Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 301(5): H1872-81, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21856908

ABSTRACT

The unique contributions of connexin (Cx)37 and Cx40, gap junction-forming proteins that are coexpressed in vascular endothelium, to the recovery of tissues from ischemic injury are unknown. We recently reported that Cx37-deficient (Cx37(-/-)) animals recovered ischemic hindlimb function more quickly and to a greater extent than wild-type (WT) or Cx40(-/-) animals, suggesting that Cx37 limits recovery in the WT animal. Here, we tested the hypothesis that enhanced angiogenesis, arteriogenesis, and vasculogenesis contribute to improved postischemic hindlimb recovery in Cx37(-/-) animals. Ischemia was induced unilaterally in the hindlimbs of WT or Cx37(-/-) mice (isoflurane anesthesia). Postsurgical limb appearance, use, and perfusion were documented during recovery, and the number (and size) of large and small vessels was determined. Native collateral number, predominantly established during embryonic development (vasculogenesis), was also determined in the pial circulation. Both microvascular density in the gastrocnemius of the ischemic limb (an angiogenic field) and the number and tortuosity of larger vessels in the gracilis vasculature (an arteriogenic field) were increased in Cx37(-/-) animals compared with WT animals. Cx37(-/-) mice also had an increased (vs. WT) number of collateral vessels in the pial circulation. These findings suggest that in Cx37(-/-) animals, improved recovery of the ischemic hindlimb involves enhanced vasculogenesis, resulting in increased numbers of collaterals in the hindlimb (and pial circulations) and more extensive collateral remodeling and angiogenesis. These results are consistent with Cx37 exerting a growth-suppressive effect in the vasculature that limits embryonic vasculogenesis as well as arteriogenic and angiogenic responses to ischemic injury in the adult animal.


Subject(s)
Angiostatic Proteins/metabolism , Connexins/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Angiostatic Proteins/deficiency , Angiostatic Proteins/genetics , Animals , Blood Flow Velocity , Cerebrovascular Circulation , Collateral Circulation , Connexins/deficiency , Connexins/genetics , Disease Models, Animal , Hindlimb , Ischemia/diagnosis , Ischemia/genetics , Ischemia/physiopathology , Laser-Doppler Flowmetry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/metabolism , Microvessels/physiopathology , Pia Mater/blood supply , Recovery of Function , Regional Blood Flow , Time Factors , X-Ray Microtomography , Gap Junction alpha-4 Protein
2.
J Thromb Haemost ; 9(1): 209-19, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20961394

ABSTRACT

BACKGROUND AND OBJECTIVES: The non-allelic variant of CXCL4/PF4, CXCL4L1/PF4alt, differs from CXCL4 in three amino acids of the C-terminal α-helix and has been characterized as a potent anti-angiogenic regulator. Although CXCL4 structurally belongs to the chemokine family, it does not behave like a 'classical' chemokine, lacking significant chemotactic properties. Specific hallmarks are its angiostatic, anti-proliferative activities, and proinflammatory functions, which can be conferred by heteromer-formation with CCL5/RANTES enhancing monocyte recruitment. METHODS AND RESULTS: Here we show that tube formation of endothelial cells was inhibited by CXCL4L1 and CXCL4, while only CXCL4L1 triggered chemokinesis of endothelial cells. The chemotactic response towards VEGF and bFGF was attenuated by both variants and CXCL4L1-induced chemokinesis was blocked by bFGF or VEGF. Endothelial cell proliferation was inhibited by CXCL4 (IC(50) 6.9 µg mL(-1)) but not by CXCL4L1, while both chemokines bound directly to VEGF and bFGF. Moreover, CXCL4 enhanced CCL5-induced monocyte arrest in flow adhesion experiments and monocyte recruitment into the mouse peritoneal cavity in vivo, whereas CXCL4L1 had no effect. CXCL4L1 revealed lower affinity to CCL5 than CXCL4, as quantified by isothermal fluorescence titration. As evidenced by the reduction of the activated partial thromboplastin time, CXCL4L1 showed a tendency towards less heparin-neutralizing activity than CXCL4 (IC(50) 2.45 vs 0.98 µg mL(-1)). CONCLUSIONS: CXCL4L1 may act angiostatically by causing random endothelial cell locomotion, disturbing directed migration towards angiogenic chemokines, serving as a homeostatic chemokine with a moderate structural distinction yet different functional profile from CXCL4.


Subject(s)
Angiostatic Proteins/metabolism , Cell Movement , Cell Proliferation , Chemotaxis, Leukocyte , Endothelial Cells/immunology , Inflammation Mediators/metabolism , Monocytes/immunology , Neovascularization, Physiologic , Platelet Factor 4/metabolism , Angiostatic Proteins/chemical synthesis , Angiostatic Proteins/genetics , Animals , Blood Coagulation , Cells, Cultured , Chemokine CCL5/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Inflammation Mediators/chemical synthesis , Mice , Mice, Inbred C57BL , Partial Thromboplastin Time , Platelet Factor 4/chemical synthesis , Platelet Factor 4/genetics , Protein Multimerization , Recombinant Proteins/metabolism , Time Factors , Vascular Endothelial Growth Factor A/metabolism
3.
Biochemistry ; 49(14): 3156-60, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20225827

ABSTRACT

Human tryptophanyl-tRNA synthetase (TrpRS) catalyzes the aminoacylation of tRNA(Trp). Human TrpRS exists in two forms: a major form that is the full-length protein and a truncated form (mini TrpRS) in which most of the N-terminal extension is absent. Human mini, but not full-length, TrpRS has angiostatic activity. Because the full-length protein, which lacks angiostatic activity, has all of the amino acid determinants of the mini form, which has activity, I searched for conformational differences between the two proteins. Using a disulfide cross-linking assay, I showed that the molecular environment around Cys62 is significantly different between the two proteins. This difference can be explained by inspection of the three-dimensional structure of the full-length protein. These results give a clear demonstration of a significant difference, around a specific residue (Cys62), between a potent angiostatic and nonangiostatic version of human TrpRS.


Subject(s)
Angiostatic Proteins/chemistry , Cysteine/chemistry , Tryptophan-tRNA Ligase/chemistry , Aminoacylation , Angiostatic Proteins/genetics , Chromatography, Gel , Circular Dichroism , Disulfides/chemistry , Electrophoresis, Polyacrylamide Gel , Humans , Models, Molecular , Mutation , Protein Conformation , Protein Multimerization , RNA, Transfer, Trp/chemistry , Tryptophan-tRNA Ligase/genetics
4.
Microvasc Res ; 80(1): 31-6, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20219495

ABSTRACT

The effects of the antiangiogenic cytokine PEDF on key steps in retinal angiogenesis, specifically endothelial cell proliferation and vascular tubule formation, and the regulation of PEDF expression in retinal capillary endothelial cells were evaluated. HUVECs were co-cultured with fibroblasts to construct a model of angiogenesis using the Angiokit assay, and image analysis software was used to measure the effects of PEDF and VEGF on vascular tubule formation. Quantitative real-time PCR analysis was used to determine the expression of PEDF in microvascular endothelial cells exposed to glucose 20 mM, insulin 100 nM and VEGF 10 ng/ml. PEDF inhibited endothelial cell proliferation and significantly decreased the number of tubules (629+93 AU vs 311+31, p=0.001), number of branching points (145+19 AU vs 46+5, p=0.03) and total tubule length (4848+748 AU vs 11,172+2353, p=0.001). In bovine retinal capillary endothelial cells (BRCECs), PEDF mRNA and protein expression was suppressed by insulin (22%) in a rapamycin-sensitive manner; wortmannin had no effect. PEDF mRNA expression was also significantly reduced in the presence of high glucose (23%) and VEGF (25%). In conclusion, PEDF inhibits key steps in the angiogenic response of BRCECs, including endothelial cell proliferation and vascular tubule formation. Gene expression of PEDF is negatively regulated by glucose, insulin (via an mTOR-dependent pathway) and VEGF.


Subject(s)
Angiostatic Proteins/metabolism , Diabetic Retinopathy/metabolism , Eye Proteins/metabolism , Gene Expression Regulation/physiology , Neovascularization, Physiologic/drug effects , Nerve Growth Factors/metabolism , Serpins/metabolism , Androstadienes/pharmacology , Angiostatic Proteins/genetics , Angiostatic Proteins/pharmacology , Animals , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Eye Proteins/genetics , Eye Proteins/pharmacology , Gene Expression/drug effects , Glucose/pharmacology , Humans , Insulin/pharmacology , Nerve Growth Factors/genetics , Nerve Growth Factors/pharmacology , Retinal Vessels/cytology , Serpins/genetics , Serpins/pharmacology , Sirolimus/pharmacology , Umbilical Cord/cytology , Vascular Endothelial Growth Factor A/pharmacology , Wortmannin
5.
Cytokine ; 48(3): 295-302, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19782578

ABSTRACT

Blood vessel growth is regulated by angiogenic and angiostatic CXC chemokines, and radiation is a vasculogenic stimulus. We investigated the effect of radiation on endothelial cell chemokine signaling, receptor expression, and migration and apoptosis. Human umbilical vein endothelial cells were exposed to a single fraction of 0, 5, or 20 Gy of ionizing radiation (IR). All vasculogenic chemokines (CXCL1-3/5-8) increased 3-13-fold after 5 or 20 Gy IR. 20 Gy induced a marked increase (1.6-4-fold) in angiostatic CXC chemokines. CXCR4 expression increased 3.5 and 7-fold at 48 h after 5 and 20 Gy, respectively. Bone marrow progenitor cell chemotaxis was augmented by conditioned media from cells treated with 5 Gy IR. Whereas 5 Gy markedly decreased intrinsic cell apoptosis (0 Gy=16%+/-3.6 vs. 5 Gy=4.5%+/-0.3), 20 Gy increased it (21.4%+/-1.2); a reflection of pro-survival angiogenic chemokine expression. Radiation induces a dose-dependent increase in pro-angiogenic CXC chemokines and CXCR4. In contrast, angiostatic chemokines and apoptosis were induced at higher (20 Gy) radiation doses. Cell migration improved significantly following 5 Gy, but not 20 Gy IR. Collectively, these data suggest that lower doses of IR induce an angiogenic cascade while higher doses produce an angiostatic profile.


Subject(s)
Angiostatic Proteins/metabolism , Chemokines, CXC/metabolism , Endothelial Cells/radiation effects , Gene Expression Regulation/radiation effects , Angiogenesis Inducing Agents/metabolism , Angiostatic Proteins/genetics , Angiostatic Proteins/radiation effects , Apoptosis/radiation effects , Cell Line , Cell Movement/radiation effects , Dose-Response Relationship, Immunologic , Dose-Response Relationship, Radiation , Endothelial Cells/immunology , Flow Cytometry , Humans , Immunoblotting , RNA, Messenger/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR4/radiation effects , Signal Transduction/radiation effects
6.
Blood ; 114(1): 128-43, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19299335

ABSTRACT

Abundant bone marrow angiogenesis is present in almost all myeloma patients requiring therapy and correlated to treatment response and survival. We assessed the expression of 402 angiogenesis-associated genes by Affymetrix DNA microarrays in 466 samples, including CD138-purified myeloma cells (MMCs) from 300 previously untreated patients, in vivo microcirculation by dynamic contrast-enhanced magnetic resonance imaging, and in vitro angiogenesis (AngioKit-assay). Normal bone marrow plasma cells (BMPCs) express a median of 39 proangiogenic (eg, VEGFA, ADM, IGF-1) and 28 antiangiogenic genes (eg, TIMP1, TIMP2). Supernatants of BMPCs unlike those of memory B cells induce angiogenesis in vitro. MMCs do not show a significantly higher median number of expressed proangiogenic (45) or antiangiogenic (31) genes, but 97% of MMC samples aberrantly express at least one of the angiogenic factors HGF, IL-15, ANG, APRIL, CTGF, or TGFA. Supernatants of MMCs and human myeloma cell lines induce significantly higher in vitro angiogenesis compared with BMPCs. In conclusion, BMPCs express a surplus of proangiogenic over antiangiogenic genes transmitting to the ability to induce in vitro angiogenesis. Aberrant expression of proangiogenic and down-regulation of antiangiogenic genes by MMCs further increases the angiogenic stimulus, together leading to bone marrow angiogenesis at various degrees in all myeloma patients.


Subject(s)
Multiple Myeloma/blood supply , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/genetics , Plasma Cells/pathology , Plasma Cells/physiology , Angiogenic Proteins/genetics , Angiostatic Proteins/genetics , B-Lymphocytes/pathology , B-Lymphocytes/physiology , Bone Marrow/blood supply , Case-Control Studies , Cell Line, Tumor , Gene Expression , Gene Expression Profiling , Humans , In Vitro Techniques , Models, Biological , Multiple Myeloma/genetics , Neovascularization, Physiologic/genetics , Oligonucleotide Array Sequence Analysis
7.
FEBS Lett ; 582(29): 4059-65, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-19007777

ABSTRACT

HDAC inhibitors are promising anticancer agents that induce cell cycle arrest and apoptosis. However, the role of HDACs in cancer progression, such as angiogenesis and metastasis, remains largely unexplored. Among various HDAC inhibitors, we demonstrate that TSA and SAHA upregulated the expression of angiostatic ADAMTS1 in A549 cells. HDAC6 inhibitor tubacin, and knockdown of HDAC6, also lead to ADAMTS1 upregulation. By reporter, DAPA, and ChIP assays, the proximal GC boxes were demonstrated to be essential for ADAMTS1 induction. Decreased binding of SP1 and HDAC6 to the ADAMTS1 promoter after TSA treatment was also seen. These data suggest the involvement of HDAC6 and SP1 in the HDACi-induced expression of angiostatic ADAMTS1.


Subject(s)
ADAM Proteins/biosynthesis , Angiostatic Proteins/biosynthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , ADAM Proteins/genetics , ADAMTS1 Protein , Angiostatic Proteins/genetics , Cell Line, Tumor , Histone Deacetylase 6 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Up-Regulation , Vorinostat
8.
Biol Chem ; 389(6): 765-71, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18627292

ABSTRACT

Kallikrein-related peptidase 3 (KLK3, also known as prostate-specific antigen, PSA) is a chymotrypsin-like kallikrein that has anti-angiogenic properties. We have previously shown in a human umbilical vein endothelial cell (HUVEC) model that the anti-angiogenic effect of KLK3 is related to its enzyme activity. However, the mechanism of this effect remains to be clarified. To this end, we used a DNA microarray to study KLK3-induced changes in gene expression associated with reduction of HUVEC tube formation. Among the 41,000 genes studied, 311 were differentially expressed between control and KLK3-treated cells. These changes were enriched in several pathways, including those associated with proteasome, ubiquitin-mediated proteolysis, focal adhesion and regulation of the actin cytoskeleton. Furthermore, the changes were opposite to those previously described to occur during tubulogenesis. In conclusion, our results show that KLK3 induces gene expression changes in HUVECs. Although these changes might be relevant for the mechanism by which KLK3 exerts its anti-angiogenic activity, it cannot be judged from the present results whether they reflect the primary mechanism mediating the effect of KLK3 or are secondary to morphogenic differentiation.


Subject(s)
Angiostatic Proteins/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation , Kallikreins/metabolism , Umbilical Veins/cytology , Angiostatic Proteins/genetics , Humans , Kallikreins/genetics , Oligonucleotide Array Sequence Analysis , Up-Regulation
9.
J Biol Chem ; 283(4): 2070-7, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-17999956

ABSTRACT

In mammalian cells, specific aminoacyl-transfer RNA (tRNA) synthetases have cytokine functions that require interactions with partners outside of the translation apparatus. Little is known about these interactions and how they facilitate expanded functions that link protein translation to other cellular pathways. For example, an alternative splice fragment of tryptophanyl-tRNA synthetase (TrpRS) and a similar natural proteolytic fragment are potent angiostatic factors that act through the vascular endothelial-cadherin receptor and Akt signaling pathway. Here we demonstrate mobilization of TrpRS for exocytosis from endothelial cells and the potential for plasmin to activate the cytokine function of the extracellular synthetase. Direct physical evidence showed that the annexin II-S100A10 complex, which regulates exocytosis, forms a ternary complex with TrpRS. Functional studies demonstrate that both annexin II and S100A10 regulate trafficking of TrpRS. Thus, complexes of mammalian tRNA synthetases with seemingly disparate proteins may in general be relevant to understanding how their expanded functions are implemented.


Subject(s)
Angiostatic Proteins/metabolism , Annexin A2/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Fibrinolysin/metabolism , S100 Proteins/metabolism , Tryptophan-tRNA Ligase/metabolism , Alternative Splicing/physiology , Angiostatic Proteins/genetics , Annexin A2/genetics , Cells, Cultured , Cytokines/genetics , Endothelial Cells/cytology , Exocytosis/physiology , Fibrinolysin/genetics , Humans , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Biosynthesis/physiology , Protein Transport/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , S100 Proteins/genetics , Signal Transduction/physiology , Tryptophan-tRNA Ligase/genetics
10.
J Biol Chem ; 283(4): 2335-43, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-18024432

ABSTRACT

Endorepellin, the C-terminal module of perlecan, has angiostatic activity. Here we provide definitive genetic and biochemical evidence that the functional endorepellin receptor is the alpha2beta1 integrin. Notably, the specific endorepellin binding to the receptor was cation-independent and was mediated by the alpha2 I domain. We show that the anti-angiogenic effects of endorepellin cannot occur in the absence of alpha2beta1. Microvascular endothelial cells from alpha2beta1(-/-) mice, but not those isolated from either wild-type or alpha1beta1(-/-) mice, did not respond to endorepellin. Moreover, syngeneic Lewis lung carcinoma xenografts in alpha2beta1(-/-) mice failed to respond to systemic delivery of endorepellin. In contrast, endorepellin inhibited tumor growth and angiogenesis in the wild-type mice expressing integrin alpha2beta1. We conclude that the angiostatic effects of endorepellin in vivo are mediated by a specific interaction of endorepellin with the alpha2beta1 integrin receptor.


Subject(s)
Angiostatic Proteins/genetics , Endothelium, Vascular/metabolism , Heparan Sulfate Proteoglycans/metabolism , Integrin alpha2beta1/metabolism , Neovascularization, Pathologic/metabolism , Peptide Fragments/metabolism , Angiostatic Proteins/metabolism , Angiostatic Proteins/pharmacology , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Endothelium, Vascular/pathology , Female , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/pharmacology , Humans , Integrin alpha1beta1/genetics , Integrin alpha1beta1/metabolism , Integrin alpha2beta1/genetics , Mice , Mice, Knockout , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Transplantation, Heterologous
11.
Drug News Perspect ; 20(2): 77-86, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17440630

ABSTRACT

With their biodegradability and diversity of permutations, peptides have significant potential as carriers of nucleic acids. This review will focus on the sequence and branching patterns of peptide carriers composed primarily of histidines and lysines. While lysines within peptides are important for binding to the negatively charged phosphates, histidines are critical for endosomal lysis enabling nucleic acids to reach the cytosol. Histidine-lysine (HK) polymers by either covalent or ionic bonds with liposomes augment transfection compared to liposome carriers alone. More recently, we have examined peptides as sole carriers of nucleic acids because of their intrinsic advantages compared to the bipartite HK/liposome carriers. With a protocol change and addition of a histidine-rich tail, HK peptides as sole carriers were more effective than liposomes alone in several cell lines. While four-branched polymers with a primary repeating sequence pattern of -HHK- were more effective as carriers of plasmids, eight-branched polymers with a sequence pattern of -HHHK- were more effective as carriers of siRNA. Compared to polyethylenimine, HK carriers of siRNA and plasmids had reduced toxicity. When injected intravenously, HK polymers in complex with plasmids encoding antiangiogenic proteins significantly decreased tumor growth. Furthermore, modification of HK polymers with polyethylene glycol and vascular-specific ligands increased specificity of the polyplex to the tumor by more than 40-fold. Together with further development and insight on the structure of HK polyplexes, HK peptides may prove to be useful as carriers of different forms of nucleic acids both in vitro and in vivo.


Subject(s)
DNA/metabolism , Genetic Therapy/methods , Neoplasms/blood supply , Neoplasms/therapy , Peptides/metabolism , RNA, Small Interfering/metabolism , Transfection/methods , Angiostatic Proteins/genetics , Angiostatic Proteins/metabolism , Animals , DNA/chemistry , Gene Expression Regulation, Neoplastic , Histidine , Humans , Lysine , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic , Nucleic Acid Conformation , Peptides/chemistry , Peptides/toxicity , Protein Conformation , RNA Interference , RNA, Small Interfering/chemistry , Tumor Burden
12.
Proc Natl Acad Sci U S A ; 102(6): 2040-5, 2005 Feb 08.
Article in English | MEDLINE | ID: mdl-15687502

ABSTRACT

A group of angiogenesis inhibitors are derived from fragments of extracellular matrix or blood proteins. Endostatin, antithrombin, and anastellin are members of this group of substances. The plasma adhesion proteins fibronectin and vitronectin serve as cofactors for these three antiangiogenic proteins. Anginex is a synthetic 33-amino acid peptide that was originally modeled to reproduce the beta-sheet structure of antiangiogenic proteins. Here, we show that anginex initiates fibronectin polymerization and is inactive in mice that lack plasma fibronectin. Anginex shares these characteristics with anastellin. Fluorescein-labeled anginex and anastellin specifically localized in angiogenic vessels in vivo. This localization was dependent on plasma fibronectin and inhibited by an Arg-Gly-Asp peptide. Thus, anginex shares with several physiological angiogenesis inhibitors a dependence on plasma adhesion proteins. The role of the adhesion protein interaction apparently is to form integrin-binding complexes that deliver the antiangiogenic proteins to sites of angiogenesis. This functional convergence of several antiangiogenic factors has important implications for antiangiogenic therapies.


Subject(s)
Angiogenesis Inhibitors/metabolism , Angiostatic Proteins/metabolism , Blood Vessels/metabolism , Fibronectins/metabolism , Neovascularization, Physiologic , Proteins/metabolism , Angiostatic Proteins/genetics , Animals , Biocompatible Materials/metabolism , Blood Vessels/anatomy & histology , Collagen/metabolism , Drug Combinations , Fibronectins/genetics , Laminin/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oligopeptides/metabolism , Peptide Fragments/metabolism , Peptides , Plasma/chemistry , Polymers/metabolism , Proteins/genetics , Proteoglycans/metabolism , Vitronectin/genetics , Vitronectin/metabolism
13.
Circ Res ; 95(9): 855-7, 2004 Oct 29.
Article in English | MEDLINE | ID: mdl-15459074

ABSTRACT

Platelet factor-4 (PF-4)/CXCL4 was the first chemokine described to inhibit neovascularization. Here, the product of the nonallelic variant gene of CXCL4, PF-4var1/PF-4alt, designated CXCL4L1, was isolated for the first time from thrombin-stimulated human platelets and purified to homogeneity. Although secreted CXCL4 and CXCL4L1 differ in only three amino acids, CXCL4L1 was more potent in inhibiting chemotaxis of human microvascular endothelial cells toward interleukin-8 (IL-8)/CXCL8 or basic fibroblast growth factor (bFGF). In vivo, CXCL4L1 was also more effective than CXCL4 in inhibiting bFGF-induced angiogenesis in rat corneas. Thus, activated platelets release CXCL4L1, a potent regulator of endothelial cell biology, which affects angiogenesis and vascular diseases.


Subject(s)
Angiostatic Proteins/metabolism , Blood Platelets/metabolism , Platelet Factor 4/metabolism , Alleles , Amino Acid Sequence , Angiostatic Proteins/genetics , Angiostatic Proteins/isolation & purification , Angiostatic Proteins/pharmacology , Animals , Blood Platelets/drug effects , Cells, Cultured/drug effects , Cells, Cultured/physiology , Chemotaxis/drug effects , Corneal Neovascularization/chemically induced , Culture Media, Conditioned/pharmacology , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Endothelial Cells/drug effects , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/pharmacology , Humans , Interleukin-8/antagonists & inhibitors , Interleukin-8/pharmacology , Molecular Sequence Data , Platelet Activation , Platelet Factor 4/genetics , Platelet Factor 4/isolation & purification , Platelet Factor 4/pharmacology , Rats , Structure-Activity Relationship , Thrombin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...