Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 482(3): 482-490, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28212735

ABSTRACT

High fidelity of biological systems is frequently achieved by duplication of the essential intracellular machineries or, removal of the entire cell, which becomes unnecessary or even harmful in altered physiological environments. Carefully controlled removal of these cells, without damaging normal cells, requires precise signaling, and is critical to maintaining homeostasis. This review describes how two anionic phospholipids - phosphatidylserine (PS) and cardiolipin (CL) - residing in distinct compartments of the cell, signal removal of "the unnecessary" using several uniform principles. One of these principles is realized by collapse of inherent transmembrane asymmetry and the externalization of the signal on the outer membrane surface - mitochondria for CL and the plasma membrane for PS - to trigger mitophagy and phagocytosis, respectively. Release from damaged cells of intracellular structures with externalized CL or externalized PS triggers their elimination by phagocytosis. Another of these principles is realized by oxidation of polyunsaturated species of CL and PS. Highly specific oxidation of CL by cytochrome c serves as a signal for mitochondria-dependent apoptosis, while oxidation of externalized PS improves its effectiveness to trigger phagocytosis of effete cells.


Subject(s)
Phospholipids/metabolism , Animals , Apoptosis , Cardiolipins/metabolism , Extracellular Space/metabolism , Humans , Intracellular Space/metabolism , Mitophagy , Oxidation-Reduction , Phagocytosis , Phosphatidylserines/metabolism , Signal Transduction
2.
Sci Signal ; 8(395): ra95, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26396268

ABSTRACT

Among the distinct molecular signatures present in the mitochondrion is the tetra-acylated anionic phospholipid cardiolipin, a lipid also present in primordial, single-cell bacterial ancestors of mitochondria and multiple bacterial species today. Cardiolipin is normally localized to the inner mitochondrial membrane; however, when cardiolipin becomes externalized to the surface of dysregulated mitochondria, it promotes inflammasome activation and stimulates the elimination of damaged or nonfunctional mitochondria by mitophagy. Given the immunogenicity of mitochondrial and bacterial membranes that are released during sterile and pathogen-induced trauma, we hypothesized that cardiolipins might function as "eat me" signals for professional phagocytes. In experiments with macrophage cell lines and primary macrophages, we found that membranes with mitochondrial or bacterial cardiolipins on their surface were engulfed through phagocytosis, which depended on the scavenger receptor CD36. Distinct from this process, the copresentation of cardiolipin with the Toll-like receptor 4 (TLR4) agonist lipopolysaccharide dampened TLR4-stimulated production of cytokines. These data suggest that externalized, extracellular cardiolipins play a dual role in host-host and host-pathogen interactions by promoting phagocytosis and attenuating inflammatory immune responses.


Subject(s)
CD36 Antigens/immunology , Cardiolipins/immunology , Macrophages/immunology , Phagocytosis , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Cell Line, Tumor , Humans
3.
ACS Nano ; 8(6): 5610-21, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24871084

ABSTRACT

In contrast to short-lived neutrophils, macrophages display persistent presence in the lung of animals after pulmonary exposure to carbon nanotubes. While effective in the clearance of bacterial pathogens and injured host cells, the ability of macrophages to "digest" carbonaceous nanoparticles has not been documented. Here, we used chemical, biochemical, and cell and animal models and demonstrated oxidative biodegradation of oxidatively functionalized single-walled carbon nanotubes via superoxide/NO* → peroxynitrite-driven oxidative pathways of activated macrophages facilitating clearance of nanoparticles from the lung.


Subject(s)
Lung/drug effects , Macrophages/drug effects , Nanotechnology/methods , Nanotubes, Carbon/chemistry , Peroxynitrous Acid/chemistry , Superoxides/chemistry , Acoustics , Animals , Biocompatible Materials/chemistry , Bronchoalveolar Lavage , Carbon/chemistry , Humans , Inflammation/metabolism , Lung/pathology , Macrophages/metabolism , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Nitric Oxide/chemistry , Oxygen/chemistry , Rats
4.
Nat Cell Biol ; 15(10): 1197-1205, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24036476

ABSTRACT

Recognition of injured mitochondria for degradation by macroautophagy is essential for cellular health, but the mechanisms remain poorly understood. Cardiolipin is an inner mitochondrial membrane phospholipid. We found that rotenone, staurosporine, 6-hydroxydopamine and other pro-mitophagy stimuli caused externalization of cardiolipin to the mitochondrial surface in primary cortical neurons and SH-SY5Y cells. RNAi knockdown of cardiolipin synthase or of phospholipid scramblase-3, which transports cardiolipin to the outer mitochondrial membrane, decreased the delivery of mitochondria to autophagosomes. Furthermore, we found that the autophagy protein microtubule-associated-protein-1 light chain 3 (LC3), which mediates both autophagosome formation and cargo recognition, contains cardiolipin-binding sites important for the engulfment of mitochondria by the autophagic system. Mutation of LC3 residues predicted as cardiolipin-interaction sites by computational modelling inhibited its participation in mitophagy. These data indicate that redistribution of cardiolipin serves as an 'eat-me' signal for the elimination of damaged mitochondria from neuronal cells.


Subject(s)
Cardiolipins/metabolism , Mitochondrial Membranes/metabolism , Mitophagy/physiology , Neurons/physiology , Signal Transduction , Amino Acid Sequence , Animals , Autophagy/drug effects , Biological Transport/drug effects , Cardiolipins/genetics , Cell Line, Tumor , Cells, Cultured , Gene Knockdown Techniques , HeLa Cells , Humans , Mitochondria/drug effects , Mitophagy/drug effects , Models, Molecular , Molecular Sequence Data , Neurons/drug effects , Oxidopamine/pharmacology , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Rotenone/pharmacology , Uncoupling Agents/pharmacology
5.
ACS Nano ; 6(5): 4147-56, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22463369

ABSTRACT

The pulmonary route represents one of the most important portals of entry for nanoparticles into the body. However, the in vivo interactions of nanoparticles with biomolecules of the lung have not been sufficiently studied. Here, using an established mouse model of pharyngeal aspiration of single-walled carbon nanotubes (SWCNTs), we recovered SWCNTs from the bronchoalveolar lavage fluid (BALf), purified them from possible contamination with lung cells, and examined the composition of phospholipids adsorbed on SWCNTs by liquid chromatography mass spectrometry (LC-MS) analysis. We found that SWCNTs selectively adsorbed two types of the most abundant surfactant phospholipids: phosphatidylcholines (PC) and phosphatidylglycerols (PG). Molecular speciation of these phospholipids was also consistent with pulmonary surfactant. Quantitation of adsorbed lipids by LC-MS along with the structural assessments of phospholipid binding by atomic force microscopy and molecular modeling indicated that the phospholipids (∼108 molecules per SWCNT) formed an uninterrupted "coating" whereby the hydrophobic alkyl chains of the phospholipids were adsorbed onto the SWCNT with the polar head groups pointed away from the SWCNT into the aqueous phase. In addition, the presence of surfactant proteins A, B, and D on SWCNTs was determined by LC-MS. Finally, we demonstrated that the presence of this surfactant coating markedly enhanced the in vitro uptake of SWCNTs by macrophages. Taken together, this is the first demonstration of the in vivo adsorption of the surfactant lipids and proteins on SWCNTs in a physiologically relevant animal model.


Subject(s)
Lipids/chemistry , Lung/metabolism , Nanotubes, Carbon , Pharynx/metabolism , Surface-Active Agents/chemistry , Adsorption , Animals , Mice , Respiratory Aspiration
6.
Neoplasia ; 13(3): 286-98, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21390191

ABSTRACT

In the United States, more than 40% of cancer patients develop brain metastasis. The median survival for untreated patients is 1 to 2 months, which may be extended to 6 months with conventional radiotherapy and chemotherapy. The growth and survival of metastasis depend on the interaction of tumor cells with host factors in the organ microenvironment. Brain metastases are surrounded and infiltrated by activated astrocytes and are highly resistant to chemotherapy. We report here that coculture of human breast cancer cells or lung cancer cells with murine astrocytes (but not murine fibroblasts) led to the up-regulation of survival genes, including GSTA5, BCL2L1, and TWIST1, in the tumor cells. The degree of up-regulation directly correlated with increased resistance to all tested chemotherapeutic agents. We further show that the up-regulation of the survival genes and consequent resistance are dependent on the direct contact between the astrocytes and tumor cells through gap junctions and are therefore transient. Knocking down these genes with specific small interfering RNA rendered the tumor cells sensitive to chemotherapeutic agents. These data clearly demonstrate that host cells in the microenvironment influence the biologic behavior of tumor cells and reinforce the contention that the organ microenvironment must be taken into consideration during the design of therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Astrocytes/metabolism , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis , Astrocytes/pathology , Biomarkers, Tumor/metabolism , Blotting, Western , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Profiling , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Nude , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Twist-Related Protein 1/antagonists & inhibitors , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Up-Regulation , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism
7.
Neoplasia ; 12(9): 748-54, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20824051

ABSTRACT

Brain metastases are highly resistant to chemotherapy. Metastatic tumor cells are known to exploit the host microenvironment for their growth and survival. We report here that melanoma brain metastases are surrounded and infiltrated by activated astrocytes, and we hypothesized that these astrocytes can play a role similar to their established ability to protect neurons from apoptosis. In coculture experiments, astrocytes, but not fibroblasts, reduced apoptosis in human melanoma cells treated with various chemotherapeutic drugs. This chemoprotective effect was dependent on physical contact and gap junctional communication between astrocytes and tumor cells. Moreover, the protective effect of astrocytes resulted from their sequestering calcium from the cytoplasm of tumor cells. These data suggest that brain tumors can, in principle, harness the neuroprotective effects of reactive astrocytes for their own survival and implicate a heretofore unrecognized mechanism for resistance in brain metastasis that might be of relevance in the clinic.


Subject(s)
Antineoplastic Agents/therapeutic use , Astrocytes/physiology , Calcium/metabolism , Connexins/physiology , Drug Resistance, Neoplasm , Animals , Antineoplastic Agents/pharmacology , Astrocytes/metabolism , Cell Communication/physiology , Cell Line, Tumor , Cells, Cultured , Connexins/metabolism , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/physiology , Humans , Intracellular Fluid/metabolism , Male , Mice , Mice, Nude , Mice, Transgenic , Models, Biological , NIH 3T3 Cells
8.
Mol Cells ; 30(2): 93-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20799011

ABSTRACT

The process of metastasis consists of a series of sequential, selective steps that few cells can complete. The outcome of cancer metastasis depends on multiple interactions between metastatic cells and homeostatic mechanisms that are unique to one or another organ microenvironment. The specific organ microenvironment determines the extent of cancer cell proliferation, angiogenesis, invasion and survival. Many lung cancer, breast cancer, and melanoma patients develop fatal brain metastases that do not respond to therapy. The blood-brain barrier is intact in and around brain metastases that are smaller than 0.25 mm in diameter. Although the blood-brain barrier is leaky in larger metastases, the lesions are resistant to many chemotherapeutic drugs. Activated astrocytes surround and infiltrate brain metastases. The physiological role of astrocytes is to protect against neurotoxicity. Our current data demonstrate that activated astrocytes also protect tumor cells against chemotherapeutic drugs.


Subject(s)
Brain Neoplasms/secondary , Tumor Microenvironment , Animals , Apoptosis , Astrocytes/pathology , Blood-Brain Barrier/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Disease Models, Animal , Humans , Neovascularization, Pathologic
9.
J Biol Chem ; 284(42): 28845-55, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19703903

ABSTRACT

The cellular and molecular pathways that regulate platelet activation, blood coagulation, and inflammation are emerging as critical players in cancer progression and metastasis. Here, we demonstrate a novel signaling mechanism whereby protease-activated receptor 1 (PAR1) mediates expression of melanoma cell adhesion molecule MCAM/MUC18 (MUC18), a critical marker of melanoma metastasis, via activation of platelet-activating factor receptor (PAFR) and cAMP-responsive element-binding protein (CREB). We found that PAR1 silencing with small hairpin RNA inhibits MUC18 expression in metastatic melanoma cells by inhibiting CREB phosphorylation, activity, and binding to the MUC18 promoter. We further demonstrate that the PAF/PAFR pathway mediates MUC18 expression downstream of PAR1. Indeed, PAR1 silencing down-regulates PAFR expression and PAF production, PAFR silencing blocks MUC18 expression, and re-expression of PAFR in PAR1-silenced cells rescues MUC18 expression. We further demonstrate that the PAR1-PAFR-MUC18 pathway mediates melanoma cell adhesion to microvascular endothelial cells, transendothelial migration, and metastatic retention in the lungs. Rescuing PAFR expression in PAR1-silenced cells fully restores metastatic phenotype of melanoma, indicating that PAFR plays critical role in the molecular mechanism of PAR1 action. Our results link the two pro-inflammatory G-protein-coupled receptors, PAR1 and PAFR, with the metastatic dissemination of melanoma and suggest that PAR1, PAFR, and MUC18 are attractive therapeutic targets for preventing melanoma metastasis.


Subject(s)
Melanoma/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptor, PAR-1/metabolism , Receptors, G-Protein-Coupled/metabolism , Skin Neoplasms/metabolism , CD146 Antigen/metabolism , CREB-Binding Protein/metabolism , Cell Line, Tumor , Gene Silencing , Humans , Melanoma/pathology , Neoplasm Metastasis , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Skin Neoplasms/pathology , Transcription Factors/metabolism
10.
J Biol Chem ; 284(34): 22512-6, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19561081

ABSTRACT

One of the hallmarks of apoptosis is the redistribution of phosphatidylserine (PS) from the inner-to-outer plasma membrane (PM) leaflet, where it functions as a ligand for phagocyte recognition and the suppression of inflammatory responses. The mechanism by which apoptotic cells externalize PS has been assumed to involve "scramblases" that randomize phospholipids across the PM bilayer. These putative activities, however, have not been unequivocally proven to be responsible for the redistribution of lipids. Because elevated cytosolic Ca(2+) is critical to this process and is also required for activation of lysosome-PM fusion during membrane repair, we hypothesized that apoptosis could activate a "pseudo"-membrane repair response that results in the fusion of lysosomes with the PM. Using a membrane-specific probe that labels endosomes and lysosomes and fluorescein-labeled annexin 5 that labels PS, we show that the appearance of PS at the cell surface during apoptosis is dependent on the fusion of lysosomes with the PM, a process that is inhibited with the lysosomotrophe, chloroquine. We demonstrate that apoptotic cells evoke a persistent pseudo-membrane repair response that likely redistributes lysosomal-derived PS to the PM outer leaflet that leads to membrane expansion and the formation of apoptotic blebs. Our data suggest that inhibition of lysosome-PM fusion-dependent redistribution of PS that occurs as a result of chemotherapy- and radiotherapy-induced apoptosis will prevent PS-dependent anti-inflammatory responses that preclude the development of tumor- and patient-specific immune responses.


Subject(s)
Apoptosis/physiology , Cell Membrane/metabolism , Phosphatidylserines/metabolism , Animals , Cells, Cultured , Lysosomes/metabolism , Mice , Microscopy, Confocal
11.
J Biol Chem ; 284(11): 6918-23, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19126538

ABSTRACT

A hallmark of apoptotic cells is the Ca2+-dependent appearance of phosphatidylserine (PS) at the cell surface as a result of its redistribution from the inner-to-outer plasma membrane leaflet. Although endoplasmic reticulum and mitochondrial Ca2+ are known to participate in apoptosis, their role in PS externalization has not been established. In this study, several organelle-specific fluorescent markers and Ca2+-sensitive probes were used to identify the source of Ca2+ critical to PS externalization. By employing Rhod-2AM, fluorescein-labeled high molecular weight dextran, and Calcium Green 1, we provide evidence that lysosomes respond to apoptotic stimuli by releasing their luminal Ca2+ to the cytosol. Cells treated with the cytosolic phospholipase A2 inhibitor, cPLA2alpha, had no effect on caspase activation but exhibited a significant decrease in lysosomal Ca2+ release and externalization of PS in response to apoptotic stimuli. Similarly, cells depleted of lysosomal Ca2+ underwent programmed cell death yet failed to externalize PS. These data indicate that although Ca2+ release from other intracellular organelles to the cytosol is adequate for apoptosis, the release of Ca2+ from lysosomes is critical for PS externalization.


Subject(s)
Apoptosis/physiology , Calcium/metabolism , Cytosol/metabolism , Lysosomes/metabolism , Phosphatidylserines/metabolism , Animals , Apoptosis/drug effects , Caspases/metabolism , Cell Membrane/metabolism , Cells, Cultured , Endoplasmic Reticulum/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Group IV Phospholipases A2/antagonists & inhibitors , Group IV Phospholipases A2/metabolism , Mice , Molecular Probes/pharmacology
12.
J Biol Chem ; 283(7): 3761-6, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18073216

ABSTRACT

The recognition and removal of apoptotic cells is critical to development, tissue homeostasis, and the resolution of inflammation. Many studies have shown that phagocytosis is regulated by signaling mechanisms that involve distinct ligand-receptor interactions that drive the engulfment of apoptotic cells. Studies from our laboratory have shown that the plasma protein beta-2-glycoprotein 1 (beta2GP1), a member of the short consensus repeat superfamily, binds phosphatidylserine-containing vesicles and apoptotic cells and promotes their bridging and subsequent engulfment by phagocytes. The phagocyte receptor for the protein/apoptotic cell complex, however, is unknown. Here we report that a member of the low density lipoprotein receptor-related protein family on phagocytes binds and facilitates engulfment of beta2GP1-phosphatidylserine and beta2GP1-apoptotic cell complexes. Using recombinant beta2GP1, we also show that beta2GP1-dependent uptake is mediated by bridging of the target cell to the phagocyte through the protein C- and N-terminal domains, respectively.


Subject(s)
Apoptosis , Macrophages/physiology , Receptors, LDL/metabolism , beta 2-Glycoprotein I/physiology , Cell Line, Tumor , Humans , Microscopy, Fluorescence , Phagocytosis
13.
Am J Pathol ; 171(5): 1659-69, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17872974

ABSTRACT

beta-2-Glycoprotein 1, an abundant plasma glycoprotein, binds anionic cell surfaces and functions as a regulator of thrombosis. Here, we show that cleavage of the kringle domain at Lys317/Thr318 switches its function to a regulator of angiogenesis. In vitro, the cleaved protein specifically inhibited the proliferation and migration of endothelial cells. The protein was without effect on preformed endothelial cell tubes. In vivo, the cleaved protein inhibited neovascularization into subcutaneously implanted Matrigel and Gelfoam sponge implants and the growth of orthotopically injected tumors. Collectively, these data indicate that plasmin-cleaved beta-2-glycoprotein 1 is a potent antiangiogenic and antitumor molecule of potential therapeutic significance.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Fibrinolysin/physiology , Neovascularization, Pathologic , beta 2-Glycoprotein I/physiology , Amino Acid Sequence , Animals , Cell Movement/drug effects , Cells, Cultured , Collagen , Drug Combinations , Endothelial Cells/drug effects , Endothelial Cells/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Gelatin Sponge, Absorbable , Kringles , Laminin , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Neoplasm Transplantation , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Proteoglycans , beta 2-Glycoprotein I/pharmacology
14.
J Biol Chem ; 282(25): 18357-18364, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17470427

ABSTRACT

The regulated loss of plasma membrane phosphatidylserine (PS) asymmetry is critical to many biological processes. In particular, the appearance of PS at the cell surface, a hallmark of apoptosis, prepares the dying cell for engulfment and elimination by phagocytes. While it is well established that PS externalization is regulated by activation of a calcium-dependent phospholipid scramblase activity in concert with inactivation of the aminophospholipid translocase, there is no evidence indicating that these processes are triggered and regulated by apoptotic regulatory mechanisms. Using a novel model system, we show that PS externalization is inducible, reversible, and independent of cytochrome c release, caspase activation, and DNA fragmentation. Additional evidence is presented indicating that the outward movement of plasma membrane PS requires sustained elevation in cytosolic Ca2+ in concert with inactivation of the aminophospholipid translocase and is inhibited by calcium channel blockers.


Subject(s)
Apoptosis , Cell Membrane/metabolism , Phosphatidylserines/chemistry , Calcium/metabolism , Cytochromes c/metabolism , Cytosol/metabolism , DNA Fragmentation , Erythrocytes/metabolism , Humans , Jurkat Cells , K562 Cells , Lipids/chemistry , Models, Biological , Phagocytes/metabolism , Phospholipid Transfer Proteins/metabolism
15.
Biochem J ; 386(Pt 2): 271-9, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15527422

ABSTRACT

The plasma protein beta2GPI (beta2-glycoprotein I) has been proposed to mediate phagocytosis of apoptotic cells and to play a role in the antiphospholipid syndrome. This suggestion is based mainly on the presumption that beta2GPI has an appreciable interaction with PS (phosphatidylserine)-exposing cell membranes. However, quantitative data on the binding of beta2GPI to PS-exposing cells under physiologically relevant conditions are scarce and conflicting. Therefore we evaluated the binding of beta2GPI to PS-expressing blood platelets. Flow cytometry showed that binding of beta2GPI is negligible at physiological ionic strength, in contrast with significant binding occurring at low ionic strength. Binding parameters of beta2GPI and (for comparison) prothrombin were quantified by ellipsometric measurement of protein depletion from the supernatant following incubation with platelets. At low ionic strength (20 mM NaCl, no CaCl2), a dissociation constant (K(d)) of 0.2 microM was found for beta2GPI, with 7.4x10(5) binding sites per platelet. Under physiologically relevant conditions (120 mM NaCl and 3 mM CaCl2), binding of beta2GPI was not detectable (extrapolated K(d)>80 microM). Prothrombin binding (at 3 mM CaCl2) was much less affected by ionic strength: K(d) values of 0.5 and 1.4 muM were observed at 20 and 120 mM NaCl respectively. The low affinity and the presence of many lipid-binding proteins in plasma that can compete with the binding of beta2GPI suggest that only a small fraction (<5%) of the binding sites on PS-exposing blood cells are likely to be occupied by beta2GPI. These findings are discussed in relation to the alleged (patho-)physiological functions of beta2GPI.


Subject(s)
Blood Platelets/chemistry , Glycoproteins/metabolism , Phosphatidylserines/metabolism , Prothrombin/metabolism , Binding Sites , Blood Platelets/drug effects , Blood Platelets/metabolism , Flow Cytometry/methods , Humans , Ionomycin/pharmacology , Osmolar Concentration , Protein Binding , beta 2-Glycoprotein I
16.
Annu Rev Physiol ; 65: 701-34, 2003.
Article in English | MEDLINE | ID: mdl-12471163

ABSTRACT

Maintenance of membrane lipid asymmetry is a dynamic process that influences many events over the lifespan of the cell. With few exceptions, most cells restrict the bulk of the aminophospholipids to the inner membrane leaflet by means of specific transporters. Working in concert with each other, these proteins correct for sporadic incursions of the aminophospholipids to the outer membrane leaflet as a result of bilayer imbalances created by various cellular events. A shift in the relative contribution in each of these activities can result in sustained exposure of the aminophospholipids at the cell surface, which allows capture of the cells by phagocytes before the integrity of the plasma membrane is compromised. The absence of an efficient recognition and elimination mechanism can result in uncontrolled and persistent presentation of self-antigens to the immune system, with development of autoimmune syndromes. To prevent this, phagocytes have developed a diverse array of distinct and redundant receptor systems that drive the postphagocytic events along pathways that facilitate cross-talk between the homeostatic and the immune systems. In this work, we review the basis for the proposed mechanism(s) by which apoptotic ligands appear on the target cell surface and the phagocyte receptors that recognize these moieties.


Subject(s)
Apoptosis/physiology , Phosphatidylethanolamines/metabolism , Phosphatidylserines/metabolism , Animals , Cell Membrane/metabolism , Humans , Phagocytosis/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...