Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters










Publication year range
1.
Oncogenesis ; 5(5): e230, 2016 May 30.
Article in English | MEDLINE | ID: mdl-27239962

ABSTRACT

Thrombospondin 1 is a glycoprotein that regulates cellular phenotype through interactions with its cellular receptors and extracellular matrix-binding partners. Thrombospondin 1 locally regulates angiogenesis and inflammatory responses that contribute to colorectal carcinogenesis in Apc(Min/+) mice. The ability of thrombospondin 1 to regulate responses of cells and tissues to a variety of stresses suggested that loss of thrombospondin 1 may also have broader systemic effects on metabolism to modulate carcinogenesis. Apc(Min/+):Thbs1(-/-) mice exhibited decreased survival and higher tumor multiplicities in the small and large intestine relative to Apc(Min/+) mice when fed a low (5%) fat western diet. However, the protective effect of endogenous thrombospondin 1 was lost when the mice were fed a western diet containing 21% fat. Biochemical profiles of liver tissue identified systemic metabolic changes accompanying the effects of thrombospondin 1 and dietary lipid intake on tumorigenesis. A high-fat western diet differentially regulated elements of amino acid, energy and lipid metabolism in Apc(Min/+):Thbs1(-/-) mice relative to Apc(Min/+):Thbs1(+/+)mice. Metabolic changes in ketone body and tricarboxylic acid cycle intermediates indicate functional interactions between Apc and thrombospondin 1 signaling that control mitochondrial function. The cumulative diet-dependent differential changes observed in Apc(Min/+):Thbs1(-/-) versus Apc(Min/+) mice include altered amino acid and lipid metabolism, mitochondrial dysfunction, eicosanoids and ketone body formation. This metabolic profile suggests that the protective role of thrombospondin 1 to decrease adenoma formation in Apc(Min/+) mice results in part from improved mitochondrial function.

2.
Gene Ther ; 17(9): 1085-97, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20463761

ABSTRACT

We have developed a multi-disciplinary approach combining molecular biology, delivery technology, combinatorial chemistry and reversible masking to create improved systemic, targeted delivery of plasmid DNA while avoiding nonspecific uptake in vivo. We initially used a well-characterized model targeting the asialolglycoprotein receptor in the liver. Using our bilamellar invaginated vesicle (BIV) liposomal delivery system with reversible masking, we increased expression in the liver by 76-fold, nearly equaling expression in first-pass organs using non-targeted complexes, with no expression in other organs. The same technology was then applied to efficiently target delivery to a human tumor microenvironment model. We achieved efficient, targeted delivery by attachment of specific targeting ligands to the surface of our BIV complexes in conjunction with reversible masking to bypass nonspecific tissues and organs. We identified ligands that target a human tumor microenvironment created in vitro by co-culturing primary human endothelial cells with human lung or pancreatic cancer cells. The model was confirmed by increased expression of tumor endothelial phenotypes including CD31 and vascular endothelial growth factor-A, and prolonged survival of endothelial capillary-like structures. The co-cultures were used for high-throughput screening of a specialized small molecule library to identify ligands specific for human tumor-associated endothelial cells in vitro. We identified small molecules that enhanced the transfection efficiency of tumor-associated endothelial cells, but not normal human endothelial cells or cancer cells. Intravenous (i.v.) injection of our targeted, reversibly masked complexes into mice, bearing human pancreatic tumor and endothelial cells, specifically increased transfection to this tumor microenvironment approximately 200-fold. Efficacy studies using our optimized targeted delivery of a plasmid encoding thrombospondin-1 eliminated tumors completely after five i.v. injections administered once every week.


Subject(s)
Genetic Therapy/methods , Neoplasms/therapy , Transfection , Animals , Asialoglycoprotein Receptor/metabolism , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Endothelial Cells/metabolism , Humans , Liposomes/chemistry , Liposomes/metabolism , Mice , Plasmids/genetics , Plasmids/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor A/metabolism
3.
Oncogene ; 28(43): 3837-46, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19701246

ABSTRACT

The chemopreventative effects of dithiolethione compounds are attributed to their activation of antioxidant response elements (AREs) by reacting with the Nrf2/Keap1 protein complex. In this study, we show antiproliferative effects of the dithiolethione compound ACS-1 in human cancer cell lines (A549 and MDA-MB-231) by increasing the activity of the tumor suppressor protein phoshatase 2A (PP2A). ACS-1 inhibited epidermal growth factor (EGF)-induced cellular proliferation in a concentration- and time-dependent manner. Akt activation, as determined by serine-473 phosphorylation, was inhibited by ACS-1 in cells stimulated with either EGF or fibronectin. Furthermore, ACS-1 inhibited mammalian target of rapamycin signaling and decreased c-myc protein levels. ACS-1 did not proximally alter EGF receptor or integrin signaling, but caused a concentration-dependent increase in PP2A activity. The effect of ACS-1 on Akt activation was not observed in the presence of the PP2A inhibitor okadaic acid. ACS-1 effects on PP2A activity were independent of ARE activation and cAMP formation. In addition to ACS-1, other dithiolethione compounds showed similar effects in reducing Akt activation, suggesting that this class of compounds may have other effects beyond chemoprevention.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
4.
Cell Mol Life Sci ; 65(5): 728-42, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18193160

ABSTRACT

Thrombospondin-1 is a secreted protein that modulates vascular cell behavior via several cell surface receptors. In vitro, nanomolar concentrations of thrombospondin-1 are required to alter endothelial and vascular smooth muscle cell adhesion, proliferation, motility, and survival. Yet, much lower levels of thrombospondin-1 are clearly functional in vivo. This discrepancy was explained with the discovery that the potency of thrombospondin-1 increases more than 100-fold in the presence of physiological levels of nitric oxide (NO). Thrombospondin-1 binding to CD47 inhibits NO signaling by preventing cGMP synthesis and activation of its target cGMP-dependent protein kinase. This potent antagonism of NO signaling allows thrombospondin-1 to acutely constrict blood vessels, accelerate platelet aggregation, and if sustained, inhibit angiogenic responses. Acute antagonism of NO signaling by thrombospondin-1 is important for hemostasis but becomes detrimental for tissue survival of ischemic injuries. New therapeutic approaches targeting thrombospondin-1 or CD47 can improve recovery from ischemic injuries and overcome a deficit in NO-responsiveness in aging. (Part of a Multi-author Review).


Subject(s)
Nitric Oxide/metabolism , Signal Transduction , Thrombospondin 1/physiology , Animals , Humans , Neovascularization, Physiologic
6.
Br J Pharmacol ; 151(1): 63-72, 2007 May.
Article in English | MEDLINE | ID: mdl-17351657

ABSTRACT

BACKGROUND AND PURPOSE: Angiogenesis involves multiple signaling pathways that must be considered when developing agents to modulate pathological angiogenesis. Because both cyclooxygenase inhibitors and dithioles have demonstrated anti-angiogenic properties, we investigated the activities of a new class of anti-inflammatory drugs containing dithiolethione moieties (S-NSAIDs) and S-valproate. EXPERIMENTAL APPROACH: Anti-angiogenic activities of S-NSAIDS, S-valproate, and the respective parent compounds were assessed using umbilical vein endothelial cells, muscle and tumor tissue explant angiogenesis assays, and developmental angiogenesis in Fli:EGFP transgenic zebrafish embryos. KEY RESULTS: Dithiolethione derivatives of diclofenac, valproate, and sulindac inhibited endothelial cell proliferation and induced Ser(78) phosphorylation of hsp27, a known molecular target of anti-angiogenic signaling. The parent drugs lacked this activity, but dithiolethiones were active at comparable concentrations. Although dithiolethiones can potentially release hydrogen sulphide, NaSH did not reproduce some activities of the S-NSAIDs, indicating that the dithioles regulate angiogenesis through mechanisms other than release of H(2)S. In contrast to the parent drugs, S-NSAIDs, S-valproate, NaSH, and dithiolethiones were potent inhibitors of angiogenic responses in muscle and HT29 tumor explants assessed by 3-dimensional collagen matrix assays. Dithiolethiones and valproic acid were also potent inhibitors of developmental angiogenesis in zebrafish embryos, but the S-NSAIDs, remarkably, lacked this activity. CONCLUSIONS AND IMPLICATION: S-NSAIDs and S-valproate have potent anti-angiogenic activities mediated by their dithiole moieties. The novel properties of S-NSAIDs and S-valproate to inhibit pathological versus developmental angiogenesis suggest that these agents may have a role in cancer treatment.


Subject(s)
Anethole Trithione/pharmacology , Angiogenesis Inhibitors/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Valproic Acid/pharmacology , Animals , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , HSP27 Heat-Shock Proteins , Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Phosphorylation , Solubility , Zebrafish
7.
Oncogene ; 25(4): 536-45, 2006 Jan 26.
Article in English | MEDLINE | ID: mdl-16247480

ABSTRACT

Three-dimensional explant cultures of muscle tissue were used to characterize secreted proteins regulated by endogenous levels of the angiogenesis modulator thrombospondin (TSP)-1. Explants from TSP1 null mice exhibit enhanced neovascularization associated with increased endothelial outgrowth but decreased outgrowth of perivascular smooth muscle cells . The absence of endogenous TSP1 did not diminish activation of latent transforming growth factor-beta and moderately decreased matrix metalloproteinase levels. However, significant changes in other secreted proteins were observed. Endogenous TSP1 decreased mRNA levels for collagens Ialpha1, Ialpha2, and IIIalpha1 and laminin alpha4 and increased collagen IValpha1 mRNA expression. Endogenous TSP1 also decreased the level of type I collagen protein produced by the vascular outgrowths. Collagens Ialpha1, Ialpha2, and IIIalpha1 are known tumor endothelial markers, suggesting that TSP1 coordinately regulates a set of extracellular matrix genes that reverse the angiogenic switch. Suppression of collagen Ialpha1 or Ialpha2 mRNAs using antisense morpholinos inhibited outgrowth in TSP1 null explants and proliferation of TSP1 null endothelial cells, indicating that type I collagen synthesis is limiting for this neovascularization response.


Subject(s)
Angiogenesis Inhibitors/physiology , Collagen Type I/genetics , Gene Expression Regulation , Thrombospondin 1/physiology , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Laminin/genetics , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 9/analysis , Mice , RNA, Messenger/analysis , Transforming Growth Factor beta/metabolism
8.
J Biol Chem ; 276(30): 27913-22, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11358957

ABSTRACT

The recognition of extracellular matrix components can be regulated by conformational changes that alter the activity of cell surface integrins. We now demonstrate that conformational regulation of the matrix glycoprotein thrombospondin-1 (TSP1) can also modulate its binding to an integrin receptor. F18 1G8 is a conformation-sensitive TSP1 antibody that binds weakly to soluble TSP1 in the presence of divalent cations. However, binding of the antibody to melanoma cells was strongly stimulated by adding exogenous TSP1 in the presence of calcium, suggesting that TSP1 undergoes a conformational change following its binding to the cell surface. This conformation was not induced by known cell surface TSP1 receptors, whereas binding of F18 was stimulated when TSP1 bound to fibronectin but not to heparin or fibrinogen. Conversely, binding of F18 to TSP1 enhanced TSP1 binding to fibronectin. Exogenous fibronectin also stimulated TSP1-dependent binding of F18 to melanoma cells. Binding of the fibronectin-TSP1 complex to melanoma cells was mediated by alpha4beta1 and alpha5beta1 integrins. Furthermore, binding to F18 or fibronectin strongly enhanced the adhesive activity of immobilized TSP1 for some cell types. This enhancement of adhesion was mediated by alpha3beta1 integrin and required that the alpha3beta1 integrin be in an active state. Fibronectin also enhanced TSP1 binding to purified alpha3beta1 integrin. Therefore, both fibronectin and the F18 antibody induce conformational changes in TSP1 that enhance the ability of TSP1 to be recognized by alpha3beta1 integrin. The conformational and functional regulation of TSP1 activity by fibronectin represents a novel mechanism for extracellular signal transduction.


Subject(s)
Fibronectins/chemistry , Fibronectins/metabolism , Integrins/chemistry , Integrins/metabolism , Thrombospondins/metabolism , Animals , Antibodies/metabolism , Calcium/pharmacology , Cell Adhesion , Dose-Response Relationship, Drug , Epitopes , Humans , Integrin alpha3beta1 , Integrin beta1/metabolism , Kinetics , Ligands , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Models, Biological , Peptides/chemistry , Protein Binding , Protein Conformation , Signal Transduction , Thrombospondins/chemistry , Tumor Cells, Cultured
9.
Clin Cancer Res ; 7(4): 854-60, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309333

ABSTRACT

Two proteins recovered from cell surface adhesion complexes in a small cell lung carcinoma (SCLC) cell line were identified as fragments of the seminal plasma proteins semenogelin I and semenogelin II. Association of both proteins with the adhesion complexes was induced by epidermal growth factor. Expression of semenogelins was previously thought to be highly specific to seminal vesicles, but Western blot analysis demonstrated that semenogelin II is widely expressed in SCLC cell lines and occasionally in other malignant cell lines. Although semenogelin expression is normally restricted to males, two SCLC cell lines from female patients were also positive for semenogelin II expression. Immunohistochemical analysis demonstrated diffuse expression of semenogelins in 12 of 13 SCLC tumors and focal expression in a minority of lung squamous and adenocarcinomas. Semenogelins were secreted into the medium by cultured SCLC cells, which suggested that these proteins may be useful markers for detecting residual tumor burden or recurrence of SCLC after treatment.


Subject(s)
Biomarkers, Tumor/isolation & purification , Carcinoma, Small Cell/metabolism , Gonadal Steroid Hormones/isolation & purification , Lung Neoplasms/metabolism , Seminal Plasma Proteins , Seminal Vesicle Secretory Proteins , Biomarkers, Tumor/analysis , Carcinoma, Small Cell/pathology , Female , Gonadal Steroid Hormones/analysis , Humans , Immunohistochemistry , Lung Neoplasms/pathology , Male , Tumor Cells, Cultured
10.
Biochemistry ; 39(51): 16110-8, 2000 Dec 26.
Article in English | MEDLINE | ID: mdl-11123939

ABSTRACT

Hemoglobin (Hb) is a host factor that induces expression of a promiscuous receptor on Candida albicans for fibronectin (FN) and several other extracellular matrix proteins. FN receptor expression was induced by ferric (Hb(+)Met and Hb(+)CN), ferrous (HbCO and HbO(2)), and cobalt-protoporphyrin derivatives of Hb, whereas globin was inactive. The Hb derivatives all exhibited saturable, dose-dependent kinetics of FN receptor induction, suggesting that Hb may be acting as a receptor ligand. Soluble Hb bound saturably to a low-affinity binding site [K(d) = (1.1+/-0.2) x 10(-6) M] on C. albicans blastospores. However, uptake of (55)FeHb revealed that heme or iron transport into the cell is not required for induction, since internalization of (55)Fe from Hb did not occur until after induction of FN binding. The serum Hb-binding protein, haptoglobin, specifically abrogated this response, indicating that protein structure rather than the heme ligand or iron is necessary for induction of this signaling pathway. C. albicans also adhered to immobilized Hb, which was sufficient to induce FN receptor expression, and to Hb polymers that formed in defined Hb liquid media in the presence of cells. Formation of Hb polymers in solution required metabolic energy, since the aggregation process was halted with azide addition. Collectively, these data demonstrate that C. albicans recognizes polymerized Hb through multivalent low-affinity interactions, and this may be a host environmental cue that triggers extracellular matrix receptor expression at a septic site.


Subject(s)
Candida albicans/metabolism , Hemoglobins/chemistry , Hemoglobins/physiology , Receptors, Fibronectin/biosynthesis , Candida albicans/genetics , Candida albicans/growth & development , Candida albicans/physiology , Cell Adhesion , Dose-Response Relationship, Drug , Energy Metabolism , Fibronectins/metabolism , Gene Expression Regulation, Fungal , Haptoglobins/chemistry , Hemoglobins/antagonists & inhibitors , Hemoglobins/metabolism , Humans , Protein Binding , Receptors, Fibronectin/antagonists & inhibitors , Receptors, Fibronectin/genetics , Spores, Fungal/growth & development , Spores, Fungal/metabolism , Spores, Fungal/physiology , Structure-Activity Relationship
11.
Mol Biol Cell ; 11(9): 2885-900, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10982388

ABSTRACT

Thrombospondin-1 (TSP1) can inhibit angiogenesis by interacting with endothelial cell CD36 or proteoglycan receptors. We have now identified alpha3beta1 integrin as an additional receptor for TSP1 that modulates angiogenesis and the in vitro behavior of endothelial cells. Recognition of TSP1 and an alpha3beta1 integrin-binding peptide from TSP1 by normal endothelial cells is induced after loss of cell-cell contact or ligation of CD98. Although confluent endothelial cells do not spread on a TSP1 substrate, alpha3beta1 integrin mediates efficient spreading on TSP1 substrates of endothelial cells deprived of cell-cell contact or vascular endothelial cadherin signaling. Activation of this integrin is independent of proliferation, but ligation of the alpha3beta1 integrin modulates endothelial cell proliferation. In solution, both intact TSP1 and the alpha3beta1 integrin-binding peptide from TSP1 inhibit proliferation of sparse endothelial cell cultures independent of their CD36 expression. However, TSP1 or the same peptide immobilized on the substratum promotes their proliferation. The TSP1 peptide, when added in solution, specifically inhibits endothelial cell migration and inhibits angiogenesis in the chick chorioallantoic membrane, whereas a fragment of TSP1 containing this sequence stimulates angiogenesis. Therefore, recognition of immobilized TSP1 by alpha3beta1 integrin may stimulate endothelial cell proliferation and angiogenesis. Peptides that inhibit this interaction are a novel class of angiogenesis inhibitors.


Subject(s)
Cell Communication/physiology , Endothelium, Vascular/physiology , Integrins/physiology , Thrombospondin 1/physiology , Allantois/blood supply , Allantois/physiology , Animals , Aorta , CD36 Antigens/physiology , Cadherins/physiology , Cattle , Cell Division , Cells, Cultured , Chick Embryo , Chorion/blood supply , Chorion/physiology , Endothelium, Vascular/cytology , Fibronectins/pharmacology , Fibronectins/physiology , Integrin alpha3beta1 , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Signal Transduction , Thrombospondin 1/pharmacology
12.
Br J Cancer ; 83(3): 298-306, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10917542

ABSTRACT

We have previously shown that platelet-produced thrombospondin-1 up-regulates the urokinase plasminogen activator and its receptor and promotes tumour cell invasion. Although tumour cells produce thrombospondin-1 in vivo, they produce only minimal amounts of thrombospondin-1 in vitro. To determine the effect of tumour cell-produced thrombospondin-1 in the regulation of the plasminogen/plasmin system and tumour cell invasion, we studied THBS-1-transfected MDA-MB-435 breast cancer cells that overexpress thrombospondin-1. The role of urokinase plasminogen receptor in thrombospondin-1-mediated adhesion and invasion was studied by antisense inhibition, enzymatic cleavage and antibody neutralization. Tumour cell adhesion to collagen and laminin was evaluated. Tumour cell invasion was studied in a modified Boyden chamber collagen invasion assay. Tumour cell thrombospondin-1 induced a 2-7 fold increase in urokinase plasminogen activator receptor and cell-associated urokinase plasminogen activator expression and a 50-65% increase in cell-associated urokinase plasminogen activator and plasmin activities. Furthermore, tumour cell thrombospondin-1 promoted tumour cell invasion and decreased tumour cell adhesion through up-regulation of urokinase plasminogen activator receptor-controlled urokinase plasminogen activator and plasmin activities. We conclude that tumour cell-produced thrombospondin-1 may play a critical role in the regulation of tumour cell adhesion and tumour cell invasion.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Cell Surface/metabolism , Thrombospondin 1/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Blotting, Western , Breast Neoplasms/pathology , Cell Adhesion , Enzyme-Linked Immunosorbent Assay , Fibrinolysin/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Plasminogen/metabolism , Receptors, Urokinase Plasminogen Activator , Transfection , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured , Up-Regulation
13.
Invest Ophthalmol Vis Sci ; 41(8): 2378-88, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10892887

ABSTRACT

PURPOSE: Thrombospondin (TSP)1 is a tumor suppressor with activity that is associated with its ability to inhibit neovascularization. Previous studies have mapped this antiangiogenic activity to the type 1 repeats and the amino-terminal portion of the molecule within the procollagen-like domain. The present study was performed to investigate the ability of TSP-1 and peptides derived from the type 1 repeats to inhibit retinal angiogenesis. METHODS: TSP-1 and peptides with tryptophan-rich, heparin-binding sequences and transforming growth factor (TGF)-beta1 activation sequences were evaluated in two models of retinal angiogenesis: a retinal explant assay and a rat model of retinopathy of prematurity (ROP). RESULTS: Platelet-derived TSP-1 inhibited angiogenesis in both experimental models. Peptides from the native TSP-1 sequence, which contained both the tryptophan-rich repeat and the TGF-beta1 activation sequence, were the most potent inhibitors of endothelial cell outgrowth in the retinal explant assay. In contrast, a peptide containing only the tryptophan-rich, heparin-binding sequence was most active in inhibiting neovascular disease in the rat ROP model. CONCLUSIONS: These results indicate that the type 1 repeats of TSP-1 contain two subdomains that may independently influence the process of neovascularization, and that peptides derived from these type 1 repeats may be promising pharmacologic agents for treatment of retinal angiogenesis.


Subject(s)
Peptide Fragments/pharmacology , Retinal Neovascularization/prevention & control , Thrombospondin 1/pharmacology , Amino Acid Sequence , Animals , Animals, Newborn , Cattle , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Infant, Newborn , Male , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Retinal Neovascularization/pathology , Retinal Vessels/drug effects , Retinal Vessels/pathology , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/prevention & control , Time Factors
14.
J Agric Food Chem ; 48(6): 2430-7, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10888563

ABSTRACT

Solid-phase microextraction (SPME) fibers were evaluated for their ability to adsorb volatile flavor compounds under various conditions with coffee and aqueous flavored solutions. Experiments comparing different fibers showed that poly(dimethylsiloxane)/divinylbenzene had the highest overall sensitivity. Carboxen/poly(dimethylsiloxane) was the most sensitive to small molecules and acids. As the concentrations of compounds increased, the quantitative linear range was exceeded as shown by competition effects with 2-isobutyl-3-methoxypyrazine at concentrations above 1 ppm. A method based on a short-time sampling of the headspace (1 min) was shown to better represent the equilibrium headspace concentration. Analysis of coffee brew with a 1-min headspace adsorption time was verified to be within the linear range for most compounds and thus appropriate for relative headspace quantification. Absolute quantification of volatiles, using isotope dilution assays (IDA), is not subject to biases caused by excess compound concentrations or complex matrices. The degradation of coffee aroma volatiles during storage was followed by relative headspace measurements and absolute quantifications. Both methods gave similar values for 3-methylbutanal, 4-ethylguaiacol, and 2,3-pentanedione. Acetic acid, however, gave higher values during storage upon relative headspace measurements due to concurrent pH decreases that were not seen with IDA.


Subject(s)
Coffee/chemistry , Taste , Chromatography, Gas/instrumentation , Chromatography, Gas/methods , Chromatography, High Pressure Liquid/instrumentation , Chromatography, High Pressure Liquid/methods , Gas Chromatography-Mass Spectrometry/methods , Microchemistry
15.
Cancer Res ; 60(2): 457-66, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10667601

ABSTRACT

Although human small cell lung carcinoma (SCLC) cell lines are typically anchorage-independent and do not attach on most extracellular matrix proteins, OH-1, and several other SCLC cell lines attached on substrates coated with thrombospondin-1 (TSP1). SCLC cells grew long-term as adherent cells on a TSP1-coated substrate. Adhesion of SCLC cells on TSP1 was inhibited by heparin, function-blocking antibodies recognizing alpha3 or beta1 integrin subunits, and by soluble alpha3beta1 integrin ligands. SCLC cells extended neurite-like processes on a TSP1 substrate, which was also mediated by alpha3beta1 integrin. Process formation on a TSP1 substrate was specifically stimulated by epidermal growth factor and somatostatin. Adhesion on TSP1 weakly inhibited SCLC cell proliferation, but this inhibition was strongly enhanced in the presence of epidermal growth factor. TSP1 and an alpha3beta1 integrin-binding peptide from TSP1 also inhibited proliferation when added in solution. High-affinity binding of 125I-labeled TSP1 to OH-1 cells was heparin-dependent and may be mediated by sulfated glycolipids, which are the major sulfated glycoconjugates synthesized by these cells. Synthesis or secretion of TSP1 by SCLC cells could not be detected. On the basis of these results, the alpha3beta1 integrin and sulfated glycolipids cooperate to mediate adhesion of SCLC cells on TSP1. Interaction with TSP1 through this integrin inhibits growth and induces neurotypic differentiation, which suggests that this response to TSP1 may be exploited to inhibit the progression of SCLC.


Subject(s)
Cell Adhesion/drug effects , Integrins/physiology , Neurites/physiology , Thrombospondin 1/physiology , Carcinoma, Small Cell , Cell Division/drug effects , Epidermal Growth Factor/pharmacology , Humans , Integrin alpha3beta1 , Integrins/drug effects , Kinetics , Lung Neoplasms , Neurites/drug effects , Neurites/ultrastructure , Somatostatin/pharmacology , Thrombospondin 1/pharmacokinetics , Thrombospondin 1/pharmacology , Tumor Cells, Cultured
16.
Arch Biochem Biophys ; 374(1): 13-23, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10640391

ABSTRACT

Interactions of heparin with intact human thrombospondin-1 (TSP1) and with two heparin-binding fragments of TSP1 were characterized using chemically modified heparins, a vascular heparan sulfate proteoglycan, and a series of heparin oligosaccharides prepared by partial deaminative cleavage. The avidity of TSP1 binding increased with oligosaccharide size, with plateaus at 4 to 6 and at 8 to 10 monosaccharide units. The dependence on oligosaccharide size for binding to the recombinant amino-terminal heparin-binding domain of TSP1 was the same as that of the intact TSP1 molecule but differed from that of a synthetic heparin-binding peptide from the type 1 repeats, suggesting that the interaction between intact TSP1 and heparin is primarily mediated by the amino-terminal domain. Based on activities of chemically modified heparins, binding to TSP1 depended primarily on 2-N- and 6-O-sulfation of glucosamine and to a lesser degree on 2,3-O-sulfation and the carboxyl residues of the uronic acids. In contrast, all of these modifications were required for binding of heparin to the type 1 repeat peptides. Affinity purification of heparin octasaccharides on immobilized TSP1 type 1 repeat peptides revealed a preference for oligosaccharides containing the disaccharide sequence IdoA(2-OSO(3))alpha1-4-GlcNS(6-OSO(3)). Binding of these oligosaccharides to the peptide required the Trp residues. These data demonstrate that the heparin-binding specificities of intact TSP1 and peptides from the type 1 repeats overlap with that of basic fibroblast growth factor (FGF2) and are consistent with the ability of these TSP1-derived molecules to inhibit FGF2-stimulated angiogenesis.


Subject(s)
Heparin/chemistry , Recombinant Proteins/chemistry , Thrombospondin 1/chemistry , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Cattle , Chromatography, Affinity , Chromatography, High Pressure Liquid/methods , Heparin/metabolism , Humans , Kinetics , Molecular Sequence Data , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Radioligand Assay , Recombinant Proteins/metabolism , Repetitive Sequences, Amino Acid , Swine , Thrombospondin 1/genetics , Thrombospondin 1/metabolism
17.
J Immunol ; 163(7): 3621-8, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10490955

ABSTRACT

Cell-cell and cell-matrix interactions play important regulatory roles in lymphocyte homeostasis. Thrombospondin-1 (TSP1) is a matricellular protein that differentially promotes the adhesion of resting and activated T cells. In this work, we show that adhesion of Jurkat T cells on substrates coated with TSP1 or TSP1-derived peptides is mediated by beta 1 integrins, CD47, and heparan sulfate proteoglycans. Interactions with TSP1 or TSP1 peptides stimulated CD3-induced Ras activation and tyrosine phosphorylation of several T cell proteins. The signals from TSP1 and its derived peptides differentially synergized with activation of the TCR to induce phosphorylation of linker for activation of T cells (LAT) and extracellular signal-regulated kinase (ERK) 1/2, c-Jun N-terminal kinase, and p38 kinases. The phosphorylation of ERK in the presence of full-length TSP1 was transient and dependent on a beta 1 integrin receptor. Interestingly, peptides derived from the type 1 repeats of TSP1 and a CD47-binding peptide from the carboxyl-terminal domain of TSP1 also stimulated mitogen-activated protein (MAP) kinase phosphorylation. Moreover, the TSP1 heparin-binding peptide synergized with Ab-ligated TCR to transduce signals to the nucleus, detected by activation of AP-1- and Elk-dependent transcription. This TSP1 peptide-dependent activation of AP-1 was inhibited by both heparin and the MAP/ERK kinase inhibitor PD98059, providing a functional link between adhesion molecule interaction and nuclear transactivation events via the MAP kinase pathways. These findings have implications for the role of extracellular TSP1 and TSP1 fragments in the regulation of T cell function during hemostasis, wound repair, and other inflammatory responses.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , DNA-Binding Proteins , Integrin beta1/physiology , Lymphocyte Activation/immunology , Lymphoma, T-Cell/immunology , Mitogen-Activated Protein Kinases , Peptide Fragments/physiology , Proteoglycans/physiology , Signal Transduction/immunology , Thrombospondin 1/physiology , Transcription Factors , Amino Acid Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Adhesion/immunology , Cell Nucleus/metabolism , GTP-Binding Proteins/physiology , Glycosaminoglycans/physiology , Heparin/metabolism , Humans , JNK Mitogen-Activated Protein Kinases , Jurkat Cells , Lymphoma, T-Cell/metabolism , Molecular Sequence Data , Oncogene Protein p21(ras)/biosynthesis , Oncogene Protein p21(ras)/metabolism , Peptide Fragments/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/physiology , Sulfates/metabolism , Thrombospondin 1/metabolism , Transcription Factor AP-1/physiology , Transcriptional Activation/immunology , Tyrosine/metabolism , ets-Domain Protein Elk-1 , p38 Mitogen-Activated Protein Kinases
18.
Circulation ; 100(13): 1423-31, 1999 Sep 28.
Article in English | MEDLINE | ID: mdl-10500044

ABSTRACT

BACKGROUND: Suppression of tumor growth by thrombospondin-1 (TSP-1) has been associated with its ability to inhibit neovascularization. The antiangiogenic activity of TSP-1, as defined by cornea pocket assays, was previously mapped to the amino-terminal portion of the protein within the procollagen region and the type 1 repeats. METHODS AND RESULTS: We evaluated the specificity and efficacy of different regions of TSP-1 using recombinant fragments of the protein on chorioallantoic membrane (CAM) angiogenesis and endothelial cell proliferation assays. In both assays, fragments containing the second and third type 1 repeats but not the procollagen region inhibited angiogenesis and endothelial cell proliferation. To further define the sequences responsible for the angiostatic effect of TSP-1, we used synthetic peptides. The CAM assay defined 2 sequences that independently suppressed angiogenesis. The amino-terminal end of the type 1 repeats showed higher potency for inhibiting angiogenesis driven by basic fibroblast growth factor (FGF-2), whereas the second region equally blocked angiogenesis driven by either FGF-2 or vascular endothelial growth factor (VEGF). Modifications of the active peptides revealed the specific amino acids required for the inhibitory response. One sequence included the conserved tryptophan residues in the amino-terminal end of the second and third type 1 repeats, and the other involved the amino acids that follow the CSVTCG sequence in the carboxy-terminus of these repeats. Both inhibition in the CAM assay and inhibition of breast tumor xenograft growth in nude mice were independent of the TGF-beta-activating sequence located in the second type 1 repeat. CONCLUSIONS: These results indicate that the type 1 repeats of TSP-1 contain 2 subdomains that may independently inhibit neovascularization. They also identify 2 independent pathways by which TSP-1 can block FGF-2 and VEGF angiogenic signals on endothelial cells.


Subject(s)
Neovascularization, Pathologic/prevention & control , Thrombospondin 1/genetics , Thrombospondin 1/pharmacology , Amino Acid Sequence/genetics , Animals , Cell Division/drug effects , Chick Embryo , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Extraembryonic Membranes/blood supply , Extraembryonic Membranes/drug effects , Female , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblast Growth Factor 2/physiology , Lymphokines/antagonists & inhibitors , Lymphokines/physiology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Nude , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Recombinant Proteins , Repetitive Sequences, Amino Acid , Signal Transduction/drug effects , Transplantation, Heterologous , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
J Biol Chem ; 274(34): 24080-6, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10446179

ABSTRACT

A synthetic peptide containing amino acid residues 190-201 of thrombospondin-1 (TSP1) promoted adhesion of MDA-MB-435 breast carcinoma cells when immobilized and inhibited adhesion of the same cells to TSP1 when added in solution. Adhesion to this peptide was enhanced by a beta(1) integrin-activating antibody, Mn(2+), and insulin-like growth factor I and was inhibited by an alpha(3)beta(1) integrin function-blocking antibody. The soluble peptide inhibited adhesion of cells to the immobilized TSP1 peptide or spreading on intact TSP1 but at the same concentrations did not inhibit attachment or spreading on type IV collagen or fibronectin. Substitution of several residues in the TSP1 peptide with Ala residues abolished or diminished the inhibitory activity of the peptide in solution, but only substitution of Arg-198 completely inactivated the adhesive activity of the immobilized peptide. The essential residues for activity of the peptide as a soluble inhibitor are Asn-196, Val-197, and Arg-198, but flanking residues enhance the inhibitory activity of this core sequence, either by altering the conformation of the active sequence or by interacting with the integrin. This functional sequence is conserved in all known mammalian TSP1 sequences and in TSP1 from Xenopus laevis. The TSP1 peptide also inhibited adhesion of MDA-MB-435 cells to the laminin-1 peptide GD6, which contains a potential integrin-recognition sequence Asn-Leu-Arg and is derived from a similar position in a pentraxin module. Adhesion studies using recombinant TSP1 fragments also localized beta1 integrin-dependent adhesion to residues 175-242 of this region, which contain the active sequence.


Subject(s)
Integrins/chemistry , Thrombospondin 1/chemistry , Amino Acid Sequence , Animals , Binding Sites , Calcium/pharmacology , Cell Adhesion , Chemotaxis/drug effects , Humans , Integrin alpha3beta1 , Integrins/metabolism , Laminin/chemistry , Magnesium/pharmacology , Mice , Molecular Sequence Data , Structure-Activity Relationship , Thrombospondin 1/metabolism , Thrombospondin 1/pharmacology
20.
Mycoses ; 42(4): 255-9, 1999.
Article in English | MEDLINE | ID: mdl-10424092

ABSTRACT

In order to study the interactions between Candida species and uroepithelial tissue, a tissue explant assay was developed using bladder mucosa harvested from New Zealand white rabbits. Blastoconidia of Candida albicans, Candida tropicalis and Candida glabrata attached to the uroepithelial tissue in similar quantities. However, there was significantly more adherence to the uroepithelium by pre-germinated C. albicans compared with C. albicans blastoconidia. Furthermore, the amount of uroepithelial tissue injury was directly related to the length of exposure of the tissue to Candida. Thus, this tissue explant assay may provide a useful method for investigating properties related to fungal adherence to transitional uroepithelium and organism-mediated tissue injury.


Subject(s)
Candida albicans/physiology , Urinary Bladder/microbiology , Animals , Cell Adhesion/physiology , Culture Techniques , Female , Mucous Membrane/microbiology , Rabbits , Specific Pathogen-Free Organisms
SELECTION OF CITATIONS
SEARCH DETAIL
...