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










Publication year range
1.
J Am Heart Assoc ; 7(15)2018 07 18.
Article in English | MEDLINE | ID: mdl-30021807

ABSTRACT

BACKGROUND: The proximal aorta normally functions as a critical shock absorber that protects small downstream vessels from damage by pressure and flow pulsatility generated by the heart during systole. This shock absorber function is impaired with age because of aortic stiffening. METHODS AND RESULTS: We examined the contribution of common genetic variation to aortic stiffness in humans by interrogating results from the AortaGen Consortium genome-wide association study of carotid-femoral pulse wave velocity. Common genetic variation in the N-WASP (WASL) locus is associated with carotid-femoral pulse wave velocity (rs600420, P=0.0051). Thus, we tested the hypothesis that decoy proteins designed to disrupt the interaction of cytoskeletal proteins such as N-WASP with its binding partners in the vascular smooth muscle cytoskeleton could decrease ex vivo stiffness of aortas from a mouse model of aging. A synthetic decoy peptide construct of N-WASP significantly reduced activated stiffness in ex vivo aortas of aged mice. Two other cytoskeletal constructs targeted to VASP and talin-vinculin interfaces similarly decreased aging-induced ex vivo active stiffness by on-target specific actions. Furthermore, packaging these decoy peptides into microbubbles enables the peptides to be ultrasound-targeted to the wall of the proximal aorta to attenuate ex vivo active stiffness. CONCLUSIONS: We conclude that decoy peptides targeted to vascular smooth muscle cytoskeletal protein-protein interfaces and microbubble packaged can decrease aortic stiffness ex vivo. Our results provide proof of concept at the ex vivo level that decoy peptides targeted to cytoskeletal protein-protein interfaces may lead to substantive dynamic modulation of aortic stiffness.


Subject(s)
Aging , Aorta, Thoracic/physiopathology , Cytoskeletal Proteins/genetics , Hypertension/physiopathology , Muscle, Smooth, Vascular/physiopathology , Polymorphism, Single Nucleotide , Vascular Stiffness/physiology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Blood Pressure , Cells, Cultured , Cytoskeletal Proteins/metabolism , DNA/genetics , Genome-Wide Association Study/methods , Humans , Hypertension/genetics , Hypertension/pathology , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Pulse Wave Analysis
2.
Invest Ophthalmol Vis Sci ; 56(9): 5218-28, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26244298

ABSTRACT

PURPOSE: Chronic inflammation of the lacrimal gland results in changes in the composition of the extracellular matrix (ECM), which is believed to compromise tissue repair. We hypothesized that increased production/activity of matrix metalloproteinases (MMPs), especially MMP-2 and -9, in inflamed lacrimal glands modifies the ECM environment, therefore disrupting tissue repair. METHODS: The lacrimal glands from female MRL/lpr and male NOD mice along with their respective control strains were harvested and divided into three pieces and processed for histology, immunohistochemistry, zymography, Western blotting, and RNA analyses. In another study, MRL/lpr mice were treated for 5 weeks with a selective MMP2/9 inhibitor peptide or a control peptide. At the end of treatment, the lacrimal glands were excised and the tissue was processed as described above. RESULTS: There was a 2.5- and 2.7-fold increase in MMP2 gene expression levels in MRL/lpr and NOD mice, respectively. Matrix metalloproteinase 2 and 9 enzymatic activities and protein expression levels were significantly upregulated in the lacrimal glands of MRL/lpr and NOD mice compared to controls. Treatment with the MMP2/9 inhibitor resulted in decreased activity of MMP-2 and -9 both in vitro and in vivo. Importantly, MMP2/9 inhibitor treatment of MRL/lpr mice improved aqueous tear production and resulted in reduced number and size of lymphocytic foci in diseased lacrimal glands. CONCLUSIONS: We conclude that MMP2/9 expression and activity are elevated in lacrimal glands of two murine models of Sjögren's syndrome, suggesting that manipulation of MMP2/9 activity might be a potential therapeutic target in chronically inflamed lacrimal glands.


Subject(s)
Gene Expression Regulation , Lacrimal Apparatus/enzymology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , RNA/genetics , Sjogren's Syndrome/genetics , Tears/enzymology , Animals , Blotting, Western , Disease Models, Animal , Female , Immunohistochemistry , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Mice, Inbred NOD , Real-Time Polymerase Chain Reaction , Sjogren's Syndrome/enzymology
3.
Chem Biol ; 21(3): 331-7, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24529990

ABSTRACT

Arginylation is an emerging posttranslational modification mediated by Arg-tRNA-protein-transferase (ATE1). It is believed that ATE1 links Arg solely to the N terminus of proteins, requiring prior proteolysis or action by Met-aminopeptidases to expose the arginylated site. Here, we tested the possibility of Arg linkage to midchain sites within intact protein targets and found that many proteins in vivo are modified on the side chains of Asp and Glu by unconventional chemistry that targets the carboxy rather than the amino groups at the target sites. Such arginylation appears to be functionally regulated, and it can be directly mediated by ATE1, in addition to the more conventional ATE1-mediated linkage of Arg to the N-terminal alpha amino group. This midchain arginylation implies an unconventional mechanism of ATE1 action that likely facilitates its major biological role.


Subject(s)
Aminoacyltransferases/metabolism , Aminoacyltransferases/chemistry , Aminoacyltransferases/genetics , Angiotensin II/analysis , Angiotensin II/chemistry , Angiotensin II/metabolism , Animals , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Biocatalysis , Chromatography, High Pressure Liquid , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Humans , Mice , Protein Processing, Post-Translational , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity
4.
Biosci Rep ; 33(2)2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23289753

ABSTRACT

CaMKII (Ca²âº/calmodulin-dependent kinase II) is a serine/threonine phosphotransferase that is capable of long-term retention of activity due to autophosphorylation at a specific threonine residue within each subunit of its oligomeric structure. The γ isoform of CaMKII is a significant regulator of vascular contractility. Here, we show that phosphorylation of CaMKII γ at Ser²6, a residue located within the ATP-binding site, terminates the sustained activity of the enzyme. To test the physiological importance of phosphorylation at Ser²6, we generated a phosphospecific Ser²6 antibody and demonstrated an increase in Ser²6 phosphorylation upon depolarization and contraction of blood vessels. To determine if the phosphorylation of Ser²6 affects the kinase activity, we mutated Ser²6 to alanine or aspartic acid. The S26D mutation mimicking the phosphorylated state of CaMKII causes a dramatic decrease in Thr²87 autophosphorylation levels and greatly reduces the catalytic activity towards an exogenous substrate (autocamtide-3), whereas the S26A mutation has no effect. These data combined with molecular modelling indicate that a negative charge at Ser²6 of CaMKII γ inhibits the catalytic activity of the enzyme towards its autophosphorylation site at Thr²87 most probably by blocking ATP binding. We propose that Ser²6 phosphorylation constitutes an important mechanism for switching off CaMKII activity.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Protein Isoforms/metabolism , Animals , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calmodulin/genetics , Calmodulin/metabolism , Ferrets/genetics , Ferrets/metabolism , Mutation , Phosphorylation/genetics , Protein Isoforms/genetics , Serine/genetics , Serine/metabolism , Signal Transduction , Threonine/genetics , Threonine/metabolism
5.
Am J Physiol Cell Physiol ; 300(6): C1356-65, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21289288

ABSTRACT

Tropomyosin (Tm) is known to be an important gatekeeper of actin function. Tm isoforms are encoded by four genes, and each gene produces several variants by alternative splicing, which have been proposed to play roles in motility, proliferation, and apoptosis. Smooth muscle studies have focused on gizzard smooth muscle, where a heterodimer of Tm from the α-gene (Tmsm-α) and from the ß-gene (Tmsm-ß) is associated with contractile filaments. In this study we examined Tm in differentiated mammalian vascular smooth muscle (dVSM). Liquid chromatography-tandem mass spectrometry (LC MS/MS) analysis and Western blot screening with variant-specific antibodies revealed that at least five different Tm proteins are expressed in this tissue: Tm6 (Tmsm-α) and Tm2 from the α-gene, Tm1 (Tmsm-ß) from the ß-gene, Tm5NM1 from the γ-gene, and Tm4 from the δ-gene. Tm6 is by far most abundant in dVSM followed by Tm1, Tm2, Tm5NM1, and Tm4. Coimmunoprecipitation and coimmunofluorescence studies demonstrate that Tm1 and Tm6 coassociate with different actin isoforms and display different intracellular localizations. Using an antibody specific for cytoplasmic γ-actin, we report here the presence of a γ-actin cortical cytoskeleton in dVSM cells. Tm1 colocalizes with cortical cytoplasmic γ-actin and coprecipitates with γ-actin. Tm6, on the other hand, is located on contractile bundles. These data indicate that Tm1 and Tm6 do not form a classical heterodimer in dVSM but rather describe different functional cellular compartments.


Subject(s)
Cell Differentiation/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Tropomyosin/chemistry , Tropomyosin/metabolism , Actins/genetics , Actins/metabolism , Amino Acid Sequence , Animals , Chickens , Ferrets , Humans , Molecular Sequence Data , Myocytes, Smooth Muscle/cytology , Protein Binding , Protein Isoforms/genetics , Sequence Alignment , Tropomyosin/genetics
6.
J Mol Biol ; 403(1): 11-23, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20804767

ABSTRACT

Actin filament nucleators initiate polymerization in cells in a regulated manner. A common architecture among these molecules consists of tandem WASP homology 2 domains (W domains) that recruit three to four actin subunits to form a polymerization nucleus. We describe a low-resolution crystal structure of an actin dimer assembled by tandem W domains, where the first W domain is cross-linked to Cys374 of the actin subunit bound to it, whereas the last W domain is followed by the C-terminal pointed end-capping helix of thymosin ß4. While the arrangement of actin subunits in the dimer resembles that of a long-pitch helix of the actin filament, important differences are observed. These differences result from steric hindrance of the W domain with intersubunit contacts in the actin filament. We also determined the structure of the first W domain of Vibrio parahaemolyticus VopL cross-linked to actin Cys374 and show it to be nearly identical with non-cross-linked W-Actin structures. This result validates the use of cross-linking as a tool for the study of actin nucleation complexes, whose natural tendency to polymerize interferes with most structural methods. Combined with a biochemical analysis of nucleation, the structures may explain why nucleators based on tandem W domains with short inter-W linkers have relatively weak activity, cannot stay bound to filaments after nucleation, and are unlikely to influence filament elongation. The findings may also explain why nucleation-promoting factors of the Arp2/3 complex, which are related to tandem-W-domain nucleators, are ejected from branch junctions after nucleation. We finally show that the simple addition of the C-terminal pointed end-capping helix of thymosin ß4 to tandem W domains can change their activity from actin filament nucleation to monomer sequestration.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/chemistry , Actins/metabolism , Protein Multimerization , Animals , Bacterial Proteins/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Structure, Quaternary , Rabbits , Vibrio parahaemolyticus/chemistry
7.
Am J Physiol Cell Physiol ; 299(5): C988-93, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20686075

ABSTRACT

Here we report and validate a new method, suitable broadly, for use in differentiated cells and tissues, for the direct visualization of actin polymerization under physiological conditions. We have designed and tested different versions of fluorescently labeled actin, reversibly attached to the protein transduction tag TAT, and have introduced this novel reagent into intact differentiated vascular smooth muscle cells (dVSMCs). A thiol-reactive version of the TAT peptide was synthesized by adding the amino acids glycine and cysteine to its NH(2)-terminus and forming a thionitrobenzoate adduct: viz. TAT-Cys-S-STNB. This peptide reacts readily with G-actin, and the complex is rapidly taken up by freshly enzymatically isolated dVSMC, as indicated by the fluorescence of a FITC tag on the TAT peptide. By comparing different versions of the construct, we determined that the optimal construct for biological applications is a nonfluorescently labeled TAT peptide conjugated to rhodamine-labeled actin. When TAT-Cys-S-STNB-tagged rhodamine actin (TSSAR) was added to live, freshly enzymatically isolated cells, we observed punctae of incorporated actin at the cortex of the cell. The punctae are indistinguishable from those we have previously reported to occur in the same cell type when rhodamine G-actin is added to permeabilized cells. Thus this new method allows the delivery of labeled G-actin into intact cells without disrupting the native state and will allow its further use to study the effect of physiological intracellular Ca(2+) concentration transients and signal transduction on actin dynamics in intact cells.


Subject(s)
Actins/metabolism , Cell Differentiation , Fluorescent Dyes/metabolism , Muscle, Smooth, Vascular/metabolism , Protein Multimerization , Staining and Labeling/methods , Actins/chemistry , Fluorescent Dyes/chemistry , Gene Products, tat/genetics , Gene Products, tat/metabolism , Molecular Structure , Peptides/chemical synthesis , Peptides/genetics , Peptides/metabolism
8.
J Biol Chem ; 284(26): 17607-15, 2009 Jun 26.
Article in English | MEDLINE | ID: mdl-19406750

ABSTRACT

ERK influences a number of pathways in all cells, but how ERK activities are segregated between different pathways has not been entirely clear. Using immunoprecipitation and pulldown experiments with domain-specific recombinant fragments, we show that smooth muscle archvillin (SmAV) binds ERK and members of the ERK signaling cascade in a domain-specific, stimulus-dependent, and pathway-specific manner. MEK binds specifically to the first 445 residues of SmAV. B-Raf, an upstream regulator of MEK, constitutively interacts with residues 1-445 and 446-1250. Both ERK and 14-3-3 bind to both fragments, but in a stimulus-specific manner. Phosphorylated ERK is associated only with residues 1-445. An ERK phosphorylation site was determined by mass spectrometry to reside at Ser132. A phospho-antibody raised to this site shows that the site is phosphorylated during alpha-agonist-mediated ERK activation in smooth muscle tissue. Phosphorylation of SmAV by ERK decreases the association of phospho-ERK with SmAV. These results, combined with previous observations, indicate that SmAV serves as a new ERK scaffolding protein and provide a mechanism for regulation of ERK binding, activation, and release from the signaling complex.


Subject(s)
Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Muscle, Smooth/metabolism , 14-3-3 Proteins/metabolism , Amino Acid Sequence , Animals , Aorta/cytology , Aorta/metabolism , Ferrets , Immunoprecipitation , Mass Spectrometry , Molecular Sequence Data , Muscle, Smooth/cytology , Phosphorylation , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction
9.
Am J Physiol Cell Physiol ; 295(3): C768-78, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18596213

ABSTRACT

Dynamic remodeling of the actin cytoskeleton plays an essential role in the migration and proliferation of vascular smooth muscle cells. It has been suggested that actin remodeling may also play an important functional role in nonmigrating, nonproliferating differentiated vascular smooth muscle (dVSM). In the present study, we show that contractile agonists increase the net polymerization of actin in dVSM, as measured by the differential ultracentrifugation of vascular smooth muscle tissue and the costaining of single freshly dissociated cells with fluorescent probes specific for globular and filamentous actin. Furthermore, induced alterations of the actin polymerization state, as well as actin decoy peptides, inhibit contractility in a stimulus-dependent manner. Latrunculin pretreatment or actin decoy peptides significantly inhibit contractility induced by a phorbol ester or an alpha-agonist, but these procedures have no effect on contractions induced by KCl. Aorta dVSM expresses alpha-smooth muscle actin, beta-actin, nonmuscle gamma-actin, and smooth muscle gamma-actin. The incorporation of isoform-specific cell-permeant synthetic actin decoy peptides, as well as isoform-specific probing of cell fractions and two-dimensional gels, demonstrates that actin remodeling during alpha-agonist contractions involves the remodeling of primarily gamma-actin and, to a lesser extent, beta-actin. Taken together, these results show that net isoform- and agonist-dependent increases in actin polymerization regulate vascular contractility.


Subject(s)
Actins/metabolism , Cell Differentiation , Cytoskeleton/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Cytoskeleton/drug effects , Depsipeptides/pharmacology , Ferrets , In Vitro Techniques , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Peptide Fragments/pharmacology , Phalloidine/pharmacology , Phenylephrine/pharmacology , Phorbol Esters/pharmacology , Potassium Chloride/pharmacology , Protein Isoforms , Vasoconstriction , Vasoconstrictor Agents/pharmacology
10.
Am J Physiol Heart Circ Physiol ; 286(1): H91-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-12969891

ABSTRACT

Caveolin is a principal component of caveolar membranes. In the present study, we utilized a decoy peptide approach to define the degree of involvement of caveolin in PKC-dependent regulation of contractility of differentiated vascular smooth muscle. The primary isoform of caveolin in ferret aorta vascular smooth muscle is caveolin-1. Chemical loading of contractile vascular smooth muscle tissue with a synthetic caveolin-1 scaffolding domain peptide inhibited PKC-dependent increases in contractility induced by a phorbol ester or an alpha agonist. Peptide loading also resulted in a significant inhibition of phorbol ester-induced adducin Ser662 phosphorylation, an intracellular monitor of PKC kinase activity, ERK1/2 activation, and Ser789 phosphorylation of the actin binding protein caldesmon. alpha-Agonist-induced ERK1-1/2 activation was also inhibited by the caveolin-1 peptide. Scrambled peptide-loaded tissues or sham-loaded tissues were unaffected with respect to both contractility and signaling. Depolarization-induced activation of contraction was not affected by caveolin peptide loading. Similar results with respect to contractility and ERK1/2 activation during exposure to the phorbol ester or the alpha-agonist were obtained with the cholesterol-depleting agent methyl-beta-cyclodextrin. These results are consistent with a role for caveolin-1 in the coordination of signaling leading to the regulation of contractility of smooth muscle.


Subject(s)
Caveolins/physiology , Muscle, Smooth, Vascular/physiology , Vasoconstriction/physiology , Animals , Anticholesteremic Agents/pharmacology , Aorta/metabolism , Calmodulin-Binding Proteins/metabolism , Calmodulin-Binding Proteins/pharmacology , Caveolin 1 , Caveolins/pharmacology , Ferrets , Mitogen-Activated Protein Kinases/metabolism , Peptide Fragments/pharmacology , Phorbol Esters/pharmacology , Phosphorylation , Protein Kinase C/metabolism , Vasoconstriction/drug effects
11.
Endocrine ; 21(2): 185-95, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12897384

ABSTRACT

In this article we show that rabbit endometrial cells express leptin receptor and that human leptin triggers phosphorylation of signal transducer and activator of transcription 3 and up-regulates the expression of interleukin- 1 receptor type I as was previously found in human endometrial cells. Interestingly, leptin also upregulates the secretion of leukemia inhibitory factor and expression of its receptor by rabbit endometrial cells. Analysis of a structural model of the leptin-leptin receptor complex suggested that helices I and III of the human leptin structure were likely sites of interaction with the cytokine binding domain of leptin receptor. Accordingly, we synthesized a peptide (LPA-2) comprising helix III (residues 70-95) and investigated its ability to inhibit leptin receptor function. The effects of LPA-2 were assayed in rabbit endometrial cells, and an antileptin receptor antibody and a scrambled version of LPA-2 were used as positive and negative controls, respectively. LPA-2 binds specifically and with high affinity (Ki ~ 0.6 x 10-10 M) to leptin receptor and is a potent inhibitor of its functions in rabbit endometrial cells. Because leukemia inhibitory factor and interleukin- 1 have been implicated in embryo implantation, our results raise the possibility that the LPA-2-induced inhibition of leptin receptor may be exploited to study the actions of leptin in endometrium and in other tissues under conditions characterized by abnormal leptin production.


Subject(s)
Endometrium/metabolism , Leptin/metabolism , Peptide Fragments/metabolism , Receptors, Cell Surface/metabolism , Animals , Cells, Cultured , Endometrium/cytology , Endometrium/drug effects , Female , Humans , Interleukin-6/metabolism , Leptin/pharmacology , Leukemia Inhibitory Factor , Peptide Fragments/pharmacology , Rabbits , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/drug effects , Receptors, Interleukin-1/drug effects , Receptors, Interleukin-1/metabolism , Receptors, Leptin , Recombinant Proteins , Up-Regulation
12.
Mol Hum Reprod ; 9(3): 151-8, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12606591

ABSTRACT

We have previously shown that (i). leptin and leptin receptor (Ob-R) are expressed in the human endometrium, and (ii). leptin secretion is regulated in blastocyst and endometrial epithelial cell (EEC) co-cultures. Interleukin-1beta (IL-1beta) up-regulates leptin and Ob-R, and both cytokines up-regulate beta3 integrin expression in EEC. In the present investigation we examined the effect of leptin on the expression of the IL-1 system in EEC and endometrial stromal cells (ESC) cultured in a medium containing insulin, leptin or IL-1beta (0-3 nmol/l). Leptin stimulated IL-1 antagonist (IL-1Ra), IL-1beta secretion and expression of IL-1 receptor type I (IL-1R tI) in both cell types. IL-1beta and IL-1Ra secretion were down-regulated by IL-1R tI blockade using specific antibodies. Interestingly, leptin partially neutralized this effect. The blockade of Ob-R neutralized the effects of both leptin and IL-1beta on expression of the IL-1beta system and beta3 integrin and on phosphorylation of signal transducer and activator of transcription 3 (Stat3). These results suggest that leptin regulates the IL-1 system and that the blockade of functional Ob-R impairs leptin and IL-1beta functions at the endometrial level. Leptin could be an important molecule for implantation and a molecular mediator for actions of the IL-1 system. The fact that leptin, in the absence of IL-1, can trigger the expression of markers of endometrial receptivity and of the invasive trophoblast phenotype (as does IL-1), suggest that leptin could substitute for these IL-1 functions during the implantation process.


Subject(s)
Endometrium/metabolism , Interleukin-1/metabolism , Leptin/metabolism , Antibodies/metabolism , Cells, Cultured , Endometrium/cytology , Female , Humans , Integrin beta3/metabolism , Interleukin 1 Receptor Antagonist Protein , Receptors, Cell Surface/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Receptors, Leptin , Sialoglycoproteins/metabolism
13.
Clin Cancer Res ; 8(11): 3579-83, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429649

ABSTRACT

Prostate carcinogenesis involves transformation of zinc-accumulating normal epithelial cells to malignant cells, which do not accumulate zinc. In this study, we demonstrate by immunoblotting and immunohistochemistry that physiological levels of zinc inhibit activation of nuclear factor (NF)-kappa B transcription factor in PC-3 and DU-145 human prostate cancer cells, reduce expression of NF-kappa B-controlled antiapoptotic protein c-IAP2, and activate c-Jun NH(2)-terminal kinases. Preincubation of PC-3 cells with physiological concentrations of zinc sensitized tumor cells to tumor necrosis factor (TNF)-alpha, and paclitaxel mediated cell death as defined by terminal deoxynucleotidyl transferase-mediated nick end labeling assay. These results suggest one possible mechanism for the inhibitory effect of zinc on the development and progression of prostate malignancy and might have important consequences for the prevention and treatment of prostate cancer.


Subject(s)
NF-kappa B/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Zinc/pharmacology , Active Transport, Cell Nucleus , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis , Blotting, Western , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases , Male , Mitogen-Activated Protein Kinases/metabolism , Paclitaxel/pharmacology , Proteins/metabolism , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...