics , Cytochromes b/genetics , Cytochromes c/genetics , Cytochromes c/metabolism , Energy Metabolism/physiology , Female , Gene Expression/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/ultrastructure , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/ultrastructure , Muscle, Skeletal/ultrastructure , Myocytes, Smooth Muscle/metabolism , Myosin Heavy Chains/genetics , Nuclear Respiratory Factor 1/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Endurance/physiology , Protein Isoforms/genetics , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Thrombospondin 1/genetics , Trans-Activators/genetics , Transcription Factors , Voltage-Dependent Anion Channel 1/metabolism
12.
Nat Immunol ; 11(2): 155-61, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20037584

ABSTRACT

In atherosclerosis and Alzheimer's disease, deposition of the altered self components oxidized low-density lipoprotein (LDL) and amyloid-beta triggers a protracted sterile inflammatory response. Although chronic stimulation of the innate immune system is believed to underlie the pathology of these diseases, the molecular mechanisms of activation remain unclear. Here we show that oxidized LDL and amyloid-beta trigger inflammatory signaling through a heterodimer of Toll-like receptors 4 and 6. Assembly of this newly identified heterodimer is regulated by signals from the scavenger receptor CD36, a common receptor for these disparate ligands. Our results identify CD36-TLR4-TLR6 activation as a common molecular mechanism by which atherogenic lipids and amyloid-beta stimulate sterile inflammation and suggest a new model of TLR heterodimerization triggered by coreceptor signaling events.


Subject(s)
CD36 Antigens/immunology , Inflammation/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 6/immunology , Amyloid beta-Peptides/immunology , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , Blotting, Western , CD36 Antigens/metabolism , Cell Line , Chemokines/biosynthesis , Chemokines/immunology , Gene Expression , Humans , Immunoprecipitation , Inflammation/metabolism , Lipoproteins, LDL/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 6/metabolism
13.
Early Hum Dev ; 85(10): 659-63, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19815354

ABSTRACT

BACKGROUND: Neutrophils (PMN) are the primary leukocyte responders during acute inflammation. After migrating into the tissues, PMN undergo programmed cell death (apoptosis) and are subsequently removed via phagocytosis by resident macrophages during the resolution phase. Efficient phagocytosis of apoptotic neutrophils is necessary for successful resolution. CD47 plays a critical role in mediating the phagocytic response, although its role in the phagocytosis of apoptotic PMN is incompletely understood. AIMS: In the present study we tested the hypotheses that CD47 modulates the targeting of apoptotic PMN for phagocytosis, and that this process is altered in neonatal PMN. STUDY DESIGN: Adult and neonatal PMN were examined for their expression of CD47. To investigate CD47-mediated functions, apoptotic adult and neonatal PMN were co-cultured with monocyte-derived macrophages (MDM) and the phagocytic index was determined using a flow cytometry-based assay. RESULTS: We observed lower basal surface CD47 levels on neonatal vs. adult PMN. In both groups, spontaneous apoptosis led to decreased surface and total cellular CD47 expression. Adult and neonatal MDM ingested apoptotic neonatal target PMN more avidly than apoptotic adult target PMN. Masking of surface CD47 on PMN with a monoclonal antibody enhanced MDM phagocytic activity. CONCLUSIONS: Our results suggest that age-dependent expression of CD47 on PMN may account for differences in their ingestion by macrophages and in the resolution of inflammation.


Subject(s)
Apoptosis , CD47 Antigen/metabolism , Macrophages/physiology , Neutrophils/metabolism , Phagocytosis/physiology , Adult , Age Factors , Coculture Techniques , Female , Flow Cytometry , Humans , Infant, Newborn , Neutrophils/cytology , Placenta/cytology , Pregnancy
14.
Matrix Biol ; 28(2): 110-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19284971

ABSTRACT

Nitric oxide (NO) locally regulates vascular resistance and blood pressure by modulating blood vessel tone. Thrombospondin-1 signaling via its receptor CD47 locally limits the ability of NO to relax vascular smooth muscle cells and increase regional blood flow in ischemic tissues. To determine whether thrombospondin-1 plays a broader role in central cardiovascular physiology, we examined vasoactive stress responses in mice lacking thrombospondin-1 or CD47. Mice lacking thrombospondin-1 exhibit activity-associated increases in heart rate, central diastolic and mean arterial blood pressure and a constant decrease in pulse pressure. CD47-deficient mice have normal central pulse pressure but elevated resting peripheral blood pressure. Both null mice show exaggerated decreases in peripheral blood pressure and increased cardiac output and ejection fraction in response to NO. Autonomic blockade also induces exaggerated hypotensive responses in awake thrombospondin-1 null and CD47 null mice. Both null mice exhibit a greater hypotensive response to isoflurane, and autonomic blockage under isoflurane anesthesia leads to premature death of thrombospondin-1 null mice. Conversely, the hypertensive response to epinephrine is attenuated in thrombospondin-1 null mice. Thus, the matricellular protein thrombospondin-1 and its receptor CD47 serve as acute physiological regulators of blood pressure and exert a vasopressor activity to maintain global hemodynamics under stress.


Subject(s)
Blood Pressure/physiology , CD47 Antigen/metabolism , Heart Rate/physiology , Heart/physiology , Signal Transduction/physiology , Stress, Physiological/physiology , Thrombospondin 1/metabolism , Animals , Blood Pressure/genetics , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Echocardiography , Heart Rate/genetics , Immunoassay , Mice , Mice, Knockout , Regional Blood Flow , Skin/blood supply , Thrombospondin 1/deficiency
15.
Cancer Res ; 69(7): 3196-204, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19276363

ABSTRACT

CD47, also called integrin-associated protein, plays a critical role in the innate immune response and is an atypical member of the immunoglobulin superfamily that interacts with and activates beta3 integrins. beta3 integrin(-/-) mice have defective platelet and osteoclast function and are protected from bone metastasis. The role of CD47 in skeletal homeostasis and bone metastasis has not been described. CD47(-/-) mice had increased bone mass and defective osteoclast function in vivo. Although the number of functional osteoclasts formed by differentiating CD47(-/-) bone marrow macrophages was decreased, high doses of RANKL rescued differentiation and function of CD47(-/-) osteoclasts ex vivo and rescued the osteoclast defect in CD47(-/-) mice. Inhibition of nitric oxide (NO) synthase, which is expressed at higher levels in CD47(-/-) osteoclasts, also rescued the osteoclast defect in CD47(-/-) cells. We then examined the consequences of this osteoclast defect in bone metastasis. In a model of tumor metastasis to bone, bone tumor burden was decreased in the CD47(-/-) mice compared with wild-type (WT) controls, with no decrease in s.c. tumor growth in CD47(-/-) mice. There was decreased tumor-associated bone destruction in the CD47(-/-) mice compared with WT controls, consistent with a defect in osteoclast function that was not rescued by the presence of tumor. Our data show that CD47 regulates osteoclastogenesis, in part, via regulation of NO production, and its disruption leads to a decrease in tumor bone metastasis. CD47 is a novel therapeutic target to strengthen bone mass and diminish metastatic tumor growth in bone.


Subject(s)
Bone Neoplasms/secondary , CD47 Antigen/metabolism , Melanoma, Experimental/pathology , Animals , Bone Neoplasms/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Humans , Integrin beta3/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase/antagonists & inhibitors , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , RANK Ligand/pharmacology
16.
J Biol Chem ; 284(2): 1116-25, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-19004835

ABSTRACT

Thrombospondin-1 regulates nitric oxide (NO) signaling in vascular cells via CD47. Because CD47 binding motifs are conserved in the C-terminal signature domains of all five thrombospondins and indirect evidence has implied CD47 interactions with other family members, we compared activities of recombinant signature domains of thrombospondin-1, -2, and -4 to interact with CD47 and modulate cGMP signaling. Signature domains of thrombospondin-2 and -4 were less active than that of thrombospondin-1 for inhibiting binding of radiolabeled signature domain of thrombospondin-1 or SIRPalpha (signal-regulatory protein) to cells expressing CD47. Consistent with this binding selectivity, the signature domain of thrombospondin-1 was more potent than those of thrombospondin-2 or -4 for inhibiting NO-stimulated cGMP synthesis in vascular smooth muscle cells and downstream effects on cell adhesion. In contrast to thrombospondin-1- and CD47-null cells, primary vascular cells from thrombospondin-2-null mice lack enhanced basal and NO-stimulated cGMP signaling. Effects of endogenous thrombospondin-2 on NO/cGMP signaling could be detected only in thrombospondin-1-null cells. Furthermore, tissue survival of ischemic injury and acute recovery of blood flow in thrombospondin-2-nulls resembles that of wild type mice. Therefore, thrombospondin-1 is the dominant regulator of NO/cGMP signaling via CD47, and its limiting role in acute ischemic injury responses is not shared by thrombospondin-2.


Subject(s)
CD47 Antigen/metabolism , Cyclic GMP/metabolism , Ischemia/metabolism , Signal Transduction , Thrombospondins/metabolism , Animals , Cells, Cultured , Humans , Ischemia/genetics , Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Protein Binding , Receptors, Immunologic/metabolism , Thrombospondins/deficiency , Thrombospondins/genetics
17.
Nature ; 457(7227): 318-21, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19037245

ABSTRACT

Tissue macrophages comprise a heterogeneous group of cell types differing in location, surface markers and function. Red pulp macrophages are a distinct splenic subset involved in removing senescent red blood cells. Transcription factors such as PU.1 (also known as Sfpi1) and C/EBPalpha (Cebpa) have general roles in myelomonocytic development, but the transcriptional basis for producing tissue macrophage subsets remains unknown. Here we show that Spi-C (encoded by Spic), a PU.1-related transcription factor, selectively controls the development of red pulp macrophages. Spi-C is highly expressed in red pulp macrophages, but not monocytes, dendritic cells or other tissue macrophages. Spic(-/-) mice have a cell-autonomous defect in the development of red pulp macrophages that is corrected by retroviral Spi-C expression in bone marrow cells, but have normal monocyte and other macrophage subsets. Red pulp macrophages highly express genes involved in capturing circulating haemoglobin and in iron regulation. Spic(-/-) mice show normal trapping of red blood cells in the spleen, but fail to phagocytose these red blood cells efficiently, and develop an iron overload localized selectively to splenic red pulp. Thus, Spi-C controls development of red pulp macrophages required for red blood cell recycling and iron homeostasis.


Subject(s)
DNA-Binding Proteins/metabolism , Erythrocytes/metabolism , Homeostasis , Iron/metabolism , Macrophages/physiology , Phagocytosis , Spleen/metabolism , Animals , Cell Count , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Erythrocytes/cytology , Gene Expression Regulation , Macrophages/cytology , Mice , Vascular Cell Adhesion Molecule-1/genetics
18.
Surgery ; 144(5): 752-61, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19081017

ABSTRACT

BACKGROUND: Ischemia-reperfusion (I/R) injury remains a primary complication of transplant surgery, accounting for about 80% of liver transplant failures, and is a major source of morbidity in other pathologic conditions. Activation of endothelium and inflammatory cell recruitment are central to the initiation and promulgation of I/R injury, which can be limited by the bioactive gas nitric oxide (NO). The discovery that thrombsospondin-1 (TSP1), via CD47, limits NO signaling in vascular cells and ischemic injuries in vivo suggested that I/R injury could be another important target of this signaling pathway. METHODS: Wild-type, TSP1-null, and CD47-null mice underwent liver I/R injury. Wild-type animals were pretreated with CD47 or control antibodies before liver I/R injury. Tissue perfusion via laser Doppler imaging, serum enzymes, histology, and immunohistology were assessed. RESULTS: TSP1-null and CD47-null mice subjected to subtotal liver I/R injury showed improved perfusion relative to wild-type mice. Null mice subjected to liver I/R had decreased liver enzyme release and less histologic evidence of injury. Elevated TSP1 expression in liver tissue after I/R injury suggested that preventing its interaction with CD47 could be protective. Thus, pretreatment of wild-type mice using a blocking CD47 antibody improved recovery of tissue perfusion and preserved liver integrity after I/R injury. CONCLUSIONS: Tissue survival and perfusion after liver I/R injury are limited by TSP1 and CD47. Targeting CD47 before I/R injury enhances tissue survival and perfusion in a model of liver I/R injury and suggests therapeutics for enhancing organ survival in transplantation surgery.


Subject(s)
CD47 Antigen/physiology , Liver/surgery , Reperfusion Injury/etiology , Reperfusion Injury/therapy , Signal Transduction/physiology , Thrombospondin 1/physiology , Animals , Antibodies, Monoclonal/therapeutic use , Female , Immunologic Factors/therapeutic use , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Recovery of Function , Reperfusion Injury/metabolism
19.
Biochemistry ; 47(44): 11616-24, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18841997

ABSTRACT

Control of alphaIIb beta3 and alphav beta3 integrin activation is critical for cardiovascular homeostasis. Mutations that perturb association of integrin alpha and beta subunits in their transmembrane and cytoplasmic regions activate the integrin heterodimer, suggesting that a low-affinity or "off" conformation is the default state, likely corresponding to the bent conformation seen in the crystal structure of alphav beta3. In this bent structure, a segment of alphav (301-308) and beta3 (560-567) are juxtaposed. Here we provide evidence that these regions of alphav/alphaIIb and beta3 function as a novel extracellular clasp to restrain activation. Synthetic peptides representing the alphaIIb and beta3 clasp regions promote integrin activation as judged by cell adhesion, cell spreading, and exposure of epitopes for three beta3 LIBS antibodies. Mutation of the clasp region of alphav or beta3 results in a constitutively activated integrin, confirming the role of the extracellular clasp in restraining integrin activation. Molecular dynamics simulations of the alphav beta3 structure yield a refined model for the alphav beta3 clasp and provide plausible explanations for the effects of the activating mutations.


Subject(s)
Integrin beta3/chemistry , Blood Platelets/metabolism , Cell Line , Dimerization , Humans , In Vitro Techniques , Integrin alphaV/chemistry , Integrin alphaV/genetics , Integrin alphaV/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism , K562 Cells , Models, Molecular , Mutagenesis, Site-Directed , Platelet Membrane Glycoprotein IIb/chemistry , Platelet Membrane Glycoprotein IIb/genetics , Platelet Membrane Glycoprotein IIb/metabolism , Protein Conformation , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
Curr Drug Targets ; 9(10): 833-41, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18855617

ABSTRACT

Activation of soluble guanylate cyclase by nitric oxide (NO) controls signaling pathways that play critical roles in normal vascular physiology and in the pathogenesis of cardiovascular disease. We have identified the secreted protein thrombospondin-1 as a key regulator of NO signaling. Thrombospondin-1 limits the angiogenic activity of NO in endothelial cells, its vasodilator activity in vascular smooth muscle, and its antithrombotic activity in platelets. Loss of either thrombospondin-1 or its receptor CD47 in transgenic mice results in hyperdynamic responses to NO and reveals the importance of this pathway in normal physiology. Thrombospondin-1 and CD47 null mice show improved abilities to respond to ischemic stress, suggesting that therapeutic targeting of this pathway could benefit patients with a variety of ischemic conditions. We review the preclinical development of therapeutics targeting thrombospondin-1 or CD47 for improving survival of fixed ischemia, ischemia due to aging and peripheral vascular disease, and skin grafting.


Subject(s)
CD47 Antigen/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Signal Transduction/physiology , Thrombospondin 1/antagonists & inhibitors , Thrombospondin 1/physiology , Animals , CD47 Antigen/genetics , CD47 Antigen/physiology , Cardiovascular Diseases/genetics , Humans , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/physiology , Signal Transduction/genetics , Thrombospondin 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...