Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 8(4): 896-905, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25492474

ABSTRACT

Neutrophil elastase (NE) and cathepsin G (CG) contribute to intracellular microbial killing but, if left unchecked and released extracellularly, promote tissue damage. Conversely, mechanisms that constrain neutrophil serine protease activity protect against tissue damage but may have the untoward effect of disabling the microbial killing arsenal. The host elaborates thrombospondin-1 (TSP-1), a matricellular protein released during inflammation, but its role during neutrophil activation following microbial pathogen challenge remains uncertain. Mice deficient in TSP-1 (thbs1(-/-)) showed enhanced lung bacterial clearance, reduced splenic dissemination, and increased survival compared with wild-type (WT) controls during intrapulmonary Klebsiella pneumoniae infection. More effective pathogen containment was associated with reduced burden of inflammation in thbs1(-/-) mouse lungs compared with WT controls. Lung NE activity was increased in thbs1(-/-) mice following K. pneumoniae challenge, and thbs1(-/-) neutrophils showed enhanced intracellular microbial killing that was abrogated with recombinant TSP-1 administration or WT serum. Thbs1(-/-) neutrophils exhibited enhanced NE and CG enzymatic activity, and a peptide corresponding to amino-acid residues 793-801 within the type-III repeat domain of TSP-1 bridled neutrophil proteolytic function and microbial killing in vitro. Thus, TSP-1 restrains proteolytic action during neutrophilic inflammation elicited by K. pneumoniae, providing a mechanism that may regulate the microbial killing arsenal.


Subject(s)
Immunity, Innate , Klebsiella Infections/immunology , Klebsiella Infections/metabolism , Klebsiella pneumoniae/immunology , Neutrophils/immunology , Neutrophils/metabolism , Serine Proteases/metabolism , Thrombospondin 1/metabolism , Animals , Cathepsin G/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Klebsiella Infections/mortality , Klebsiella Infections/pathology , Leukocyte Elastase/metabolism , Lung/immunology , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Mice, Knockout , Neutrophils/drug effects , Peptides/pharmacology , Recombinant Proteins/pharmacology , Respiratory Burst/genetics , Respiratory Burst/immunology , Spleen/immunology , Spleen/metabolism , Spleen/microbiology , Thrombospondin 1/chemistry , Thrombospondin 1/deficiency , Thrombospondin 1/genetics , Thrombospondin 1/pharmacology
2.
J Thromb Haemost ; 12(11): 1906-17, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25163645

ABSTRACT

INTRODUCTION: Activated platelets shed microparticles from plasma membranes, but also release smaller exosomes from internal compartments. While microparticles participate in athero-thrombosis, little is known of exosomes in this process. MATERIALS & METHODS: Ex vivo biochemical experiments with human platelets and exosomes, and FeCl3 -induced murine carotid artery thrombosis. RESULTS: Both microparticles and exosomes were abundant in human plasma. Platelet-derived exosomes suppressed ex vivo platelet aggregation and reduced adhesion to collagen-coated microfluidic channels at high shear. Injected exosomes inhibited occlusive thrombosis in FeCl3 -damaged murine carotid arteries. Control platelets infused into irradiated, thrombocytopenic mice reconstituted thrombosis in damaged carotid arteries, but failed to do so after prior ex vivo incubation with exosomes.CD36 promotes platelet activation, and exosomes dramatically reduced platelet CD36.CD36 is also expressed by macrophages, where it binds and internalizes oxidized LDL and microparticles, supplying lipid to promote foam cell formation. Platelet exosomes inhibited oxidized-LDL binding and cholesterol loading into macrophages. Exosomes were not competitive CD36 ligands, but instead sharply reduced total macrophage CD36 content. Exosomal proteins, in contrast to microparticle or cellular proteins, were highly adducted by ubiquitin. Exosomes enhanced ubiquitination of cellular proteins, including CD36, and blockade of proteosome proteolysis with MG-132 rescued CD36 expression. Recombinant unanchored K48 poly-ubiquitin behaved similarly to exosomes, inhibiting platelet function, macrophage CD36 expression and macrophage particle uptake. CONCLUSIONS: Platelet-derived exosomes inhibit athero-thrombotic processes by reducing CD36-dependent lipid loading of macrophages and by suppressing platelet thrombosis. Exosomes increase protein ubiquitination and enhance proteasome degradation of CD36.


Subject(s)
Blood Platelets/metabolism , CD36 Antigens/blood , Carotid Artery Diseases/prevention & control , Exosomes/metabolism , Platelet Activation , Polyubiquitin/metabolism , Thrombosis/prevention & control , Animals , Carotid Artery Diseases/blood , Carotid Artery Diseases/chemically induced , Cell-Derived Microparticles/metabolism , Chlorides , Cholesterol/blood , Collagen/metabolism , Disease Models, Animal , Ferric Compounds , Foam Cells/metabolism , Humans , Lipoproteins, LDL/blood , Mice, Inbred C57BL , Platelet Adhesiveness , Platelet Aggregation , Platelet Transfusion , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction , Thrombocytopenia/blood , Thrombosis/blood , Thrombosis/chemically induced , Time Factors , Ubiquitination
3.
Mucosal Immunol ; 7(2): 440-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24045574

ABSTRACT

Mononuclear phagocyte recognition of apoptotic cells triggering suppressive cytokine signaling is a key event in inflammation resolution from injury. Mice deficient in thrombospondin (TSP)-1 (thbs1⁻/⁻), an extracellular matrix glycoprotein that bridges cell-cell interactions, are prone to lipopolysaccharide-induced lung injury and show defective macrophage interleukin (IL)-10 production during the resolution phase of inflammation. Reconstitution of IL-10 rescues thbs1⁻/⁻ mice from persistent neutrophilic lung inflammation and injury and thbs1⁻/⁻ alveolar macrophages show defective IL-10 production following intratracheal instillation of apoptotic neutrophils despite intact efferocytosis. Following co-culture with apoptotic neutrophils, thbs1⁻/⁻ macrophages show a selective defect in IL-10 production, whereas prostaglandin E2 and transforming growth factor beta 1 responses remain intact. Full macrophage IL-10 responses require the engagement of TSP-1 structural repeat 2 domain and the macrophage scavenger receptor CD36 LIMP-II Emp sequence homology (CLESH) domain in vitro. Although TSP-1 is not essential for macrophage engulfment of apoptotic neutrophils in vivo, TSP-1 aids in the curtailment of inflammatory responses during the resolution phase of injury in the lungs by providing a means by which apoptotic cells are recognized and trigger optimal IL-10 production by macrophages.


Subject(s)
Interleukin-10/biosynthesis , Lung Injury/immunology , Lung Injury/metabolism , Macrophages/immunology , Macrophages/metabolism , Thrombospondin 1/metabolism , Animals , Apoptosis/immunology , CD36 Antigens/genetics , CD36 Antigens/metabolism , Dinoprostone/deficiency , Disease Models, Animal , Lipopolysaccharides/adverse effects , Lung Injury/chemically induced , Lung Injury/genetics , Lung Injury/pathology , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , Protein Interaction Domains and Motifs/genetics , Signal Transduction , Thrombospondin 1/chemistry , Thrombospondin 1/genetics , Transforming Growth Factor beta1/metabolism
4.
Int J Lab Hematol ; 34(5): 473-83, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22574824

ABSTRACT

INTRODUCTION: Association of P2RY1 and P2RY12 polymorphisms with on-aspirin platelet reactivity was investigated. MATERIALS AND METHODS: Platelet reactivity was assessed by the light transmission aggregometry and TxB(2) assay in 423 patients with coronary artery disease (CAD) on aspirin. High residual platelet reactivity (RPR) was defined by ≥20% and ≥70% maximal aggregation stimulated with 0.5 mg/mL arachidonic acid (AA) and 10 µm ADP, respectively. Moderate RPR was considered aggregation ≥20% with AA, ≥70% with ADP, or ≥1 ng/mL stimulated TxB(2) . Fourteen P2RY1 and 35 P2RY12 single nucleotide polymorphisms (SNPs) were genotyped. RESULTS: High RPR was detected in 24% of the patients. Moderate RPR was observed in 31% with AA, 57% with 5 µm ADP, and 82% with 10 µm ADP. Stimulated TxB(2) was ≥1 ng/mL in 23% of patients. P2RY12 SNP rs9859538 was associated with high RPR (OR = 2.16, 95% CI = 1.24-3.75, P-value = 0.004). Four P2RY12 SNPs, rs1491974, rs10513398, rs3732765, and rs10935841, showed association with moderate RPR (OR = 1.79-2.94, P-value = 0.04-0.028), while five, rs7615865, rs1388623, rs1388622, rs7634096, and rs7637803, were associated with low RPR (OR = 0.50-0.55, P-value = 0.008-0.026), following ADP stimulation. TxB(2) level <1 ng/mL was linked to five P2RY1 SNPs, rs1439010, rs1371097, rs701265, rs12497578, and rs2312265 (OR = 0.36-0.54, P-value = 0.003-0.039). CONCLUSIONS: Polymorphisms in P2RY1 and P2RY12 are associated with on-aspirin platelet reactivity in patients with CAD.


Subject(s)
Aspirin/therapeutic use , Coronary Artery Disease/drug therapy , Platelet Aggregation/drug effects , Polymorphism, Single Nucleotide , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y1/genetics , Aged , Coronary Artery Disease/blood , Coronary Artery Disease/genetics , Female , Gene Frequency , Genotype , Humans , Logistic Models , Male , Middle Aged , Odds Ratio , Platelet Aggregation Inhibitors/therapeutic use , Platelet Function Tests
5.
Matrix Biol ; 24(1): 27-34, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15748999

ABSTRACT

Thrombospondins-1 and -2 (TSP-1, TSP-2) are matricellular glycoproteins with potent antiangiogenic activity. We have previously shown that the antiangiogenic activity of TSP-1 is mediated by the interaction of the type I repeats (TSR) with the receptor CD36, although other domains of TSP-1 have also been implicated. We now show that the antiangiogenic activity of TSP-2, which contains three TSRs but, unlike TSP-1, lacks the capacity to activate TGF-beta, is similarly dependent on CD36. Using the corneal pocket assay we found that TSP-2 did not inhibit bFGF-induced angiogenesis in CD36 null mice. We then demonstrated that (125)[I]-TSP-2 bound to murine macrophages and that binding was diminished by 70% by anti-CD36 antibody or by using cells from CD36 null animals. Solid-phase binding studies revealed that (125)[I]-TSP-2 bound to CD36/glutathione-S-transferase (GST) fusion proteins encoding the region spanning amino acids 93-120, but not amino acids 298-439. This 93-120 amino acid region, previously identified as the TSP-1 binding site, is homologous to domains on other TSP binding proteins, such as LIMP-2 and histidine-rich glycoprotein (HRGP). Finally, we showed with an immunoabsorbent binding assay that TSP-2 bound HRGP with high affinity and that HRGP blocked the antiangiogenic activity of TSP-2, acting like a "decoy" receptor. These data suggest that modulation of the TSR/CD36 system may play an important role in the regulation of the angiogenic "switch," and may provide a target for therapeutic interventions.


Subject(s)
CD36 Antigens/pharmacology , Neovascularization, Pathologic , Proteins/chemistry , Thrombospondins/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , CD36 Antigens/chemistry , Cell Adhesion Molecules/metabolism , Dose-Response Relationship, Drug , Fibroblast Growth Factor 2/metabolism , Glutathione Transferase/metabolism , Macromolecular Substances/chemistry , Macrophages/metabolism , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Peptides/chemistry , Platelet Membrane Glycoproteins/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Temperature , Thrombospondins/chemistry , Time Factors
6.
Leuk Res ; 28(9): 979-85, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15234576

ABSTRACT

This meeting was convened by Richard T. Silver, M.D. and co-chaired by Jerry L. Spivak, M.D. It was held from 16 to 18 October 2003 in New York City, New York, USA. Thirty-nine invited speakers from nine different countries participated in the conference. There were more than 350 attendees. There were formal presentations and discussions on biology, clinical aspects, and management of patients with these diverse bone marrow stem cell disorders linked by a variable progression to acute myeloid leukemia. Of considerable interest, a clinical symposium exclusively for patients was held the day preceding the meeting at which John Bennett, Tiziano Barbui, Richard Silver, Jerry Spivak, and Ayalew Tefferi spoke on various topics pertaining to these diseases. This proved to be highly informative to the patients who reported that they enjoyed the program immensely. This was sponsored by the Cancer Research & Treatment Fund, Inc. Representatives of the Myelodysplasia Foundation were also present. This meeting report provides a summary of five different sections prepared by one or more of the session chairs. The keynote address was given by Shahin Rafii (Cornell Medical Center). Most appropriately, this talk focused on the expression and activation of angiogenic factors which play a crucial role in the progression of both myeloproliferative disorders and myelodysplastic syndromes (MDS). Among the known factors, vascular endothelial growth tyrosine kinase receptors (VEGF-R1, R2, and R3) support proliferation, survival, and mobility. Rafii's team has demonstrated that these receptors are expressed on subsets of primary hematopoietic cells as well as leukemic cells. Some leukemic cells express both VEGF-A and VEGF-R2, resulting in the generation of an autocrine loop that supports survival and within the osteoblastic zone translocating these cells to the vascular enriched niche for receipt of molecular instructions required for proliferation and differentiation. A pathologic correlation can be seen in some patients with the identification of abnormal localization of immature precursors (ALIP) in the central portions of the medullary cavity. Misplaced megakaryocytes can release pro-fibrotic factors, including platelet derived growth factors and transforming growth factor-beta. Collectively, these data suggest that chronic disregulation of angiogenic factors alter the microenvironment dislocating marrow stem cells that force both proliferation and differentiation in varying degrees, contributing to these hematological disorders.


Subject(s)
Myelodysplastic Syndromes , Myeloproliferative Disorders , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/genetics , Polycythemia , Primary Myelofibrosis , Thrombocytosis
7.
Mol Cell Biochem ; 239(1-2): 193-7, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12479585

ABSTRACT

Fatty acid translocase (FAT)/CD36 has been associated with diverse normal and pathologic processes. These include scavenger receptor functions (uptake of apoptotic cells and modified lipid), lipid metabolism and fatty acid transport, adhesion, angiogenesis, modulation of inflammation, transforming growth factor-beta activation, atherosclerosis, diabetes and cardiomyopathy. Although CD36 was identified more than 25 years ago, it is only with the advent of recent genetic technology that in vivo evidence has emerged for its physiologic and pathologic relevance. As these in vivo studies are expanded, we will gain further insight into the mechanism(s) by which CD36 transmits a cellular signal, and this will allow the design of specific therapeutics that impact on a particular function of CD36.


Subject(s)
Fatty Acids/metabolism , Membrane Glycoproteins/metabolism , Organic Anion Transporters/metabolism , Adipocytes/metabolism , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , Disease Models, Animal , Humans , Membrane Glycoproteins/genetics , Organic Anion Transporters/genetics
8.
Circ Res ; 90(8): 844-9, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11988484

ABSTRACT

Growth hormone-releasing peptides (GHRPs) are known as potent growth hormone secretagogues whose actions are mediated by the ghrelin receptor, a G protein-coupled receptor cloned from pituitary libraries. Hexarelin, a hexapeptide of the GHRP family, has reported cardiovascular activity. To identify the molecular target mediating this activity, rat cardiac membranes were labeled with a radioactive photoactivatable derivative of hexarelin and purified using lectin affinity chromatography and preparative gel electrophoresis. A binding protein of M(r) 84 000 was identified. The N-terminal sequence determination of the deglycosylated protein was identical to rat CD36, a multifunctional glycoprotein, which was expressed in cardiomyocytes and microvascular endothelial cells. Activation of CD36 in perfused hearts by hexarelin was shown to elicit an increase in coronary perfusion pressure in a dose-dependent manner. This effect was lacking in hearts from CD36-null mice and hearts from spontaneous hypertensive rats genetically deficient in CD36. The coronary vasoconstrictive response correlated with expression of CD36 as assessed by immunoblotting and covalent binding with hexarelin. These data suggest that CD36 may mediate the coronary vasospasm seen in hypercholesterolemia and atherosclerosis.


Subject(s)
CD36 Antigens/physiology , Heart/physiology , Oligopeptides/pharmacology , Vasoconstrictor Agents/pharmacology , Animals , Binding Sites , CD36 Antigens/genetics , CD36 Antigens/isolation & purification , Cell Membrane/chemistry , Heart/drug effects , Immunoblotting , Mice , Mice, Knockout , Myocardium/chemistry , Oligopeptides/metabolism , Organ Culture Techniques , Photoaffinity Labels/chemistry , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Receptors, Neuropeptide/isolation & purification , Receptors, Pituitary Hormone-Regulating Hormone/isolation & purification , Vasoconstriction/drug effects , Vasoconstrictor Agents/metabolism
9.
Article in English | MEDLINE | ID: mdl-11723002

ABSTRACT

This year the Hematology Grants Workshop, chaired by Dr. Todd, includes a comprehensive listing of available National Institutes of Health, Department of Veterans Affairs, and non-federal grants applicable to fellows and junior faculty as well as to established investigators. In Section II, Dr. Miller discusses the essential principles of successful grant writing with a special emphasis on the young investigator. He highlights the best strategies to take and the common mistakes to avoid. In Section III, Dr. Silverstein outlines the structure of the current NIH Integrated Review Group (IRG) system and the study sections of the most relevance to hematology. He traces the path that a grant takes from review to funding including the way in which grants are reviewed at NIH Study Section Meetings and provides advice in the preparation of revised applications.


Subject(s)
Hematology , Research Support as Topic/methods , Consensus Development Conferences as Topic , Humans , National Institutes of Health (U.S.) , Peer Review, Research/methods , Research Design/standards , Research Support as Topic/organization & administration , Research Support as Topic/standards , United States , Writing
10.
J Exp Med ; 194(9): 1289-98, 2001 Nov 05.
Article in English | MEDLINE | ID: mdl-11696594

ABSTRACT

Retinal pigment epithelial (RPE) cells employ alphavbeta5 integrin and CD36 receptors to phagocytose photoreceptor outer segment fragments (OS). We explored special properties of RPE phagocytosis to identify the contribution of CD36 to RPE phagocytosis measuring effects of CD36 antibodies on OS binding and internalization kinetics. Early, CD36 antibodies had no effect on OS binding or internalization. Both control and CD36 antibody treated RPE initiated internalization approximately 2 hours after OS challenge. Later, bivalent CD36 IgG accelerated OS engulfment while monovalent Fab fragments inhibited engulfment. Cross-linking Fab fragments restored the accelerating activity of intact IgG. Strikingly, antibodies were effective even if added to OS already bound by RPE. alphavbeta5 blocking antibody reduced OS binding equally well in the presence of CD36 antibodies but CD36 antibodies accelerated internalization of remaining bound OS. Furthermore, CD36 ligation at either apical or basal RPE surface partially substituted for soluble factors that are required for internalization but not for binding of OS at the RPE apical surface. Our results demonstrate that CD36 ligation is necessary and sufficient to activate the OS internalization mechanism of RPE. They suggest that CD36 acts as a signaling molecule in postbinding steps of RPE phagocytosis independently of the OS binding receptor alphavbeta5 integrin.


Subject(s)
CD36 Antigens/immunology , Integrins/immunology , Phagocytosis/immunology , Pigment Epithelium of Eye/immunology , Receptors, Vitronectin , Rod Cell Outer Segment/immunology , Animals , Antibodies, Monoclonal/immunology , CD36 Antigens/biosynthesis , Cell Line , Humans , Kinetics , Lipoproteins, LDL/immunology , Pigment Epithelium of Eye/cytology , Rats
12.
J Clin Invest ; 107(1): 45-52, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134179

ABSTRACT

Angiogenesis is critical for the growth and proliferation of tumors as well as for normal development. We now describe a novel role for histidine-rich glycoprotein (HRGP) in the modulation of angiogenesis. HRGP is a plasma protein that circulates in relatively high concentrations (1.5 microM), but has no known function in vivo. We have shown previously that HRGP binds with high affinity to thrombospondin-1 (TSP-1), a homotrimeric glycoprotein that is a potent inhibitor of angiogenesis. The antiangiogenic activity of TSP-1 is mediated by the binding of properdin-like type I repeats to the receptor CD36. We found that binding of HRGP to TSP-1 was similarly mediated by TSP type I repeats. HRGP colocalized with TSP-1 in the stroma of human breast cancer specimens, and this interaction masked the antiangiogenic epitope of TSP-1. In assays performed in vitro of endothelial cell migration and tube formation, and in vivo corneal angiogenesis assays, HRGP inhibited the antiangiogenic effect of TSP-1. These studies suggest that HRGP can modulate the antiangiogenic activity of TSP-1, and identify a potential mechanism of resistance to the antiangiogenic effect of TSP-1.


Subject(s)
Glycoproteins/pharmacology , Neovascularization, Physiologic/drug effects , Proteins/pharmacology , Thrombospondin 1/antagonists & inhibitors , Thrombospondin 1/pharmacology , Amino Acid Sequence , Binding Sites/genetics , Breast Neoplasms/blood supply , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , In Vitro Techniques , Models, Biological , Molecular Sequence Data , Neovascularization, Pathologic , Proteins/genetics , Proteins/metabolism , Repetitive Sequences, Amino Acid , Sequence Homology, Amino Acid , Thrombospondin 1/genetics , Thrombospondin 1/metabolism
13.
Curr Opin Lipidol ; 11(5): 483-91, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11048891

ABSTRACT

CD36 has been associated with diverse normal and pathologic processes. These include scavenger receptor functions (uptake of apoptotic cells and modified lipid), lipid metabolism and fatty acid transport, adhesion, angiogenesis, modulation of inflammation, transforming growth factor-beta activation, atherosclerosis, diabetes and cardiomyopathy. Although CD36 was identified more than 25 years ago, it is only with the advent of recent genetic technology that in-vivo evidence has emerged for its physiologic and pathologic relevance. As these in-vivo studies are expanded, we will gain further insight into the mechanism(s) by which CD36 transmits a cellular signal, and this will allow the design of specific therapeutics that impact on a particular function of CD36.


Subject(s)
Arteriosclerosis/immunology , CD36 Antigens/metabolism , Membrane Proteins , Receptors, Lipoprotein , Animals , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , CD36 Antigens/chemistry , CD36 Antigens/genetics , Cell Adhesion , Humans , Inflammation/etiology , Lipid Metabolism , Neovascularization, Pathologic , Receptors, Immunologic/metabolism , Receptors, Scavenger , Scavenger Receptors, Class B , Signal Transduction , Transforming Growth Factor beta/metabolism
14.
J Biol Chem ; 275(42): 32523-9, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-10913136

ABSTRACT

The transmembrane protein CD36 has been identified in isolated cell studies as a putative transporter of long chain fatty acids. In humans, an association between CD36 deficiency and defective myocardial uptake of the fatty acid analog 15-(p-iodophenyl)-3-(R, S)-methyl pentadecanoic acid (BMIPP) has been reported. To determine whether this association represents a causal link and to assess the physiological role of CD36, we compared tissue uptake and metabolism of two iodinated fatty acid analogs BMIPP and 15-(p-iodophenyl) pentadecanoic acid (IPPA) in CD36 null and wild type mice. We also investigated the uptake and lipid incorporation of palmitate by adipocytes isolated from both groups. Compared with wild type, uptake of BMIPP and IPPA was reduced in heart (50-80%), skeletal muscle (40-75%), and adipose tissues (60-70%) of null mice. The reduction was associated with a 50-68% decrease in label incorporation into triglycerides and in 2-3-fold accumulation of label in diglycerides. Identical results were obtained from studies of [(3)H]palmitate uptake in isolated adipocytes. The block in diglyceride to triglyceride conversion could not be explained by changes in specific activities of the key enzymes long chain acyl-CoA synthetase and diacylglycerol acyltransferase, which were similar in tissues from wild type and null mice. It is concluded that CD36 facilitates a large fraction of fatty acid uptake by heart, skeletal muscle, and adipose tissues and that CD36 deficiency in humans is the cause of the reported defect in myocardial BMIPP uptake. In CD36-expressing tissues, uptake regulates fatty acid esterification at the level of diacylglycerol acyltransferase by determining fatty acyl-CoA supply. The membrane transport step may represent an important control site for fatty acid metabolism in vivo.


Subject(s)
Adipose Tissue/metabolism , CD36 Antigens/genetics , CD36 Antigens/physiology , Fatty Acids/pharmacokinetics , Iodobenzenes/pharmacokinetics , Muscle, Skeletal/metabolism , Myocardium/metabolism , Animals , Biological Transport , Crosses, Genetic , Female , Humans , Iodine Radioisotopes/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombination, Genetic , Tissue Distribution
15.
Ann N Y Acad Sci ; 902: 128-31; discussion 131-3, 2000 May.
Article in English | MEDLINE | ID: mdl-10865832

ABSTRACT

CD36, an 88 kD transmembrane glycoprotein, is an important receptor for oxidized lipoproteins. Unlike the LDL receptor, expression of CD36 is upregulated by this pro-atherogenic particle, and binding and uptake perpetuates a cycle of lipid accumulation and receptor expression. This effect is, in part, mediated by the transcription factor, peroxisome proliferator activated receptor-gamma (PPAR gamma), and its ligands. We have found that specific inhibitors of protein kinase C (PKC) reduce basal mRNA expression of CD36 and block induction of CD36 mRNA and protein by oxidized LDL (OxLDL) and a PPAR gamma ligand. In addition, PKC inhibitors block both PPAR gamma mRNA and protein expression. These results suggest that activation of CD36 gene expression by OxLDL involves activation and translocation of PKC with subsequent PPAR gamma activation. More recently, we have generated a mouse null for CD36, and crossed it with the atherogenic Apo E null strain. Evaluation of lesion development in these animals will allow us to assess the in vivo contribution of CD36 to the pathogenesis of atherosclerosis.


Subject(s)
Arteriosclerosis/physiopathology , CD36 Antigens/physiology , Membrane Proteins , Receptors, Immunologic/physiology , Receptors, Lipoprotein , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Arteriosclerosis/genetics , CD36 Antigens/genetics , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Immunologic/genetics , Receptors, Scavenger , Scavenger Receptors, Class B , Transcription Factors/genetics
16.
J Lipid Res ; 41(5): 688-96, 2000 May.
Article in English | MEDLINE | ID: mdl-10787429

ABSTRACT

CD36, a class B scavenger receptor, is a macrophage receptor for oxidized low density lipoprotein (OxLDL) and may play a critical role in atherosclerotic foam cell formation. We have previously demonstrated that OxLDL, macrophage-colony stimulating factor (M-CSF), and interleukin-4 (IL-4) enhanced expression of CD36. The effect of OxLDL on CD36 is due, in part, to its ability to activate the transcription factor, PPAR-gamma (peroxisome proliferator activated receptor-gamma). Other PPAR-gamma ligands (15-deoxyDelta(12,14) prostaglandin J(2) (15d-PGJ(2)) and the thiazolidinedione class of antidiabetic drugs) also increase CD36 expression. We have now evaluated signaling pathways involved in the induction of CD36. Treatment of RAW264.7 cells (a murine macrophage cell line) with protein kinase C (PKC) activators (diacylglycerol and ingenol) up-regulated CD36 mRNA expression. Specific inhibitors of PKC reduced CD36 expression in a time-dependent manner, while protein kinase A (PKA) and cyclic AMP agonists had no effect on CD36 mRNA expression. PKC inhibitors reduced basal expression of CD36 and blocked induction of CD36 mRNA by 15d-PGJ(2), OxLDL and IL-4. In addition, PKC inhibitors decreased both PPAR-gamma mRNA and protein expression and blocked induction of CD36 protein surface expression by OxLDL and 15d-PGJ(2) in human monocytes, as determined by FACS. 15d-PGJ(2) had no effect on translocation of PKC-alpha from the cytosol to the plasma membrane. These results demonstrate that two divergent physiological or pathophysiological agonists utilize a common pathway to up-regulate of CD36 gene expression. This pathway involves initial activation of PKC with subsequent PPAR-gamma activation. Defining these signaling pathways is critical for understanding and modulating expression of this scavenger receptor pathway.


Subject(s)
CD36 Antigens/genetics , Interleukin-4/pharmacology , Lipoproteins, LDL/pharmacology , Protein Kinase C/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , DNA, Complementary/genetics , Enzyme Activation , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , Mice , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Protein Kinase C/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Transcription Factors/agonists , Transcription Factors/genetics
17.
J Clin Invest ; 105(8): 1049-56, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10772649

ABSTRACT

Macrophage scavenger receptors have been implicated as key players in the pathogenesis of atherosclerosis. To assess the role of the class B scavenger receptor CD36 in atherogenesis, we crossed a CD36-null strain with the atherogenic apo E-null strain and quantified lesion development. There was a 76.5% decrease in aortic tree lesion area (Western diet) and a 45% decrease in aortic sinus lesion area (normal chow) in the CD36-apo E double-null mice when compared with controls, despite alterations in lipoprotein profiles that often correlate with increased atherogenicity. Macrophages derived from CD36-apo E double-null mice bound and internalized more than 60% less copper-oxidized LDL and LDL modified by monocyte-generated reactive nitrogen species. A similar inhibition of in vitro lipid accumulation and foam cell formation after exposure to these ligands was seen. These results support a major role for CD36 in atherosclerotic lesion development in vivo and suggest that blockade of CD36 can be protective even in more extreme proatherogenic circumstances.


Subject(s)
Arteriosclerosis/prevention & control , CD36 Antigens/physiology , Receptors, Immunologic/physiology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Arteriosclerosis/etiology , CD36 Antigens/genetics , Cells, Cultured , Cholesterol/blood , Female , Lipoproteins, LDL/metabolism , Macrophages, Peritoneal/cytology , Male , Mice , Mice, Inbred C57BL , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Receptors, Scavenger , Triglycerides/blood , Weight Gain
18.
J Clin Invest ; 105(8): 1095-108, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10772654

ABSTRACT

The oxidative conversion of LDL into an atherogenic form is considered a pivotal event in the development of cardiovascular disease. Recent studies have identified reactive nitrogen species generated by monocytes by way of the myeloperoxidase-hydrogen peroxide-nitrite (MPO-H(2)O(2)-NO(2)(-)) system as a novel mechanism for converting LDL into a high-uptake form (NO(2)-LDL) for macrophages. We now identify the scavenger receptor CD36 as the major receptor responsible for high-affinity and saturable cellular recognition of NO(2)-LDL by murine and human macrophages. Using cells stably transfected with CD36, CD36-specific blocking mAbs, and CD36-null macrophages, we demonstrated CD36-dependent binding, cholesterol loading, and macrophage foam cell formation after exposure to NO(2)-LDL. Modification of LDL by the MPO-H(2)O(2)-NO(2)(-) system in the presence of up to 80% lipoprotein-deficient serum (LPDS) still resulted in the conversion of the lipoprotein into a high-uptake form for macrophages, whereas addition of less than 5% LPDS totally blocked Cu(2+)-catalyzed LDL oxidation and conversion into a ligand for CD36. Competition studies demonstrated that lipid oxidation products derived from 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphocholine can serve as essential moieties on NO(2)-LDL recognized by CD36. Collectively, these results suggest that MPO-dependent conversion of LDL into a ligand for CD36 is a likely pathway for generating foam cells in vivo. MPO secreted from activated phagocytes may also tag phospholipid-containing targets for removal by CD36-positive cells.


Subject(s)
CD36 Antigens/metabolism , Lipoproteins, LDL/metabolism , Monocytes/metabolism , Nitrogen Dioxide/metabolism , Receptors, Immunologic/metabolism , Receptors, LDL/metabolism , Animals , CHO Cells , Cell Line , Cricetinae , Glucose Oxidase/metabolism , Humans , Hydrogen Peroxide/metabolism , Ligands , Mice , Mice, Inbred C57BL , Monocytes/cytology , Peroxidase/metabolism , Receptors, Scavenger , Time Factors
19.
Nat Med ; 6(1): 41-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10613822

ABSTRACT

Thrombospondin-1 (TSP-1) is a naturally occurring inhibitor of angiogenesis that limits vessel density in normal tissues and curtails tumor growth. Here, we show that the inhibition of angiogenesis in vitro and in vivo and the induction of apoptosis by thrombospondin-1 all required the sequential activation of CD36, p59fyn, caspase-3 like proteases and p38 mitogen-activated protein kinases. We also detected increased endothelial cell apoptosis in situ at the margins of tumors in mice treated with thrombospondin-1. These results indicate that thrombospondin-1, and possibly other broad-spectrum natural inhibitors of angiogenesis, act in vivo by inducing receptor-mediated apoptosis in activated microvascular endothelial cells.


Subject(s)
Apoptosis/physiology , CD36 Antigens/physiology , Endothelium, Vascular/physiology , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins/physiology , Thrombospondin 1/pharmacology , Angiostatins , Animals , Apoptosis/drug effects , CD36 Antigens/genetics , Caspases/metabolism , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Knockout , Microcirculation , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Plasminogen/pharmacology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-fyn , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases
20.
Blood ; 94(12): 4011-9, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10590044

ABSTRACT

Homing of hematopoietic stem cells to the bone marrow (BM) involves sequential interaction with adhesion molecules expressed on BM endothelium (BMEC) and chemokine stromal derived factor-1 (SDF-1). However, the mechanism whereby adhesion molecules regulate the SDF-1-induced transendothelial migration process is not known. E-selectin is an endothelial-specific selectin that is constitutively expressed by the BMEC in vivo. Hence, we hypothesized that E-selectin may mediate SDF-1-induced transendothelial migration of CD34(+) cells. We show that CD34(+) cells express both E-selectin ligand and fucosyltransferase-VII (FucT-VII). Soluble E-selectin-IgG chimera binds avidly to 75% +/- 10% of CD34(+) cells composed mostly of progenitors and cells with long-term culture-initiating cell (LTC-IC) potential. To assess the functional capacity of E-selectin to mediate CD34(+) cell migration in a transendothelial migration system, CD34(+) cells were placed on transwell plates coated with interleukin-1beta-activated BMEC. In the absence of SDF-1, there was spontaneous migration of 7.0% +/- 1.4% of CD34(+) cells and 14.1% +/- 2.2% of LTC-IC. SDF-1 induced migration of an additional 23.0% +/- 4.4% of CD34(+) cells and 17.6% +/- 3.6% of LTC-IC. Blocking MoAb to E-selectin inhibited SDF-1-induced migration of CD34(+) cells by 42.0% +/- 2.5% and LTC-IC by 90.9% +/- 16.6%. To define the mechanism of constitutive expression of E-selectin by the BMEC in vivo, we have found that vascular endothelial growth factor (VEGF(165)) induces E-selectin expression by cultured endothelial cells. VEGF-stimulated endothelial cells support transendothelial migration of CD34(+) cells that could be blocked by MoAb to E-selectin. These results suggest that trafficking of subsets of CD34(+) cells with LTC-IC potential is determined in part by sequential interactions with E-selectin and SDF-1.


Subject(s)
Cell Communication/physiology , Chemokines, CXC/physiology , E-Selectin/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Cell Movement/physiology , Cells, Cultured , Chemokine CXCL12 , Chemokines/physiology , Fetal Blood , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...