Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Epidemiol Biomarkers Prev ; 20(6): 1112-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21527583

ABSTRACT

BACKGROUND: TGF-ß acts as a suppressor of primary tumor initiation but has been implicated as a promoter of the later malignant stages. Here associations with risk of invasive breast cancer are assessed for single-nucleotide polymorphisms (SNP) tagging 17 genes in the canonical TGF-ß ALK5/SMADs 2&3 and ALK1/SMADs 1&5 signaling pathways: LTBP1, LTBP2, LTBP4, TGFB1, TGFB2, TGFB3, TGFBR1(ALK5), ALK1, TGFBR2, Endoglin, SMAD1, SMAD2, SMAD3, SMAD4, SMAD5, SMAD6, and SMAD7 [Approved Human Gene Nomenclature Committee gene names: ACVRL1 (for ALK1) and ENG (for Endoglin)]. METHODS: Three-hundred-fifty-four tag SNPs (minor allele frequency > 0.05) were selected for genotyping in a staged study design using 6,703 cases and 6,840 controls from the Studies of Epidemiology and Risk Factors in Cancer Heredity (SEARCH) study. Significant associations were meta-analyzed with data from the NCI Polish Breast Cancer Study (PBCS; 1,966 cases and 2,347 controls) and published data from the Breast Cancer Association Consortium (BCAC). RESULTS: Associations of three SNPs, tagging TGFB1 (rs1982073), TGFBR1 (rs10512263), and TGFBR2 (rs4522809), were detected in SEARCH; however, associations became weaker in meta-analyses including data from PBCS and BCAC. Tumor subtype analyses indicated that the TGFB1 rs1982073 association may be confined to increased risk of developing progesterone receptor negative (PR(-)) tumors [1.18 (95% CI: 1.09-1.28), 4.1 × 10(-5) (P value for heterogeneity of ORs by PR status = 2.3 × 10(-4))]. There was no evidence for breast cancer risk associations with SNPs in the endothelial-specific pathway utilizing ALK1/SMADs 1&5 that promotes angiogenesis. CONCLUSION: Common variation in the TGF-ß ALK5/SMADs 2&3 signaling pathway, which initiates signaling at the cell surface to inhibit cell proliferation, might be related to risk of specific tumor subtypes. IMPACT: The subtype specific associations require very large studies to be confirmed.


Subject(s)
Breast Neoplasms/etiology , Disease Susceptibility , Polymorphism, Single Nucleotide/genetics , Signal Transduction , Transforming Growth Factor beta/genetics , Aged , Case-Control Studies , Female , Humans , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Prospective Studies
2.
Biochem Genet ; 49(3-4): 213-25, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21161366

ABSTRACT

Latent transforming growth factor-ß binding protein-1 (LTBP1) has been implicated in the control of secretion, localization, and activation of TGFß (transforming growth factor-ß). We developed a quantitative reverse-transcriptase polymerase chain reaction (Q-RT-PCR) assay using an RNA internal standard to examine the expression of three alternatively spliced isoforms of LTBP1 (LTBP1Δ41, LTBP1Δ53, and LTBP1Δ55) in a variety of human tissues. The assays were also used to determine the expression of LTBP1L and LTBP1S isoforms and total LTBP1. The Q-RT-PCR assays were highly reproducible and showed that in most tissues LTBP1Δ55 and LTBP1L were minor components of LTBP1. The proportion of LTBP1Δ41 ranged from 2% of total LTBP1 mRNA in early coronary atherosclerotic lesions to 54% in advanced lesions.


Subject(s)
Alternative Splicing , Coronary Artery Disease/genetics , Latent TGF-beta Binding Proteins/genetics , RNA, Messenger/biosynthesis , Case-Control Studies , Coronary Artery Disease/metabolism , Coronary Vessels/metabolism , Epithelial Cells/metabolism , Female , Genetic Variation , Humans , Latent TGF-beta Binding Proteins/biosynthesis , Organ Specificity , Protein Isoforms , Reverse Transcriptase Polymerase Chain Reaction
3.
Circ J ; 75(1): 196-200, 2011.
Article in English | MEDLINE | ID: mdl-21071877

ABSTRACT

BACKGROUND: Transforming growth factor-ß (TGFß) and its receptors have been detected by immunohistochemistry in the normal vessel wall and in atherosclerotic lesions of human coronary arteries. However, TGFß is normally secreted as an inactive complex associated with a latent TGFß-binding protein (LTBP). Therefore, detection of TGFß antigen only in the arterial wall does not imply the activated form of the growth factor. METHODS AND RESULTS: In situ hybridization and immunohistochemistry demonstrated LTBP1 mRNA and protein expression throughout the media and intima of early coronary artery lesions, with the highest levels of protein at the luminal surface. In advanced lesions, LTBP1 mRNA and protein were detected mainly in regions of high cell density, such as the fibrous cap. CONCLUSIONS: Assays of the TGFß signalling pathway will be required to determine the activity associated with TGFß antigen in the vessel wall.


Subject(s)
Coronary Artery Disease/metabolism , Coronary Vessels/chemistry , Latent TGF-beta Binding Proteins/analysis , Tunica Intima/chemistry , Tunica Media/chemistry , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , Coronary Vessels/pathology , Disease Progression , Humans , Immunohistochemistry , In Situ Hybridization , Latent TGF-beta Binding Proteins/genetics , RNA, Messenger/analysis , Tunica Intima/pathology , Tunica Media/pathology
4.
J Biol Chem ; 283(18): 11897-904, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18285334

ABSTRACT

To study the function of the Krüppel-like transcription factor KLF13 in vivo, we generated mice with a disrupted Klf13 allele. Although Klf13(-/-) mice are viable, fewer mice were present at 3 weeks than predicted by Mendelian inheritance. Viable Klf13(-/-) mice had reduced numbers of circulating erythrocytes and a larger spleen. The spleen contained an increased number of Ter119(med)CD71(hi), Ter119(hi)CD71(hi), and Ter119(hi)CD71(med) cells but not Ter119(hi)CD71(-) cells, indicating an increase in less mature erythroblasts. A higher proportion of the Ter119(med)CD71(hi) cells were proliferating, indicating that the mice were under a degree of erythropoietic stress. These data indicate that KLF13 is involved in the normal control of erythropoiesis.


Subject(s)
Erythropoiesis , Kruppel-Like Transcription Factors/deficiency , Splenomegaly/physiopathology , Animals , Apoptosis/genetics , Base Sequence , Blood Cell Count , Bone Marrow/pathology , Cell Cycle Proteins/genetics , Cell Differentiation , Cell Proliferation , Erythroblasts/pathology , Gene Deletion , Gene Expression Regulation , Gene Targeting , Genotype , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Organ Size , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Splenomegaly/pathology
5.
Nat Med ; 12(6): 705-10, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16732278

ABSTRACT

Multivariate analysis of 1H-NMR spectra of blood sera was reported previously to predict angiographically defined advanced coronary artery disease (CAD) with >90% accuracy and specificity. The analysis depended mainly on the major lipid regions of the spectra, but many variables, including gender and drug treatment, affect lipid composition and are potential confounders. We have determined the predictive power of the same methodology for angiographically defined CAD using plasma samples from groups of male patients, classified by statin treatment, who had normal coronary arteries (NCAs) or CAD. Predictions for NCA and CAD groups were only 80.3% correct for patients not treated with statins and 61.3% for treated patients, compared with random correct predictions of 50%. A confidence limit of >99% was achieved for 36.2% of predictions for untreated groups and 6.2% for treated groups. Detection of CAD by 1H-NMR with >99% confidence was therefore very weak compared with angiography.


Subject(s)
Coronary Artery Disease/blood , Plasma/chemistry , Protons , Coronary Artery Disease/drug therapy , Coronary Artery Disease/pathology , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Multivariate Analysis , Nuclear Magnetic Resonance, Biomolecular , Predictive Value of Tests
6.
J Vasc Res ; 40(3): 234-43, 2003.
Article in English | MEDLINE | ID: mdl-12902636

ABSTRACT

NOV is a member of the CCN family of matricellular proteins. We have shown previously that NOV is strongly expressed by vascular smooth muscle cells (VSMCs) of the rat carotid artery. However, 7 days after injury, NOV expression is down-regulated, except near the luminal surface of the developing intima, where it is strongly expressed. These data suggested that NOV might be involved in the regulation of endothelial cell adhesion. NOV promoted the adhesion of human umbilical vein endothelial cells (HUVECs), which was abolished by anti-NOV antibody. HUVEC adhesion to NOV required divalent cations and was inhibited by GRGDS peptide, implicating integrins in the adhesion mechanism. Monoclonal antibodies (mAbs) against alphavbeta3 inhibited adhesion of HUVECs to NOV, and NOV was shown to bind to alphavbeta3. Anti-alpha5beta1 mAbs also inhibited HUVEC adhesion to NOV, but adhesion via alpha5beta1 was mediated by fibronectin. HUVEC adhesion to NOV caused intracellular signalling, as evidenced by increased phosphotyrosine content of focal adhesion kinase. Together with evidence that NOV expression in a variety of tissues is restricted to blood vessels containing VSMCs, these data are consistent with a role for NOV in endothelial cell adhesion in vascular homeostasis and in the response to injury.


Subject(s)
Endothelium, Vascular/physiology , Immediate-Early Proteins/physiology , Integrin alpha5beta1/physiology , Integrin alphaVbeta3/physiology , Intercellular Signaling Peptides and Proteins/physiology , Animals , Antibodies, Monoclonal/pharmacology , Cell Adhesion/physiology , Cells, Cultured , Connective Tissue Growth Factor , Endothelium, Vascular/cytology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/pharmacology , Integrin alpha5beta1/drug effects , Integrin alpha5beta1/immunology , Integrin alphaVbeta3/drug effects , Integrin alphaVbeta3/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Nephroblastoma Overexpressed Protein , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Rats , Tissue Distribution
7.
Biochem J ; 375(Pt 2): 457-63, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-12848620

ABSTRACT

We have previously identified a C2H2 zinc-finger transcription factor [BTEB3 (basal transcription element-binding protein 3)/KLF13 (Krüppel-like factor 13)] that activates the minimal promoter for the smooth muscle-specific SM22alpha gene in other types of cell. We show that recombinant BTEB3 binds to three TGGG motifs in the minimal SM22alpha promoter. By mutation analysis, only one of these boxes is required for BTEB3-dependent promoter activation in P19 cells and BTEB3 activates or inhibits reporter gene expression depending on the TGGG box to which it binds. Transient transfection experiments show that BTEB3 also activates reporter gene expression from the SM22alpha promoter in VSMCs (vascular smooth muscle cells). Similar studies showed that BTEB3 did not activate expression from the promoter regions of the smooth muscle myosin heavy chain or smooth muscle alpha-actin promoters, which contain similar sequences, implying that promoter activation by BTEB3 is selective. The expression of BTEB3 is readily detectable in VSMCs in vitro and is modulated in response to injury in vivo.


Subject(s)
Microfilament Proteins/genetics , Microsatellite Repeats , Muscle Proteins/genetics , Promoter Regions, Genetic/genetics , Trans-Activators/metabolism , Animals , Base Sequence , Binding Sites/genetics , Carotid Artery Injuries/genetics , Cell Line, Tumor , Cells, Cultured , Conserved Sequence/genetics , DNA/genetics , DNA/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression Regulation , In Situ Hybridization , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Sequence Homology, Nucleic Acid , Trans-Activators/genetics
8.
Cancer Res ; 63(10): 2610-5, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12750287

ABSTRACT

There is evidence that transforming growth factor (TGF)beta acts as a suppressor of tumor initiation but also as a promoter of tumor progression when the antiproliferative effect of the TGFbeta signaling pathway has been overridden by other oncogenic mutations. Several somatic mutations that disrupt the TGFbeta-SMAD signaling pathway have been reported in human breast tumors. We have examined the association between single nucleotide polymorphisms (SNPs) in the TGFbeta1 gene and the incidence of invasive breast cancer in three case-control series, with a maximum of 3987 patients and 3867 controls, median age approximately 50 years, and range 22-92 years. The promoter SNP, C-509T, and the T +29C signal-peptide SNP (encoding Leu10Pro) are in strong linkage disequilibrium. They are both significantly associated with increased incidence of invasive breast cancer in a recessive manner [odds ratios: (TT versus C-carrier), 1.25; 95% confidence intervals 1.06-1.48; P = 0.009 and (ProPro versus Leu-carrier), 1.21; 95% confidence intervals 1.05-1.37; P = 0.01]. The G-800A SNP was not significantly associated with incidence of breast cancer. The C-509T SNP is not contained within a known consensus sequence for a promoter regulatory element and therefore unlikely to affect TGFbeta1 expression, whereas the Leu10Pro signal peptide substitution potentially affects TGFbeta1 secretion. Transfections of HeLa cells with constructs encoding either the Pro or Leu forms of TGFbeta1 and driven by the cytomegalovirus promoter indicate that the signal peptide with Pro at residue 10 causes a 2.8-fold increase in secretion compared with the Leu form. These data indicate that the allele encoding Pro10 is associated with increased rates of TGFbeta1 secretion and with increased incidence of invasive breast cancer for the population samples described. It is estimated that 3% of all breast cancer cases may be attributable to Pro10 homozygosity.


Subject(s)
Breast Neoplasms/pathology , Protein Sorting Signals/physiology , Transforming Growth Factor beta/physiology , Amino Acid Substitution , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Case-Control Studies , HeLa Cells , Humans , Leucine/genetics , Neoplasm Invasiveness , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Proline/genetics , Promoter Regions, Genetic , Protein Sorting Signals/genetics , Transfection , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
9.
Biochem J ; 364(Pt 2): 547-54, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12023898

ABSTRACT

Recent evidence has implicated CC(A/T(richG))GG (CArG) boxes, binding sites for serum response factor (SRF), in the regulation of expression of a number of genes in response to changes in the actin cytoskeleton. In many cases, the activity of SRF at CArG boxes is modulated by transcription factors binding to overlapping (e.g. Yin Yang 1, YY1) or adjacent (e.g. ets) binding sites. However, the mechanisms by which SRF activity is regulated by the cytoskeleton have not been determined. To investigate these mechanisms, we screened for cells that did or did not increase the activity of a fragment of the promoter for a smooth-muscle (SM)-specific gene SM22alpha, in response to changes in actin cytoskeletal polymerization induced by LIM kinase. These experiments showed that vascular SM cells (VSMCs) and C2C12 cells increased the activity of promoters containing at least one of the SM22alpha CArG boxes (CArG near) in response to LIM kinase, whereas P19 cells did not. Bandshift assays using a probe to CArG near showed that P19 cells lacked detectable YY1 DNA binding to the CArG box in contrast with the other two cell types. Expression of YY1 in P19 cells inhibited SM22alpha promoter activity and conferred responsiveness to LIM kinase. Mutation of the CArG box to inhibit YY1 or SRF binding indicated that both factors were required for the LIM kinase response in VSMCs and C2C12 cells. The data indicate that changes in the actin cytoskeletal organization modify SRF activity at CArG boxes by modulating YY1-dependent inhibition.


Subject(s)
Actins/metabolism , Biopolymers/metabolism , DNA-Binding Proteins/metabolism , Depsipeptides , Serum Response Factor/antagonists & inhibitors , Transcription Factors/metabolism , Base Sequence , Binding Sites , Cell Line , DNA Primers , Electrophoretic Mobility Shift Assay , Erythroid-Specific DNA-Binding Factors , Lim Kinases , Microfilament Proteins/genetics , Muscle Proteins/genetics , Peptides, Cyclic/pharmacology , Promoter Regions, Genetic , Protein Kinases/metabolism , Serum Response Factor/metabolism , YY1 Transcription Factor
10.
Am J Pathol ; 160(3): 1097-103, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11891206

ABSTRACT

Retinal neovascularization occurs in a variety of diseases including diabetic retinopathy, the most common cause of blindness in the developed world. There is accordingly considerable incentive to develop drugs that target the aberrant angiogenesis associated with these conditions. Previous studies have shown that a number of anti-angiogenic agents can inhibit retinal neovascularization in a well-characterized murine model of ischemia-induced proliferative retinopathy. Combretastatin-A4 (CA-4) is an anti-vascular tubulin-binding agent currently undergoing clinical evaluation for the treatment of solid tumors. We have recently shown that CA-4 is not tumor-specific but elicits anti-vascular effects in nonneoplastic angiogenic vessels. In this study we have examined the capacity of CA-4 to inhibit retinal neovascularization in vivo. CA-4 caused a dose-dependent inhibition of neovascularization with no apparent side effects. The absence of vascular abnormalities or remnants of disrupted neovessels in retinas of CA-4-treated mice suggests an anti-angiogenic mechanism in this model, in contrast to the anti-vascular effects observed against established tumor vessels. Importantly, histological and immunohistochemical analyses indicated that CA-4 permitted the development of normal retinal vasculature while inhibiting aberrant neovascularization. These data are consistent with CA-4 eliciting tissue-dependent anti-angiogenic effects and suggest that CA-4 has potential in the treatment of nonneoplastic diseases with an angiogenic component.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Neovascularization, Pathologic/drug therapy , Retinal Diseases/drug therapy , Stilbenes/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Mice , Mice, Inbred C57BL , Retinal Diseases/pathology , Stilbenes/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...