Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Oncogene ; 25(52): 6959-67, 2006 Nov 02.
Article in English | MEDLINE | ID: mdl-16715134

ABSTRACT

Understanding how RhoC expression and activation are regulated is essential for deciphering its contribution to tumorigenesis. Here, we report that RhoC expression and activation are induced by the epithelial to mesenchymal transition (EMT) of colon carcinoma. Using LIM 1863 colon cancer cells, RhoC protein expression and subsequent activation were detected coincident with the loss of E-cadherin and acquisition of mesenchymal characteristics. Several Ets-1 binding sites were identified in the RhoC promoter, and evidence was obtained using chromatin immunoprecipitation that Ets-1 can regulate RhoC expression during the EMT. Interestingly, a marked decrease in RhoA activation associated with the EMT was observed that corresponds to the increase in RhoC expression. Use of shRNA established that RhoA inhibits and RhoC promotes post-EMT cell migration, demonstrating functional significance for their coordinate regulation. To assess the importance of RhoC expression in colon cancer, immunohistochemistry was performed on 566 colorectal tumors with known clinical outcome. The level of RhoC ranged from no expression to high expression, and statistical analysis revealed that elevated RhoC expression correlates with poor outcome as well as aberrant expression and localization of E-cadherin. These data provide one mechanism for how RhoC expression is regulated in colon carcinoma and substantiate its utility as a prognostic marker.


Subject(s)
Biomarkers, Tumor/analysis , Colonic Neoplasms/enzymology , Neoplasm Invasiveness/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Cadherins/metabolism , Cell Line, Tumor , Colonic Neoplasms/pathology , Enzyme Activation/physiology , Epithelial Cells/enzymology , Humans , Immunohistochemistry , Immunoprecipitation , Prognosis , Promoter Regions, Genetic , Proto-Oncogene Protein c-ets-1/metabolism , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , rho GTP-Binding Proteins/genetics , rhoC GTP-Binding Protein
2.
Mol Cell Biol ; 14(7): 4443-54, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8007951

ABSTRACT

Induction of interleukin-6 (IL-6) gene expression is mediated by numerous agents involving all major signal transduction pathways. We have compared the effects of prostaglandins and their second messenger cyclic AMP (cAMP) with the effect of lipopolysaccharide (LPS) on IL-6 gene expression. We demonstrate that secretion of IL-6 is induced by cAMP in murine monocytic PU5-1.8 cells, even though to a lesser extent than by LPS. Nevertheless, cAMP and prostaglandins of the E series in the presence of theophylline induce transcription of the IL-6 promoter more strongly than LPS, suggesting distinctive effects of cAMP and LPS on posttranscriptional events. Mutations within four regulatory elements, namely, the multiple response element (MRE), AP-1, NF-IL6, and NF-kappa B sites, significantly reduce, but do not completely abrogate, inducibility by cAMP and prostaglandin E1, whereas alterations of four additional sites have no effects. LPS-induced promoter activity, however, is almost completely abolished by mutations in the NF-kappa B site, suggesting that a single regulatory element is crucial for inducibility by LPS. Stimulation by cAMP is correlated with the binding of inducible factors to the AP-1, NF-IL6, and NF-kappa B elements, whereas factors binding to the MRE are constitutively expressed. Recombinant cAMP response element-binding protein binds to the MRE, indicating a potential role for this factor in the cAMP response. Our results suggest that cAMP and prostaglandins act through multiple, partially redundant regulatory elements to induce IL-6 expression in monocytic cells. Nuclear events that overlap partially with the LPS response but also exhibit distinctive features are involved.


Subject(s)
Cyclic AMP/pharmacology , Gene Expression/physiology , Interleukin-6/biosynthesis , Interleukin-6/genetics , Lipopolysaccharides/pharmacology , Promoter Regions, Genetic , Prostaglandins E/pharmacology , Regulatory Sequences, Nucleic Acid , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Base Sequence , Bucladesine/pharmacology , Cell Line , Dinoprostone/pharmacology , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Humans , Mice , Misoprostol/pharmacology , Molecular Sequence Data , Monocytes , NF-kappa B/metabolism , Oligodeoxyribonucleotides , Oligonucleotide Probes , Promoter Regions, Genetic/drug effects , Rats , Sequence Homology, Nucleic Acid , Signal Transduction/drug effects , Theophylline/pharmacology
3.
Mol Cell Biol ; 14(5): 3292-309, 1994 May.
Article in English | MEDLINE | ID: mdl-7909357

ABSTRACT

The ets gene family encodes a group of proteins which function as transcription factors under physiological conditions and, if aberrantly expressed, can cause cellular transformation. We have recently identified two regulatory elements in the murine immunoglobulin heavy-chain (IgH) enhancer, pi and microB, which exhibit striking similarity to binding sites for ets-related proteins. To identify ets-related transcriptional regulators expressed in pre-B lymphocytes that may interact with either the pi or the microB site, we have used a PCR approach with degenerate oligonucleotides encoding conserved sequences in all members of the ets family. We have cloned the gene for a new ets-related transcription factor, ERP (ets-related protein), from the murine pre-B cell line BASC 6C2 and from mouse lung tissue. The ERP protein contains a region of high homology with the ETS DNA-binding domain common to all members of the ets transcription factor/oncoprotein family. Three additional smaller regions show homology to the ELK-1 and SAP-1 genes, a subgroup of the ets gene family that interacts with the serum response factor. Full-length ERP expresses only negligible DNA-binding activity by itself. Removal of the carboxy terminus enables ERP to interact with a variety of ets-binding sites including the E74 site, the IgH enhancer pi site, and the lck promoter ets site, suggesting a carboxy-terminal negative regulatory domain. At least three ERP-related transcripts are expressed in a variety of tissues. However, within the B-cell lineage, ERP is highly expressed primarily at early stages of B-lymphocyte development, and expression declines drastically upon B-cell maturation, correlating with the enhancer activity of the IgH pi site. These data suggest that ERP might play a role in B-cell development and in IgH gene regulation.


Subject(s)
B-Lymphocytes/metabolism , Oncogene Proteins , Transcription Factors/biosynthesis , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Binding, Competitive , Cell Line , Chromosome Mapping , Chromosomes, Human , Cloning, Molecular , Conserved Sequence , Cricetinae , DNA Primers , Gene Expression , Humans , Hybrid Cells , Mice , Molecular Sequence Data , Multigene Family , Oligonucleotide Probes/metabolism , Poly A/isolation & purification , Poly A/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins c-ets , RNA, Messenger/isolation & purification , RNA, Messenger/metabolism , Repetitive Sequences, Nucleic Acid , Sequence Homology, Amino Acid , Transcription Factors/genetics , Tumor Cells, Cultured
4.
Mol Cell Biol ; 16(9): 5091-106, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8756667

ABSTRACT

We have cloned the gene for a novel Ets-related transcription factor, new Ets-related factor (NERF), from human spleen, fetal liver, and brain. Comparison of the deduced amino acid sequence of NERF with those of other members of the Ets family reveals that the level of homology to ELF-1, which is involved in the regulation of several T- and B-cell-specific genes, is highest. Homologies are clustered in the putative DNA binding domain in the middle of the protein, a basic domain just upstream of this domain, and several shorter stretches of homology towards the amino terminus. The presence of two predominant NERF transcripts in various fetal and adult human tissues is due to at least three alternative splice products, NERF-1a, NERF-1b, and NERF-2, which differ in their amino termini and their expression in different tissues. Only NERF-2 and ELF-1, and not NERF-1a and NERF-1b, function as transcriptional activators of the lyn and blk gene promoters, although all isoforms of NERF bind with affinities similar to those of ELF-1 to a variety of Ets binding sites in, among others, the blk, lck, lyn, mb-1, and immunoglobulin H genes and are expressed at similar levels. Since NERF and ELF-1 are coexpressed in B and T cells, both might be involved in the regulation of the same genes.


Subject(s)
DNA-Binding Proteins/chemistry , Genes , Transcription Factors/chemistry , Transcription Factors/isolation & purification , Adult , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Cell Line, Transformed , Chlorocebus aethiops , Cloning, Molecular , Fetal Proteins/isolation & purification , Fibroblasts/metabolism , Gene Expression Regulation , HeLa Cells/metabolism , Humans , Mice , Molecular Sequence Data , Multigene Family , Nuclear Proteins , Organ Specificity , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Regulatory Sequences, Nucleic Acid , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/physiology , Transcriptional Activation , src-Family Kinases/genetics
5.
Mol Cell Biol ; 17(8): 4419-33, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9234700

ABSTRACT

We report here the isolation of a novel, highly tissue-restricted member of the ets transcription factor/oncogene family, ESE-1 (for epithelium-specific Ets), which has features distinct from those of any other ets-related factor. ESE-1 contains two putative DNA binding domains: an ETS domain, which is unique in that the 5' half shows relatively weak homology to known ets factors, and an A/T hook domain, found in HMG proteins and various other nuclear factors. In contrast to any known ets factors, ESE-1 is expressed exclusively in epithelial cells. ESE-1 expression is induced during terminal differentiation of the epidermis and in a primary human keratinocyte differentiation system. The keratinocyte terminal differentiation marker gene, SPRR2A, is a putative target for ESE-1, since SPRR2A expression during keratinocyte differentiation correlates with induction of ESE-1 expression, and ESE-1 binds with high affinity to and transactivates the ets binding site in the SPRR2A promoter. ESE-1 also binds to and transactivates the enhancer of the Endo A gene, a potential target for ESE-1 in simple epithelia. Due to the important role that other ets factors play in cellular differentiation, ESE-1 is expected to be a critical regulator of epithelial cell differentiation.


Subject(s)
Keratinocytes/chemistry , Trans-Activators/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Cells, Cultured , Cloning, Molecular , Cornified Envelope Proline-Rich Proteins , DNA-Binding Proteins/genetics , Epidermal Cells , Epithelial Cells , Epithelium/chemistry , Gene Expression Regulation, Developmental , High Mobility Group Proteins/genetics , Humans , Keratinocytes/cytology , Keratins/genetics , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Nuclear Proteins , Organ Specificity , Protein Precursors/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ets , RNA, Messenger/analysis , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Trans-Activators/chemistry , Transcription Factors/genetics
6.
Circ Res ; 89(5): 380-8, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11532898

ABSTRACT

Vascular development is a highly organized sequence of events that requires the correct spatial and temporal expression of specific sets of genes leading to the development of a primary vascular network. The first step in this process is the differentiation of pluripotent stem cells into endothelial cells. This is followed by endothelial proliferation, migration, and eventual formation of endothelial tubes. Maturation of these primitive tubes into fully developed blood vessels requires the recruitment of surrounding pericytes and their differentiation into vascular smooth muscle cells. Many of the events that occur during vasculogenesis are recapitulated during angiogenesis. Transcription factors have been shown to serve as master switches for regulating a number of developmental processes. Using a candidate gene approach, the genomic regulatory regions required to direct vascular-specific gene expression of several receptor tyrosine kinases that are critical for vasculogenesis have been characterized and some of the transcription factors that are involved in the regulation of these genes have recently been identified. Many of these factors are also involved in the regulation of hematopoiesis and may have overlapping functions in determining hematopoietic and endothelial differentiation. Targeted disruption of other transcription factors that were not previously thought to be involved in vascular development have also been recently shown to play a role in blood vessel development. The purpose of this review is to provide an update on the progress that has been made in our understanding of the transcriptional regulation of vascular development over the past few years.


Subject(s)
Cardiovascular System/embryology , Gene Expression Regulation, Developmental , Neovascularization, Physiologic/genetics , Animals , Blood Vessels/embryology , Endothelium, Vascular/embryology , Humans , Transcription Factors/genetics , Transcription, Genetic
7.
Circ Res ; 88(2): 237-44, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11157678

ABSTRACT

Vascular development requires the tightly coordinated expression of several growth factors and their receptors. Among these are the Tie1 and Tie2 receptors, which are almost exclusively endothelial cell-specific. The critical transcriptional regulators of vascular-specific gene expression remain largely unknown. The Ets factors are a family of evolutionarily conserved transcription factors that regulate genes involved in cellular growth and differentiation. We have recently shown that the Ets factor NERF is a strong transactivator of the Tie1 and Tie2 genes. To extend these studies, we have begun to identify the Ets factors that are expressed in developing blood vessels of the chicken chorioallantoic membrane (CAM), a highly vascular embryonic network. RNA was extracted from microdissected CAM blood vessels, and reverse transcriptase-polymerase chain reaction was performed using oligonucleotides encoding conserved amino acids within the Ets domain. One of the polymerase chain reaction fragments was subcloned and identified as the chicken homologue of the Ets factor ELF-1, cELF-1. ELF-1 is most closely related to the Ets factor NERF. In situ hybridization and immunohistochemistry demonstrate that cELF-1 is enriched in developing chicken blood vessels. cELF-1 is also a strong transactivator of the Tie1 and Tie2 genes and can bind to conserved Ets sites within the promoters of these genes. A complex of similar size forms when gel shifts are performed with cellular extracts derived from the CAM blood vessels, which is recognized by an antibody against cELF-1. In summary, ELF-1 belongs to a subset of Ets factors that regulate vascular-specific gene expression during blood vessel development.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Allantois/blood supply , Allantois/embryology , Allantois/metabolism , Animals , Blood Vessels/cytology , Blood Vessels/embryology , Blood Vessels/metabolism , Blotting, Northern , Cell Line , Chick Embryo , Chickens , Chorion/blood supply , Chorion/embryology , Chorion/metabolism , Cloning, Molecular , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Molecular Sequence Data , Nuclear Proteins , Organ Specificity , Promoter Regions, Genetic/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor, TIE-1 , Receptor, TIE-2 , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, TIE , Regulatory Sequences, Nucleic Acid/genetics , Sequence Alignment , Sequence Homology, Amino Acid
8.
J Am Coll Cardiol ; 25(6): 1354-61, 1995 May.
Article in English | MEDLINE | ID: mdl-7722133

ABSTRACT

OBJECTIVES: We sought to validate the safety of transesophageal echocardiographically guided early cardioversion in conjunction with short-term anticoagulation as a strategy for guiding early cardioversion in hospitalized patients with atrial fibrillation. BACKGROUND: Because atrial thrombi are poorly seen by conventional imaging techniques, several weeks of prophylactic anticoagulation is routinely prescribed before cardioversion. Transesophageal echocardiography is a superior test for identifying atrial thrombi; preliminary feasibility studies have supported its use to guide early cardioversion for patients in whom no thrombus is observed, but safety has not been validated in any large series. METHODS: All patients admitted to hospital with atrial fibrillation during a 4.5-year period were screened. The inclusion criterion was a clinical duration of atrial fibrillation > 2 days or of unknown duration. Patients received anticoagulation with heparin/warfarin and underwent conventional transthoracic echocardiography followed by transesophageal study. Patients in whom transesophageal echocardiography revealed no atrial thrombus underwent pharmacologic or electrical cardioversion followed by warfarin therapy for 1 month. Cardioversion was deferred in patients with evidence of atrial thrombi, and they received prolonged warfarin treatment. RESULTS: Two hundred thirty-three patients (86% of those eligible) agreed to participate, and 230 underwent transesophageal echocardiography. Transesophageal echocardiography identified 40 atrial thrombi (left atrium 34, right atrium 6) in 34 patients (15%). One hundred eighty-six (95%) of 196 patients without thrombi had successful cardioversion to sinus rhythm, all without prolonged anticoagulation, and none (0%, 95% confidence interval 0% to 1.6%) experienced a clinical thromboembolic event. Eighteen patients with atrial thrombi underwent uneventful cardioversion after prolonged anticoagulation. CONCLUSIONS: Compared with smaller series that have shown only feasibility, this large prospective and consecutive study of patients undergoing transesophageal echocardiographically facilitated early cardioversion in conjunction with short-term anticoagulation validates the safety of this strategy. This treatment algorithm has a safety profile similar to conventional therapy and minimizes both the period of anticoagulation and the overall duration of atrial fibrillation.


Subject(s)
Atrial Fibrillation/therapy , Echocardiography, Transesophageal , Electric Countershock , Heart Atria/diagnostic imaging , Heart Diseases/diagnostic imaging , Thromboembolism/diagnostic imaging , Adult , Aged , Aged, 80 and over , Analysis of Variance , Atrial Fibrillation/complications , Electric Countershock/methods , Female , Heart Diseases/etiology , Heart Diseases/prevention & control , Heparin/therapeutic use , Humans , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Risk Factors , Sensitivity and Specificity , Thromboembolism/etiology , Thromboembolism/prevention & control , Warfarin/therapeutic use
9.
Arch Intern Med ; 155(20): 2193-8, 1995 Nov 13.
Article in English | MEDLINE | ID: mdl-7487241

ABSTRACT

BACKGROUND: Thromboembolism related to atrial fibrillation (AF) is a major cause of morbidity and mortality. Patients with acute thromboembolism and AF are at high risk for early recurrent events. OBJECTIVE: To determine the prevalence of left atrial thrombi in patients who had acute thromboembolism and newly diagnosed AF. PATIENTS AND METHODS: Adult inpatients with AF were screened to identify those with acute (< 36 hours) systemic thromboembolism and newly recognized AF. Of 41 qualifying patients, 31 (76%) agreed to undergo transesophageal echocardiographic study, including 24 with acute neurologic events and seven with peripheral thromboembolism. A control population consisted of 88 adults with newly recognized AF without clinical thromboembolism. RESULTS: Transesophageal echocardiography identified left atrial thrombi in 13 (43%) of the 30 study patients who underwent transesophageal echocardiography compared with nine (10%) of 87 controls (P < .001). Spontaneous echo contrast was identified in 27 (87%) of the study population vs 42 (48%) of controls (P < .001). The prevalence of this marker of blood stasis did not differ between patients with left atrial thrombi without thromboembolism (P = .69). Duration of AF, prevalence of abnormal left ventricular function, left atrial size, and mitral regurgitation were similar in both groups. CONCLUSIONS: Left atrial thrombi were identified in more than 40% of patients with acute thromboembolism and newly recognized AF. These data suggest that a major source of recurrent thromboembolism in this group may be residual thrombus migration. Among patients with AF and atrial thrombi, clinical thromboembolism seems to occur randomly, or is related to an unidentified process.


Subject(s)
Atrial Fibrillation/complications , Heart Atria , Thromboembolism/complications , Thrombosis/complications , Acute Disease , Aged , Aged, 80 and over , Atrial Fibrillation/diagnostic imaging , Echocardiography, Transesophageal , Female , Heart Atria/diagnostic imaging , Heart Diseases/complications , Humans , Male , Middle Aged , Prevalence , Thromboembolism/diagnostic imaging , Thrombosis/diagnostic imaging
12.
J Biol Chem ; 271(42): 26007-12, 1996 Oct 18.
Article in English | MEDLINE | ID: mdl-8824239

ABSTRACT

We previously identified a B-cell-specific regulatory element in the immunoglobulin heavy chain (IgH) enhancer, pi, with striking similarity to binding sites for ets-related transcription factors. Whereas the ability of ets-related factors to bind to and transactivate the pi site has been substantiated, the identification of the particular member of the ets family responsible for B-cell-specific regulation of the pi site has remained controversial. We have used antibodies specific for individual members of the ets family to evaluate which ets-related factor in B-cell nuclear extracts interacts with the IgH pi site. We present strong evidence that ELF-1 is highly expressed in B-cells and is one of two major factors specifically interacting with the murine IgH enhancer pi site in B-cell nuclear extracts. Binding of ELF-1 correlates with activity of the pi site, since mutations abolishing function of pi also inhibit binding of ELF-1. Furthermore, we demonstrate that ELF-1 can transactivate the IgH enhancer in HeLa cells, suggesting a role for ELF-1 in B-cell-specific IgH gene expression.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Immunoglobulin Heavy Chains/genetics , Transcription Factors/metabolism , Transcriptional Activation , Animals , B-Lymphocytes/metabolism , Base Sequence , Cells, Cultured , Consensus Sequence , HeLa Cells , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins
13.
Circ Res ; 84(10): 1177-85, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10347092

ABSTRACT

The Tie2 gene encodes a vascular endothelium-specific receptor tyrosine kinase that is required for normal vascular development and is also upregulated during angiogenesis. The regulatory regions of the Tie2 gene that are required for endothelium-specific gene expression in vivo have been identified. However, the transcription factors required for Tie2 gene expression remain largely unknown. We have identified highly conserved binding sites for Ets transcription factors in the Tie2 promoter. Mutations in 2 particular binding sites lead to a 50% reduction in the endothelium-specific activity of the promoter. We have compared the ability of several members of the Ets family to transactivate the Tie2 promoter. Our results demonstrate that 1 of 3 distinct isoforms of the novel Ets transcription factor NERF, NERF2, is expressed in endothelial cells and can strongly transactivate the regulatory regions of the Tie2 gene in comparison to other Ets factors, which have little or no effect. NERF2 can bind to the Tie2 promoter Ets sites in electrophoretic mobility shift assays. These studies support a role for Ets factors in the regulation of vascular-specific gene expression and suggest that the novel Ets factor NERF2 may be a critical transcription factor in specifying the expression of the Tie2 gene in vascular endothelial cells.


Subject(s)
Endothelium, Vascular/enzymology , Gene Expression Regulation, Enzymologic , Receptor Protein-Tyrosine Kinases/genetics , Transcription Factors/metabolism , Base Sequence , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endothelium, Vascular/cytology , Genes, Reporter , Humans , Kidney/cytology , Luciferases/genetics , Molecular Sequence Data , Mutagenesis/physiology , Neovascularization, Physiologic/physiology , Point Mutation/physiology , Promoter Regions, Genetic/physiology , Receptor, TIE-2 , Transcription Factors/genetics , Transcriptional Activation/physiology , Umbilical Veins/cytology
14.
Proc Natl Acad Sci U S A ; 83(22): 8457-61, 1986 Nov.
Article in English | MEDLINE | ID: mdl-3095836

ABSTRACT

The genes coding for three proteins of the plasma lipid transport system--apolipoproteins A1 (APOA1), C3 (APOC3), and A4 (APOA4)--are closely linked and tandemly organized on the long arm of human chromosome 11. In this study the human APOA4 gene has been isolated and characterized. In contrast to APOA1 and APOC3 genes, which contain three introns, the APOA4 gene contains only two. An intron interrupting the 5' noncoding region of the APOA1 and APOC3 mRNAs is absent from the corresponding position of the APOA4 mRNA. However, similar to APOA1 and APOC3 genes, the introns of the APOA4 gene separate nucleotide sequences coding for the signal peptide and the amphipathic domains in APOA4. These results suggest that the APOA1, APOC3, and APOA4 genes were derived from a common evolutionary ancestor and indicate that during evolution the APOA4 gene lost one of its ancestral introns. Two restriction endonuclease sites, an Xba I located in the second intron of the APOA4 gene and a different Xba I located 9 kilobases 3' to the APOA4 gene, are polymorphic in Mediterranean and Northern European populations. Haplotype analysis indicated that even though these polymorphic sites are located within 9 kilobases they do not display significant nonrandom association. Finally, restriction mapping analysis of DNA from a patient with combined APOA1-APOC3 deficiency and premature coronary artery disease indicated that this patient has a structurally normal APOA4 gene.


Subject(s)
Apolipoproteins A/genetics , Polymorphism, Genetic , Apolipoprotein A-I , Apolipoprotein C-III , Apolipoproteins A/deficiency , Apolipoproteins C/deficiency , Apolipoproteins C/genetics , Base Sequence , Biological Evolution , Coronary Disease/genetics , DNA/analysis , Humans , RNA, Messenger/analysis , Recombination, Genetic
15.
Genomics ; 55(3): 358-62, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-10049594

ABSTRACT

We recently isolated a novel member of the Ets transcription factor/oncogene family, ESE-1/ESX/ELF3, with features distinct from any other Ets-related factor. ELF3 is the prototype of a new subclass of Ets factors, contains two DNA-binding domains, and, in contrast to any known Ets factor, is expressed exclusively in epithelial cells. ELF3 expression is induced during differentiation of the epidermis, indicating a role in the regulation of terminal differentiation genes in the epidermis. Due to the important role that other Ets factors play in cellular differentiation, ELF3 is expected to be a critical regulator of epithelial gene expression. We report here the cloning and the structural organization of the human ELF3 gene. The human ELF3 gene contains nine exons, which span approximately 5.8 kb of genomic DNA. Intron/exon borders and number of exons are almost identical to those in the mouse ELF3 gene. Comparison of the immediate promoter regions of the human and mouse ELF3 genes demonstrates the presence of TATA and CCAAT boxes as well as potential binding sites for Ets factors and NF-kappaB. Transfection experiments demonstrate that a 1.5-kb fragment of the 5' upstream region acts as a strong promoter in two epithelial cell lines.


Subject(s)
DNA-Binding Proteins , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Animals , Base Sequence , CHO Cells , Cricetinae , Epithelium/metabolism , Exons , Humans , Introns , Liver/metabolism , Luciferases/metabolism , Mice , Models, Genetic , Molecular Sequence Data , Oligonucleotide Probes , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ets , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Transcription, Genetic
16.
FASEB J ; 13(2): 377-86, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9973326

ABSTRACT

The Tie gene encodes an endothelial cell receptor tyrosine kinase necessary for normal vascular development. The Tie gene promoter targets expression of heterologous genes specifically to endothelial cells in transgenic mice. Here we have characterized the promoter sequences critical for endothelial cell-specific activity in cultured cells and transgenic mice. Progressive deletions and site-directed mutations of the promoter showed that the critical endothelial cell-specific elements are an octamer transcription factor binding site and several Ets binding sites located in two clusters within 300 bp upstream of the major transcription initiation site. Among members of the Ets transcription factor family tested, NERF-2 (a novel transcription factor related to the ets factor ELF-1), which is expressed in endothelial cells, and ETS2 showed the strongest transactivation of the Tie promoter; ETS1 gave lower levels of stimulation and the other Ets factors gave little or no transactivation. Furthermore, the Tie promoter directed the production of high amounts of human growth hormone into the circulation of transgenic mice. The secreted amounts correlated with transgene copy number, being relatively insensitive to the effects of the transgene integration site. These properties suggest that Tie promoter activity is controlled by endothelial cell Ets factors and that it has potential for use in vectors for endothelial cell-specific gene expression.


Subject(s)
Endothelium, Vascular/enzymology , Gene Expression Regulation, Enzymologic , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Transcription Factors/genetics , Animals , Base Sequence , Cell Line , Endothelium, Vascular/embryology , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, TIE , Transcription Factors/metabolism , Transfection
17.
Hum Genet ; 80(3): 265-73, 1988 Nov.
Article in English | MEDLINE | ID: mdl-2903847

ABSTRACT

The genes coding for apolipoproteins A1, C3, and A4 (APOA1, APOC3, APOA4) are closely linked and tandemly organized within a 15-kilobase (kb) DNA segment on the long arm of human chromosome 11. The nucleotide variability of a 61-kb DNA segment containing these genes and their flanking sequences was studied by restriction analysis of a sample of 18 unrelated Northern Europeans using seven different genomic DNA probes. Eleven restriction site polymorphisms located within this DNA segment were used for haplotype analysis of 129 Mediterranean and 67 American black chromosomes. Estimation of the extent of nonrandom association between these polymorphisms indicated considerable linkage disequilibrium within the APOA1-APOC3-APOA4 gene cluster. Several haplotypes arose by recombination, and the rate of recombination within this gene cluster was estimated to be at least 4 times greater than that expected based on uniform recombination. The polymorphism information content of each of these polymorphisms, taken individually, ranges between 0.053 and 0.375, while that of their haplotypes ranges between 0.858 and 0.862. Therefore, DNA polymorphism haplotypes in the APOA1-APOC3-APOA4 gene cluster constitute a highly informative genetic marker on the long arm of human chromosome 11.


Subject(s)
Apolipoproteins A/genetics , Apolipoproteins C/genetics , DNA/genetics , Haplotypes , Multigene Family , Polymorphism, Genetic , Polymorphism, Restriction Fragment Length , Apolipoprotein A-I , Apolipoprotein C-III , Chromosome Mapping , Genetic Linkage , Humans , Nucleic Acid Hybridization
18.
J Biol Chem ; 276(12): 9421-36, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11108721

ABSTRACT

We report here the isolation of Tel-2, a novel member of the Ets transcription factor family, with high homology to Tel/ETV-6. Tel-2 is the second mammalian member of the Tel Ets family subclass whose prototype Tel is involved in various chromosomal translocations in human cancers. Six differentially expressed alternative splice products of Tel-2 were characterized encoding different Tel-2 isoforms which either contain or lack the amino-terminal Pointed domain and also vary at the carboxyl terminus. In contrast to Tel, which is highly expressed in several different cell types and tissues, Tel-2 is only weakly expressed in a variety of tissues and cell types, including placenta, prostate, spleen, liver, and lung. Tel-2 binds to functionally relevant Ets-binding sites of several genes and only the Tel-2 isoform containing the Pointed domain and the DNA-binding domain acts as a strong repressor of transcription. The retinoic acid receptor alpha and bone morphogenetic protein-6B (BMP-6) genes are specifically repressed by Tel-2 indicating a function for Tel-2 as an inhibitor of differentiation. Due to the important involvement of Tel in human cancer and the location of Tel-2 within the MHC cluster region, Tel-2 might be involved in chromosomal translocations in human cancer as well.


Subject(s)
DNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Bone Morphogenetic Protein 6 , Bone Morphogenetic Proteins/genetics , Cell Line , Cloning, Molecular , DNA, Complementary , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Humans , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-ets , Receptors, Retinoic Acid/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Retinoic Acid Receptor alpha , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Transcription Factors/chemistry , Transcription Factors/genetics
19.
Proc Natl Acad Sci U S A ; 95(3): 1213-7, 1998 Feb 03.
Article in English | MEDLINE | ID: mdl-9448311

ABSTRACT

Proliferation of vascular smooth muscle cells (VSMCs) in response to injury plays a key role in the pathogenesis of vascular disorders. Fas ligand (FasL) induces apoptosis in Fas-bearing cells, and its expression on activated T cells contributes to the regulation of the immune response and physiological cell turnover. Here, we show that a replication-defective adenovirus encoding FasL (Ad-FasL) induced apoptosis in Fas-bearing VSMCs. When introduced locally to balloon-injured rat carotid arteries, a well characterized model of a VSMC-derived lesion, Ad-FasL functioned as a potent inhibitor of neointima formation. In rats immunized with an empty adenoviral vector, robust T cell infiltration of the vessel wall was detected after local delivery of a beta-galactosidase-expressing virus (Ad-betagal), whereas T cell infiltrates were not detected after local delivery of Ad-FasL. Prior immunization prevented beta-galactosidase expression from Ad-betagal, whereas the expression of the FasL transgene was unaffected. When Ad-betagal and Ad-FasL were delivered together to preimmunized animals, T cell infiltration was reduced and beta-galactosidase expression was restored. These data demonstrate that Fas ligand gene transfer can effectively inhibit injury-induced vessel lesion formation and can allow adenovirus-harboring cells to evade immune destruction.


Subject(s)
Adenoviridae Infections/pathology , Gene Transfer Techniques , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Muscle, Smooth, Vascular/pathology , T-Lymphocytes/immunology , T-Lymphocytes/virology , fas Receptor/genetics , fas Receptor/immunology , Angioplasty, Balloon/adverse effects , Animals , Apoptosis , DNA Fragmentation , Endothelium, Vascular/injuries , Endothelium, Vascular/pathology , Fas Ligand Protein , Humans , Jurkat Cells , Ligands , Male , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/injuries , Rats , Rats, Sprague-Dawley
20.
Ann Rheum Dis ; 62(2): 100-7, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12525377

ABSTRACT

OBJECTIVES: To examine the potential role of the angiogenic growth factor angiopoietin-1 (Ang-1) in inflammatory arthritis. METHODS: Eighteen synovial tissue samples were obtained from 17 patients with a clinical diagnosis of rheumatoid arthritis (RA) and compared with six synovial tissue samples from six patients with osteoarthritis (OA). Ang-1 expression in synovial tissues was determined by immunohistochemistry and in situ hybridisation. Ang-1 mRNA and protein expression were also examined by northern blot analysis and enzyme linked immunosorbent assay (ELISA) in cultured synovial fibroblasts and human umbilical vein endothelial cells (HUVECs) before and after treatment with tumour necrosis factor (TNF)alpha. RESULTS: Ang-1 protein expression was detected by immunohistochemistry in 16/18 RA synovial tissue samples. Ang-1 protein was frequently observed in the synovial lining layer and in cells within the sublining synovial tissue, in both perivascular areas and in areas remote from vessels. In contrast, Ang-1 was only weakly detected in these sites in OA samples. Ang-1 mRNA and protein were also expressed in cultured synovial fibroblasts derived from patients with RA. In addition, induction of Ang-1 mRNA and protein was observed by northern blot analysis and ELISA after stimulation of RA synovial fibroblasts, but not HUVECs, with the proinflammatory cytokine TNF alpha. CONCLUSIONS: Ang-1 mRNA and protein are expressed in the synovium of patients with RA, and are up regulated in synovial fibroblasts by TNF alpha. Ang-1 may therefore be an important regulator of angiogenesis in inflammatory arthritis.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Arthritis, Rheumatoid/metabolism , Membrane Glycoproteins/metabolism , Synovial Membrane/metabolism , Angiogenesis Inducing Agents/genetics , Angiopoietin-1 , Angiopoietin-2 , Arthritis, Rheumatoid/pathology , Blotting, Northern/methods , Culture Techniques , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Immunoenzyme Techniques , In Situ Hybridization , Interleukin-1/pharmacology , Membrane Glycoproteins/genetics , Osteoarthritis/metabolism , RNA, Messenger/genetics , Synovial Membrane/blood supply , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL