Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Angiogenesis ; 25(3): 397-410, 2022 08.
Article in English | MEDLINE | ID: mdl-35212873

ABSTRACT

Anthrax protective antigen (PA) is a potent inhibitor of pathological angiogenesis with an unknown mechanism. In anthrax intoxication, PA interacts with capillary morphogenesis gene 2 (CMG2) and tumor endothelial marker 8 (TEM8). Here, we show that CMG2 mediates the antiangiogenic effects of PA and is required for growth-factor-induced chemotaxis. Using specific inhibitors of CMG2 and TEM8 interaction with natural ligand, as well as mice with the CMG2 or TEM8 transmembrane and intracellular domains disrupted, we demonstrate that inhibiting CMG2, but not TEM8 reduces growth-factor-induced angiogenesis in the cornea. Furthermore, the antiangiogenic effect of PA was abolished when the CMG2, but not the TEM8, gene was disrupted. Binding experiments demonstrated a broad ligand specificity for CMG2 among extracellular matrix (ECM) proteins. Ex vivo experiments demonstrated that CMG2 (but not TEM8) is required for PA activity in human dermal microvascular endothelial cell (HMVEC-d) network formation assays. Remarkably, blocking CMG2-ligand binding with PA or CRISPR knockout abolishes endothelial cell chemotaxis but not chemokinesis in microfluidic migration assays. These effects are phenocopied by Rho inhibition. Because CMG2 mediates the chemotactic response of endothelial cells to peptide growth factors in an ECM-dependent fashion, CMG2 is well-placed to integrate growth factor and ECM signals. Thus, CMG2 targeting is a novel way to inhibit angiogenesis.


Subject(s)
Chemotaxis , Endothelial Cells , Neovascularization, Pathologic , Receptors, Peptide , Animals , Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Ligands , Mice , Receptors, Peptide/genetics , Receptors, Peptide/metabolism
2.
Bioorg Med Chem Lett ; 22(18): 5885-8, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22910038

ABSTRACT

Targeting and inhibiting CMG2 (Capillary Morphogenesis Gene protein 2) represents a new strategy for therapeutic agents for cancer and retinal diseases due to CMG2's role in blood vessel growth (angiogenesis). A high throughput FRET (Förster Resonance Energy Transfer) assay was developed for the identification of CMG2 inhibitors as anti-angiogenetic agents. Bioassay-guided separation led to the isolation and identification of two new compounds (1 and 2) from CR252M, an endophytic fungus Coccomyces proteae collected from a Costa Rican rainforest, and one known compound (3) from CR1207B (Aurapex penicillata). Secondary in vitro assays indicated anti-angiogenic activity. Compound 3 inhibited the endothelial cell migration at 52 µM, but did not show any endothelial cell antiproliferative effect at 156 µM. The structure of the two new compounds, A (1) and B (2), were elucidated on the basis of extensive spectroscopic analysis, including 1D and 2D NMR experiments.


Subject(s)
Ascomycota/chemistry , Membrane Proteins/antagonists & inhibitors , Phenols/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Costa Rica , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Humans , Molecular Structure , Phenols/chemistry , Phenols/isolation & purification , Receptors, Peptide , Stereoisomerism , Structure-Activity Relationship
3.
J Biomed Biotechnol ; 2010: 107859, 2010.
Article in English | MEDLINE | ID: mdl-20368775

ABSTRACT

Proteomic approaches have proven powerful at identifying large numbers of proteins, but there are fewer reports of functional characterization of proteins in biological tissues. Here, we describe an experimental approach that fractionates proteins released from human platelets, linking bioassay activity to identity. We used consecutive orthogonal separation platforms to ensure sensitive detection: (a) ion-exchange of intact proteins, (b) SDS-PAGE separation of ion-exchange fractions and (c) HPLC separation of tryptic digests coupled to electrospray tandem mass spectrometry. Migration of THP-1 monocytes in response to complete or fractionated platelet releasate was assessed and located to just one of the forty-nine ion-exchange fractions. Over 300 proteins were identified in the releasate, with a wide range of annotated biophysical and biochemical properties, in particular platelet activation, adhesion, and wound healing. The presence of PEDF and involucrin, two proteins not previously reported in platelet releasate, was confirmed by western blotting. Proteins identified within the fraction with monocyte promigratory activity and not in other inactive fractions included vimentin, PEDF, and TIMP-1. We conclude that this analytical platform is effective for the characterization of complex bioactive samples.


Subject(s)
Blood Platelets/chemistry , Blood Proteins/chemistry , Complex Mixtures/analysis , Proteomics/methods , Blood Platelets/metabolism , Blood Proteins/metabolism , Blotting, Western , Cell Movement , Chromatography, Ion Exchange , Humans , Monocytes/metabolism , Organ Specificity , Reproducibility of Results
4.
ACS Chem Biol ; 15(2): 587-596, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32003961

ABSTRACT

Capillary Morphogenesis Gene 2 protein (CMG2) is a transmembrane, integrin-like receptor and the primary receptor for the anthrax toxin. CMG2 also plays a role in angiogenic processes. However, the molecular mechanism that mediates the observed CMG2-related angiogenic effects is not fully elucidated. Previous studies have reported that CMG2 binds type IV collagen (Col-IV), a vital component of the vascular basement membrane, as well as other ECM proteins. Here, we further characterize the interaction between CMG2 and individual peptides from Col-IV and explore the effects of this interaction on angiogenesis. Using a peptide array, we observed that CMG2 preferentially binds peptide fragments of the NC1 (noncollagenous domain 1) domains of Col-IV. These domains are also known as the fragments arresten (from the α1 chain) and canstatin (from the α2 chain) and have documented antiangiogenic properties. A second peptide array was probed to map a putative peptide-binding epitope onto the Col-IV structure. A top hit from the initial array, a canstatin-derived peptide, binds to the CMG2 ligand-binding von Willebrand factor A (vWA) domain with a submicromolar affinity (peptide S16, Kd = 400 ± 200 nM). This peptide competes with anthrax protective antigen (PA) for CMG2 binding and does not bind CMG2 in the presence of EDTA. Together these data suggest that, like PA, S16 interacts with CMG2 at the metal-ion dependent adhesion site (MIDAS) of its vWA domain. CMG2 specifically mediates endocytic uptake of S16; both CMG2-/- endothelial cells and WT cells treated with PA show markedly reduced S16 uptake. Furthermore, S16 dramatically reduces directional endothelial cell migration with no impact on cell proliferation. These data demonstrate that this canstatin-derived peptide acts via CMG2 to elicit a marked effect on a critical process required for angiogenesis.


Subject(s)
Collagen Type IV/metabolism , Endocytosis/physiology , Peptide Fragments/metabolism , Receptors, Peptide/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line, Tumor , Cell Movement/drug effects , Endothelial Cells/metabolism , Humans , Mice , Protein Binding , Protein Domains , Receptors, Peptide/chemistry
5.
Curr Eye Res ; 33(2): 177-84, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18293189

ABSTRACT

PURPOSE: The expression of cyclooxygenase-2 (COX-2) and its prognostic value in uveal melanoma was examined. METHODS: Paraffin-embedded sections from 32 clinicopathologically well-characterized cases of primary uveal melanoma were immunohistochemically stained for COX-2. COX-2 expression was evaluated in terms of both the intensity and the extent of staining for each tumor. A COX-2 score encompassing both intensity and extent was also calculated for each specimen. RESULTS: 29 specimens (90.6%) contained moderate or intense positive immunoreactivity for COX-2. A statistically significant association (p<0.05) between COX-2 expression (intensity and score) and metastatic death was established. CONCLUSION: Upregulation of COX-2 expression appears to be associated with poor prognosis in uveal melanoma.


Subject(s)
Choroid Neoplasms/enzymology , Cyclooxygenase 2/metabolism , Melanoma/enzymology , Adult , Aged , Aged, 80 and over , Choroid Neoplasms/mortality , Choroid Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Male , Melanoma/mortality , Melanoma/secondary , Middle Aged , Prognosis , Survival Rate
6.
Mol Vis ; 12: 405-14, 2006 Apr 20.
Article in English | MEDLINE | ID: mdl-16688111

ABSTRACT

PURPOSE: Cyclooxygenases (COX-1 and COX-2) and prostaglandins regulate angiogenesis in several settings, including cancer and ischemia. In the eye, both selective inhibitors of COX-2 and nonselective COX inhibitors are reported to suppress ischemia-related retinal angiogenesis. Such studies however, may be confounded by the nonspecific effects of inhibitors. METHODS: Mice lacking either the COX-1 (COX-1(-/-)) or COX-2 isoform (COX-2(-/-)) were employed in a model of oxygen-induced retinopathy. Vascular responses were examined by histology, isolectin B4 staining of the abluminal endothelium, and retinal fluorescein angiography. RESULTS: There was an increase in intravitreal endothelial nuclei in hyperoxia-treated mice compared to normoxic controls irrespective of the genotype. Quantitative analysis of fluorescein-perfused and isolectin B4-stained retinal angiograms at postnatal day 18 (P18) revealed similar global levels of neovascular tufts in hyperoxia-treated wild-type, COX-1(-/-), and COX-2(-/-) mice. However, hyperoxia-treated COX-2(-/-) mice had increased areas of retinal nonperfusion (29.2+/-1.9 compared to 16.3+/-2.7; n=6; p<0.001). COX-1 disruption had no effect (15.6+/-2.6; n=8). Platelet deposition within retinal vessels was increased in hyperoxia-treated COX-2(-/-) mice (p<0.05). CONCLUSIONS: Genetic disruption of a single COX isoform is not sufficient to prevent oxygen-induced retinopathy. COX-2 protects retinal vessels from thrombosis, limiting the area of retinal nonperfusion in oxygen-induced retinopathy.


Subject(s)
Cyclooxygenase 2/metabolism , Retinal Diseases/metabolism , Retinal Diseases/prevention & control , Thrombosis/prevention & control , Animals , Animals, Newborn , Blood Platelets/pathology , Cell Nucleus/ultrastructure , Cyclooxygenase 1/deficiency , Endothelial Cells/pathology , Fibrinogen/metabolism , Immunohistochemistry , Lectins , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen , Prostaglandins/biosynthesis , Regional Blood Flow , Retina/pathology , Retinal Diseases/chemically induced , Retinal Diseases/pathology , Retinal Vessels/metabolism , Retinal Vessels/physiopathology , Staining and Labeling , Vitreous Body/blood supply , Vitreous Body/pathology
7.
Mol Cancer Res ; 12(5): 754-64, 2014 May.
Article in English | MEDLINE | ID: mdl-24574516

ABSTRACT

UNLABELLED: The angiogenic switch, a rate-limiting step in tumor progression, has already occurred by the time most human tumors are detectable. However, despite significant study of the mechanisms controlling this switch, the kinetics and reversibility of the process have not been explored. The stability of the angiogenic phenotype was examined using an established human liposarcoma xenograft model. Nonangiogenic cells inoculated into immunocompromised mice formed microscopic tumors that remained dormant for approximately 125 days (vs. <40 days for angiogenic cells) whereupon the vast majority (>95%) initiated angiogenic growth with second-order kinetics. These original, clonally derived angiogenic tumor cells were passaged through four in vivo cycles. At each cycle, a new set of single-cell clones was established from the most angiogenic clone and characterized for in vivo for tumorigenic activity. A total of 132 single-cell clones were tested in the second, third, and fourth in vivo passage. Strikingly, at each passage, a portion of the single-cell clones formed microscopic, dormant tumors. Following dormancy, like the original cell line, these revertant tumors spontaneously switched to the angiogenic phenotype. Finally, revertant clones were transcriptionally profiled and their angiogenic output determined. Collectively, these data demonstrate that the angiogenic phenotype in tumors is malleable and can spontaneously revert to the nonangiogenic phenotype in a population of human tumor cells. IMPLICATIONS: Leveraging the rate of reversion to the nonangiogenic phenotype and tumor dormancy may be a novel anticancer strategy.


Subject(s)
Liposarcoma/blood supply , Liposarcoma/pathology , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Gene Expression , Heterografts , Humans , Male , Mice , Mice, SCID , Neovascularization, Pathologic/pathology , Phenotype
8.
J Med Chem ; 56(5): 1940-5, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23394144

ABSTRACT

Capillary morphogenesis gene 2 (CMG2) is a transmembrane extracellular matrix binding protein that is also an anthrax toxin receptor. We have shown that high-affinity CMG2 binders can inhibit angiogenesis and tumor growth. We recently described a high-throughput FRET assay to identify CMG2 inhibitors. We now report the serendipitous discovery that PGG (1,2,3,4,6-penta-O-galloyl-ß-D-glucopyranose) is a CMG2 inhibitor with antiangiogenic activity. PGG is a gallotannin produced by a variety of medicinal plants that exhibits a wide variety of antitumor and other activities. We find that PGG inhibits CMG2 with a submicromolar IC50 and it also inhibits the migration of human dermal microvascular endothelial cells at similar concentrations in vitro. Finally, oral or intraperitoneal administration of PGG inhibits angiogenesis in the mouse corneal micropocket assay in vivo. Together, these results suggest that a portion of the in vivo antitumor activity of PGG may be the result of antiangiogenic activity mediated by inhibition of CMG2.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Hydrolyzable Tannins/pharmacology , Neovascularization, Pathologic/drug therapy , Receptors, Peptide/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Humans , Mice , Receptors, Peptide/physiology
9.
J Biomol Screen ; 18(6): 714-25, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23479355

ABSTRACT

Tumor marker endothelial 8 (TEM8) is a receptor for the protective antigen (PA) component of anthrax toxin. TEM8 is upregulated on endothelial cells lining the blood vessels within tumors, compared with normal blood vessels. A number of studies have demonstrated a pivotal role for TEM8 in developmental and tumor angiogenesis. We have also shown that targeting the anthrax receptors with a mutated form of PA inhibits angiogenesis and tumor formation in vivo. Here we describe the development and testing of a high-throughput fluorescence resonance energy transfer assay to identify molecules that strongly inhibit the interaction of PA and TEM8. The assay we describe is sensitive and robust, with a Z' value of 0.8. A preliminary screen of 2310 known bioactive library compounds identified ebselen and thimerosal as inhibitors of the TEM8-PA interaction. These molecules each contain a cysteine-reactive transition metal, and complementary studies indicate that their inhibition of interaction is due to modification of a cysteine residue in the TEM8 extracellular domain. This is the first demonstration of a high-throughput screening assay that identifies inhibitors of TEM8, with potential application for antianthrax and antiangiogenic diseases.


Subject(s)
Antigens, Bacterial/metabolism , Neoplasm Proteins/antagonists & inhibitors , Protective Agents/metabolism , Receptors, Cell Surface/metabolism , Small Molecule Libraries/pharmacology , Bacillus anthracis/immunology , Biomarkers, Tumor/metabolism , Fluorescence Resonance Energy Transfer , High-Throughput Screening Assays , Humans , Microfilament Proteins , Neoplasm Proteins/metabolism , Pilot Projects , Receptors, Cell Surface/antagonists & inhibitors
10.
PLoS One ; 7(6): e39911, 2012.
Article in English | MEDLINE | ID: mdl-22768167

ABSTRACT

Anti-angiogenic therapies are effective for the treatment of cancer, a variety of ocular diseases, and have potential benefits in cardiovascular disease, arthritis, and psoriasis. We have previously shown that anthrax protective antigen (PA), a non-pathogenic component of anthrax toxin, is an inhibitor of angiogenesis, apparently as a result of interaction with the cell surface receptors capillary morphogenesis gene 2 (CMG2) protein and tumor endothelial marker 8 (TEM8). Hence, molecules that bind the anthrax toxin receptors may be effective to slow or halt pathological vascular growth. Here we describe development and testing of an effective homogeneous steady-state fluorescence resonance energy transfer (FRET) high throughput screening assay designed to identify molecules that inhibit binding of PA to CMG2. Molecules identified in the screen can serve as potential lead compounds for the development of anti-angiogenic and anti-anthrax therapies. The assay to screen for inhibitors of this protein-protein interaction is sensitive and robust, with observed Z' values as high as 0.92. Preliminary screens conducted with a library of known bioactive compounds identified tannic acid and cisplatin as inhibitors of the PA-CMG2 interaction. We have confirmed that tannic acid both binds CMG2 and has anti-endothelial properties. In contrast, cisplatin appears to inhibit PA-CMG2 interaction by binding both PA and CMG2, and observed cisplatin anti-angiogenic effects are not mediated by interaction with CMG2. This work represents the first reported high throughput screening assay targeting CMG2 to identify possible inhibitors of both angiogenesis and anthrax intoxication.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Fluorescence Resonance Energy Transfer/methods , High-Throughput Screening Assays/methods , Membrane Proteins/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Cisplatin/pharmacology , Immobilized Proteins/metabolism , Inhibitory Concentration 50 , Kinetics , Membrane Proteins/antagonists & inhibitors , Mice , Pilot Projects , Protein Binding/drug effects , Receptors, Peptide , Reproducibility of Results , Surface Plasmon Resonance , Tannins/pharmacology , Time Factors
11.
Front Biosci (Landmark Ed) ; 16(4): 1574-88, 2011 01 01.
Article in English | MEDLINE | ID: mdl-21196249

ABSTRACT

The anthrax toxin receptors tumor endothelial marker-8 (TEM-8) and capillary morphogenesis gene-2 (CMG-2) are responsible for allowing entry of anthrax toxin into host cells. These receptors were first discovered due to their enhanced expression on endothelial cells undergoing blood vessel growth or angiogenesis in model systems. Inhibition of angiogenesis is an important strategy for current anti-cancer therapies and treatment of retinal diseases. Functional roles for TEM-8 and CMG-2 in angiogenesis have recently emerged. TEM-8 appears to regulate endothelial cell migration and tubule formation whereas a role for CMG-2 in endothelial proliferation has been documented. TEM-8 and CMG-2 bind differentially to extracellular matrix proteins including collagen I, collagen IV and laminin and these properties may be responsible for their apparent roles in regulating endothelial cell behavior during angiogenesis. TEM-8-binding moieties have also been suggested to be useful in selectively targeting anti-angiogenic and anti-tumorigenic therapies to tumor endothelium. Additionally, studies of modified forms of lethal toxin (LeTx) have demonstrated that targeted inhibition of MAPKs within tumor vessels may represent an efficacious anti-angiogenic strategy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Membrane Proteins/drug effects , Neoplasm Proteins/drug effects , Neovascularization, Pathologic/drug therapy , Receptors, Cell Surface/drug effects , Animals , Antigens, Bacterial/therapeutic use , Antineoplastic Agents/therapeutic use , Bacterial Toxins/therapeutic use , Endothelial Cells/physiology , Extracellular Matrix/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microfilament Proteins , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Peptide
12.
Cardiovasc Res ; 89(3): 661-70, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21030535

ABSTRACT

AIMS: Aberrant retinal blood flow is a hallmark of various retinopathies and may be a causative factor in the pathology associated with these conditions. We examined the effects of pulsatile flow on bovine retinal endothelial cell (BREC) and bovine retinal pericyte (BRP) apoptosis and proliferation. METHODS AND RESULTS: Co-cultured BRECs and BRPs were exposed to low (0.3 mL/min) or high (25 mL/min) pulsatile flow for 72 h using a perfused transcapillary culture system. Pulsatile flow increased BREC nitric oxide synthase (eNOS) and cyclooxygenase-2 (COX-2) expression and activity concomitant with a significant decrease in pre-pro-endothelin-1 (ET-1) mRNA and peptide. BREC apoptosis was significantly attenuated following exposure to high flow. The inhibition of NOS, COX, and ET receptors significantly reduced the pro-survival effects of flow on BREC. In contrast, BRP apoptosis was significantly enhanced following exposure to high flow. The inhibition of COX and ET receptors significantly attenuated the high flow-induced increase in BRP apoptosis when compared with untreated controls. Treatment of static BREC with NO donor (S-nitroso-N-acetylpenicillamine, SNAP), ET-1, or iloprost inhibited serum deprivation-induced apoptosis, whereas treatment of BRP with ET-1 and iloprost, but not SNAP, was ineffective. High pulsatile flow decreased BRP proliferation, in the absence of any changes in BREC proliferation. CONCLUSION: Increased pulsatile flow promotes BREC survival and enhances BRP apoptosis through the activation of endothelial-derived vasoactive substances. Altered pulsatile flow does not alter BREC proliferation in co-culture with BRP, whereas BRP proliferation was significantly decreased at high flow rates. These interactions have important implications for vessel growth and regression during retinal vascular pathogenesis.


Subject(s)
Apoptosis/physiology , Endothelial Cells/cytology , Microcirculation/physiology , Pericytes/cytology , Pulsatile Flow/physiology , Retinal Vessels/cytology , Animals , Cattle , Cell Division/physiology , Cells, Cultured , Coculture Techniques , Endothelial Cells/physiology , Endothelin-1/metabolism , Epoprostenol/metabolism , Nitric Oxide/metabolism , Pericytes/physiology , Regional Blood Flow/physiology , Retina/cytology , Retina/physiology , Retinal Vessels/physiology , Stress, Mechanical
13.
Invest Ophthalmol Vis Sci ; 52(7): 4472-83, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21498615

ABSTRACT

PURPOSE: Aberrant retinal blood flow is a hallmark of retinopathies and may be a causative factor in their pathophysiology. In this study, the effects of pulsatile flow on hedgehog and Notch control of retinal endothelial cell and pericyte apoptosis were examined. METHODS: The levels of hedgehog and Notch signaling components in bovine retinal endothelial cells (BRECs) and pericytes (BRPs) were examined in vitro under static conditions and after exposure to pulsatile flow, with a perfused transcapillary co-culture system. Notch and hedgehog signaling was examined by immunocytochemistry, immunoblot, and real-time PCR. RESULTS: Notch and hedgehog proteins were present in BRECs and BRPs in vitro and in human retinal vasculature in vivo. Inhibition of hedgehog with cyclopamine and Notch with DAPT decreased hedgehog target gene levels and Notch intracellular receptor expression, respectively, concomitant with an increase in BREC and BRP apoptosis. Sonic hedgehog (Shh) mediated upregulation of Notch1 receptor levels was attenuated after cyclopamine treatment in both cell types. Exposure of co-cultured BRECs and BRPs to pulsatile flow increased apoptosis in the BRPs while concurrently decreasing apoptosis in the BRECs. These changes were concomitant with increased expression of Notch and hedgehog signaling components in the BRECs and reduced expression in the BRPs. The flow-induced decrease in apoptosis in the BRECs was associated with increased Notch receptor expression and was reversed after inhibition of hedgehog signaling with cyclopamine and inhibition of Notch signaling after ectopic expression of the CBF-1/RBP-Jκ-binding protein, RPMS-1. CONCLUSIONS: Pulsatile flow promotes BREC survival and enhances BRP apoptosis through modulation of Notch and hedgehog pathways. These interactions have important implications for the pathogenesis of retinopathies.


Subject(s)
Apoptosis/physiology , DNA/genetics , Endothelium, Vascular/cytology , Hedgehog Proteins/genetics , Pericytes/cytology , Receptors, Notch/genetics , Retina/cytology , Animals , Blotting, Western , Cattle , Cells, Cultured , Endothelial Cells/cytology , Endothelium, Vascular/metabolism , Hedgehog Proteins/biosynthesis , Humans , Immunohistochemistry , Pericytes/metabolism , Polymerase Chain Reaction , Receptors, Notch/biosynthesis , Retina/metabolism , Signal Transduction
14.
Atherosclerosis ; 211(1): 96-102, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20223456

ABSTRACT

OBJECTIVE: We have previously shown that conjugated linoleic acid (CLA) regresses pre-established murine atherosclerosis. Although the exact underlying mechanisms are unclear, accumulation of macrophages and expression of inflammatory markers were reduced in atherosclerotic plaques of CLA-fed mice, implicating the monocyte/macrophage as a target through which CLA may mediate anti-atherosclerotic effects. CLA mediates its effect at least in part via activation of the nuclear receptor, peroxisome proliferator activator receptor-gamma (PPARgamma). In this study we investigate if CLA mediates anti-atherogenic effects via modulation of monocyte/macrophage function and provide evidence for an additional PPARgamma-independent mechanism for CLA. METHODS AND RESULTS: Migration of the human monocyte cell line THP-1, and primary blood monocytes (HPBMCs) was assessed using transwell migration assays. Monocyte chemoattractant protein-1 (MCP-1) mediates chemotaxis via interaction with the chemokine (C-C motif)-2 receptor (CCR-2), which is expressed on the monocyte cell surface, and is negatively regulated by PPARgamma agonists. Incubation of THP-1 monocytes with CLA-isomers and a PPARgamma agonist inhibited MCP-1-induced monocyte migration. Prior to monocyte recruitment, activated platelets accumulate and release the contents of their secretory granules ("platelet-releasate"). Here we demonstrate that platelet-releasate is a monocyte chemoattractant, and CLA, but not the PPARgamma agonist, inhibits platelet-releasate-induced migration of THP-1 and HPBMC monocytes. CLA-treatment also suppressed the inflammatory macrophage phenotype, demonstrated by decreased induction of monocyte migration by CLA-treated macrophage-conditioned-media, as well as by decreased cyclooxygenase (COX)-2 and cytosolic phospholipase-A2 (cPLA2) expression and MCP-1, prostaglandin E2 (PGE2) and matrix metalloprotease (MMP)-9 generation. CONCLUSIONS: CLA-isomers inhibit monocyte migration and reduce the inflammatory output of the macrophage. These mechanisms may contribute to the potent anti-atherosclerotic effects of CLA in vivo.


Subject(s)
Cell Movement/drug effects , Linoleic Acids, Conjugated/pharmacology , Macrophages/drug effects , Monocytes/drug effects , Chemokine CCL2/metabolism , Chromans/pharmacology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Humans , Matrix Metalloproteinase 9/biosynthesis , PPAR gamma , Phospholipases A2/biosynthesis , Receptors, CCR2/physiology , Thiazolidinediones/pharmacology , Troglitazone
15.
Article in English | MEDLINE | ID: mdl-19767186

ABSTRACT

Prostaglandins have many important roles in ocular physiology and are used clinically for the treatment of glaucoma. The aim of this study was to analyse the contribution of each cyclooxygenase isoform to ocular prostaglandin production using isoform-specific knockout mice. Ex vivo PGE(2), 6-keto-PGF(1alpha), and TXB(2) production was measured from whole eyes, corneal tissue, uveoscleral tissue, lens, retina and optic nerve using enzyme-linked immunosorbant assays. Ocular immunohistochemical and histological analysis was also conducted for each genotype. Levels of each of the prostaglandins measured were significantly decreased in the corneal tissue, uveoscleral tissue, lens, retina and optic nerve of COX-1(-/-) mice in comparison with wild-type mice. In contrast, COX-2(-/-) mice had similar levels of ocular prostaglandin production to wild-type mice. These results suggest that COX-1 is the principal isoform responsible for prostaglandin production in the mouse eye. The absence of COX-1 or COX-2 did not appear to effect ocular development in these mice.


Subject(s)
Eye/anatomy & histology , Eye/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandins/metabolism , Age Factors , Animals , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cyclooxygenase 1/physiology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/physiology , Eye/growth & development , Genotype , Isoenzymes/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandin-Endoperoxide Synthases/physiology , Tissue Distribution
16.
Proteomics Clin Appl ; 2(5): 762-75, 2008 May.
Article in English | MEDLINE | ID: mdl-21136873

ABSTRACT

It is estimated that 37 million people worldwide suffer from blindness and 124 million people have impaired vision. While the relatively recently developed therapies, antivascular endothelial growth factor inhibitors for the treatment of age-related macular degeneration, and prostaglandin analogues for the treatment of glaucoma are beneficial for some patients, there are many individuals with sight-threatening diseases for whom no effective pharmacological therapy is available. For many of these diseases, the molecular mechanisms remain to be comprehensively elucidated, thus precluding the design of successful therapies against specific pathological targets. The current review summarises recent attempts to elucidate molecular mechanisms of ocular diseases, including diabetic retinal disease, age-related macular degeneration and inherited blindness using proteomic methodologies. A novel hypothesis can be generated from global protein expression analysis of disease tissue, which can then be addressed with cellular and in vivo functional studies. For example, the identification of extracellular carbonic anhydrase from the vitreous of diabetic retinopathy patients using MS based proteomics led to the elucidation of a new pathway involved in intraretinal edema, which could be inhibited by a number of agents targeting different proteins in this pathway in relevant animal models. The potential of protein biomarkers for diagnosis and the identification of novel disease mechanisms are also discussed.

17.
Proteomics ; 6(22): 5894-7, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17106913

ABSTRACT

This report summarizes the highlights of the recent British Society for Proteome Research (BSPR) meeting jointly organized with the European Bioinformatics Institute (EBI) which was held at the Wellcome Trust Genome Campus, Hinxton, Cambridge, UK in July 2006. This was the third annual scientific meeting organized by the BSPR and EBI and the theme of this years meeting was Integrative Proteomics: Structure, function and interaction. A wealth of local and overseas speakers were invited to discuss both their own work and specific challenges present in modern day proteomic based experiments.


Subject(s)
Computational Biology , Proteomics , Databases, Genetic , United Kingdom
SELECTION OF CITATIONS
SEARCH DETAIL