Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Cancer Sci ; 113(12): 4350-4362, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36121618

ABSTRACT

Antibody-mimetic drug conjugate is a novel noncovalent conjugate consisting of an antibody-mimetic recognizing a target molecule on the cancer cell surface and low-molecular-weight payloads that kill the cancer cells. In this study, the efficacy of a photo-activating antibody-mimetic drug conjugate targeting HER2-expressing tumors was evaluated in mice, by using the affibody that recognize HER2 (ZHER2:342 ) as a target molecule and an axially substituted silicon phthalocyanine (a novel potent photo-activating compound) as a payload. The first treatment with the photo-activating antibody-mimetic drug conjugates reduced the size of all HER2-expressing KPL-4 xenograft tumors macroscopically. However, during the observation period, relapsed tumors gradually appeared in approximately 50% of the animals. To evaluate the efficacy of repeated antibody-mimetic drug conjugate treatment, animals with relapsed tumors were treated again with the same regimen. After the second observation period, the mouse tissues were examined histopathologically. Unexpectedly, all relapsed tumors were eradicated, and all animals were diagnosed with pathological complete remission. After the second treatment, skin wounds healed rapidly, and no significant side effects were observed in other organs, except for occasional microscopic granulomatous tissues beneath the serosa of the liver in a few mice. Repeated treatments seemed to be well tolerated. These results indicate the promising efficacy of the repeated photo-activating antibody-mimetic drug conjugate treatment against HER2-expressing tumors.


Subject(s)
Immunoconjugates , Humans , Animals , Mice , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Receptor, ErbB-2/metabolism , Cell Line, Tumor , Antibodies
2.
Protein Expr Purif ; 192: 106043, 2022 04.
Article in English | MEDLINE | ID: mdl-34973460

ABSTRACT

Antibody-drug conjugates (ADCs) are a major therapeutic tool for the treatment of advanced cancer. Malignant cells in advanced cancer often display multiple genetic mutations and become resistant to monotherapy. Therefore, a therapeutic regimen that simultaneously targets multiple molecules with multiple payloads is desirable. However, the development of ADCs is hampered by issues in biopharmaceutical manufacturing and the complexity of the conjugation process of low-molecular-weight payloads to biologicals. Here, we report antibody mimetic-drug conjugates (AMDCs) developed by exploiting the non-covalent binding property of payloads based on high-affinity binding of mutated streptavidin and modified iminobiotin. Miniprotein antibodies were fused to a low immunogenic streptavidin variant, which was then expressed in Escherichia coli inclusion bodies, solubilized, and refolded into functional tetramers. The AMDC developed against human epidermal growth factor receptor 2 (HER2) effectively killed cultured cancer cells using bis-iminobiotin conjugated to photo-activating silicon phthalocyanine. The HER2-targeting AMDC was also effective in vivo against a mouse KPL-4 xenograft model. This AMDC platform provides rapid, stable, and high-yield therapeutics against multiple targets.


Subject(s)
Escherichia coli/metabolism , Gene Expression , Immunoconjugates/genetics , Animals , Biotin/administration & dosage , Biotin/analogs & derivatives , Biotin/chemistry , Biotin/genetics , Biotin/immunology , Cell Line, Tumor , Cloning, Molecular , Escherichia coli/genetics , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Immunoconjugates/immunology , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Protein Folding , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Streptavidin/administration & dosage , Streptavidin/chemistry , Streptavidin/genetics , Streptavidin/immunology
3.
Medicine (Baltimore) ; 100(15): e25526, 2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33847675

ABSTRACT

RATIONALE: In some cases, autopsy is the first opportunity to find a previously unrecognized critical infection. Pathogens are identified by various methods, such as microscopic examination, special stains, culture tests, and immunohistochemistry. Here, we report a case of 16S ribosomal RNA (rRNA) gene sequencing using a postmortem formalin-fixed, paraffin-embedded (FFPE) tissue, which was useful for identifying pathogenic microbes. PATIENT CONCERNS: Autopsy was performed on an 87-year-old man who had chronic renal failure and had developed sepsis from a central venous catheter infection 10 days before his death. Prior to these events, von Meyenburg complexes (VMCs) were also found during regular checkups. DIAGNOSIS: Postmortem microscopic examination revealed acute purulent cholangitis with numerous microabscesses, accompanied by VMCs. Gram-negative rods were observed in some microabscesses, which were considered causative pathogens. INTERVENTIONS: 16S rRNA gene sequencing using postmortem FFPE tissue. OUTCOMES: Pseudomonas aeruginosa was identified, different from the one detected in the central venous catheter culture while alive. LESSONS: 16S rRNA gene sequencing is a useful tool for identifying pathogenic microbes in postmortem FFPE tissues. This technique may be useful for amplicon sizes of approximately 100 bp or less.


Subject(s)
Biliary Tract Diseases/microbiology , Cholangitis/microbiology , Hamartoma/microbiology , Pseudomonas aeruginosa , RNA, Bacterial/analysis , Acute Disease , Aged, 80 and over , Autopsy , Fatal Outcome , Formaldehyde , Humans , Male , Medical Illustration , Paraffin Embedding , RNA, Ribosomal, 16S/analysis , Sequence Analysis, RNA
4.
EMBO J ; 39(7): e103949, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32125007

ABSTRACT

Histone H3 lysine-9 di-methylation (H3K9me2) and lysine-27 tri-methylation (H3K27me3) are linked to repression of gene expression, but the functions of repressive histone methylation dynamics during inflammatory responses remain enigmatic. Here, we report that lysine demethylases 7A (KDM7A) and 6A (UTX) play crucial roles in tumor necrosis factor (TNF)-α signaling in endothelial cells (ECs), where they are regulated by a novel TNF-α-responsive microRNA, miR-3679-5p. TNF-α rapidly induces co-occupancy of KDM7A and UTX at nuclear factor kappa-B (NF-κB)-associated elements in human ECs. KDM7A and UTX demethylate H3K9me2 and H3K27me3, respectively, and are both required for activation of NF-κB-dependent inflammatory genes. Chromosome conformation capture-based methods furthermore uncover increased interactions between TNF-α-induced super enhancers at NF-κB-relevant loci, coinciding with KDM7A and UTX recruitments. Simultaneous pharmacological inhibition of KDM7A and UTX significantly reduces leukocyte adhesion in mice, establishing the biological and potential translational relevance of this mechanism. Collectively, these findings suggest that rapid erasure of repressive histone marks by KDM7A and UTX is essential for NF-κB-dependent regulation of genes that control inflammatory responses of ECs.


Subject(s)
Endothelial Cells/immunology , Histone Demethylases/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , MicroRNAs/genetics , Animals , Cell Adhesion , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Histones/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Lysine/metabolism , Male , Methylation , Mice , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
5.
J Evol Biol ; 32(10): 1117-1123, 2019 10.
Article in English | MEDLINE | ID: mdl-31381200

ABSTRACT

Large genetic variations in starvation tolerance in animals indicate that there are multiple strategies to cope with low-nutrient conditions. Fruit flies (Drosophila melanogaster) typically respond to starvation by suppressing sleep and enhancing locomotor activity presumably to search for food. However, we hypothesized that in a natural population, there are costs and benefits to sleep suppression under low-nutrient conditions and that conserving energy through sleep could be a better strategy depending on food availability. In this study, we quantified the variation in sleep-related traits in 21 wild-derived inbred lines from Katsunuma, Japan, under fed and starved conditions and analysed the relationship between those traits and starvation tolerance. Although most of the lines responded to starvation by suppressing the total time in sleep, there were indeed two lines that responded by significantly increasing the sleep-bout durations and thus not reducing the total time in sleep. These genotypes survived longer in acute starvation conditions compared to genotypes that responded by the immediate suppression of sleep, which could be due to the reduced metabolic rate during the long uninterrupted sleep bouts. The coexistence of the enhanced foraging and resting strategies upon starvation within a single population is consistent with the presence of a behavioural trade-off between food search and energy conservation due to unpredictable food availability in nature. These results provide insights into the evolutionary mechanisms that contribute to the maintenance of genetic variations underlying environmental stress resistance.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Sleep/physiology , Starvation , Animals , Female , Humans , Motor Activity
6.
PLoS One ; 10(7): e0131793, 2015.
Article in English | MEDLINE | ID: mdl-26181717

ABSTRACT

Of late, numerous prodrugs are widely used for therapy. The hemeprotein cytochrome P450 (CYP) catalyzes the activation of prodrugs to form active metabolites. Therefore, the activation of CYP function might allow the use of lower doses of prodrugs and decrease toxicity. We hypothesized that the addition of 5-aminolevulinic acid (ALA), a precursor in the porphyrin biosynthetic pathway, enhances the synthesis of heme, leading to the up-regulation of CYP activity. To test this hypothesis, we treated a human gastric cancer cell line with ALA and determined the effect on CYP-dependent prodrug activation. For this purpose, we focused on the anticancer prodrug tegafur, which is converted to its active metabolite 5-fluorouracil (5-FU) mainly by CYP2A6. We show here that ALA increased CYP2A6-dependent tegafur activation, suggesting that ALA elevated CYP activity and potentiated the activation of the prodrug.


Subject(s)
Aminolevulinic Acid/pharmacology , Prodrugs/metabolism , Tegafur/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Cell Line, Tumor , Cholic Acid/pharmacology , Cytochrome P-450 CYP2A6/genetics , Cytochrome P-450 CYP2A6/metabolism , Heme/metabolism , Humans , Neoplasm Proteins/metabolism
7.
J Biol Chem ; 289(42): 29044-59, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25157100

ABSTRACT

VEGF is a key regulator of endothelial cell migration, proliferation, and inflammation, which leads to activation of several signaling cascades, including the calcineurin-nuclear factor of activated T cells (NFAT) pathway. NFAT is not only important for immune responses but also for cardiovascular development and the pathogenesis of Down syndrome. By using Down syndrome model mice and clinical patient samples, we showed recently that the VEGF-calcineurin-NFAT signaling axis regulates tumor angiogenesis and tumor metastasis. However, the connection between genome-wide views of NFAT-mediated gene regulation and downstream gene function in the endothelium has not been studied extensively. Here we performed comprehensive mapping of genome-wide NFATc1 binding in VEGF-stimulated primary cultured endothelial cells and elucidated the functional consequences of VEGF-NFATc1-mediated phenotypic changes. A comparison of the NFATc1 ChIP sequence profile and epigenetic histone marks revealed that predominant NFATc1-occupied peaks overlapped with promoter-associated histone marks. Moreover, we identified two novel NFATc1 regulated genes, CXCR7 and RND1. CXCR7 knockdown abrogated SDF-1- and VEGF-mediated cell migration and tube formation. siRNA treatment of RND1 impaired vascular barrier function, caused RhoA hyperactivation, and further stimulated VEGF-mediated vascular outgrowth from aortic rings. Taken together, these findings suggest that dynamic NFATc1 binding to target genes is critical for VEGF-mediated endothelial cell activation. CXCR7 and RND1 are NFATc1 target genes with multiple functions, including regulation of cell migration, tube formation, and barrier formation in endothelial cells.


Subject(s)
Endothelium, Vascular/metabolism , NFATC Transcription Factors/metabolism , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/metabolism , Animals , COS Cells , Cell Movement , Chlorocebus aethiops , Coculture Techniques , Endothelial Cells/cytology , Epigenesis, Genetic , Fibroblasts/metabolism , Genome-Wide Association Study , HEK293 Cells , Homeostasis , Human Umbilical Vein Endothelial Cells , Humans , Mice , Oligonucleotide Array Sequence Analysis , Phenotype , Receptors, CXCR4/metabolism , Signal Transduction , Transcriptional Activation , rho GTP-Binding Proteins/metabolism
8.
Cell Rep ; 4(4): 709-23, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23954784

ABSTRACT

The premetastatic niche is a predetermined site of metastases, awaiting the influx of tumor cells. However, the regulation of the angiogenic switch at these sites has not been examined. Here, we demonstrate that the calcineurin and nuclear factor of activated T cells (NFAT) pathway is activated specifically in lung endothelium prior to the detection of tumor cells that preferentially metastasize to the lung. Upregulation of the calcineurin pathway via deletion of its endogenous inhibitor Dscr1 leads to a significant increase in lung metastases due to increased expression of a newly identified NFAT target, Angiopoietin-2 (ANG2). Increased VEGF levels specifically in the lung, and not other organ microenvironments, trigger a threshold of calcineurin-NFAT signaling that transactivates Ang2 in lung endothelium. Further, we demonstrate that overexpression of DSCR1 or the ANG2 receptor, soluble TIE2, prevents the activation of lung endothelium, inhibiting lung metastases in our mouse models. Our studies provide insights into mechanisms underlying angiogenesis in the premetastatic niche and offer targets for lung metastases.


Subject(s)
Calcineurin/metabolism , Lung Neoplasms/metabolism , Lung/metabolism , NFATC Transcription Factors/metabolism , Ribonuclease, Pancreatic/metabolism , Animals , Calcium-Binding Proteins , Cell Line, Tumor , Cells, Cultured , Endothelium/metabolism , Endothelium/pathology , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung/pathology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscle Proteins/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Ribonuclease, Pancreatic/genetics , Signal Transduction , Transcriptional Activation , Tumor Microenvironment
9.
Biosystems ; 101(3): 162-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20599583

ABSTRACT

In the colour contrast effect, the impression of a colour changes according to the situation; cases occur in which the colour appearance is misunderstood. We propose an image signal processing method for preventing such misperception of colour. Many conventional image improving methods emphasize the contrast of images as same as the brain does. However, by their processes, the colour contrast effect is not canceled; we misunderstand the colour. The objective of this study is to perceive original colour. Therefore, we propose an image correction method using inverse processes of the brain in order to cancel the processes of the brain, the colour contrast effect. We verified whether the proposed method corrected the colour contrast effect by conducting a psychological experiment. The results show that the method succeeds in canceling the colour contrast effect.


Subject(s)
Biomimetic Materials , Color , Contrast Sensitivity/physiology , Image Enhancement/methods , Models, Theoretical , Visual Perception/physiology , Humans , Psychological Tests
10.
J Clin Invest ; 119(8): 2257-70, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19620774

ABSTRACT

Down syndrome critical region gene 1 (DSCR-1) short variant (DSCR-1s) is an inhibitor of calcineurin/NFAT signaling encoded by exons 4-7 of DSCR1. We previously reported that VEGF induces DSCR-1s expression in endothelial cells, which in turn negatively feeds back to attenuate endothelial cell activation. Here, in order to characterize the role of the promoter that drives DSCR-1s expression in mediating inducible expression in vivo and to determine the functional relevance of DSCR-1s in inflammation, we targeted a DNA construct containing 1.7 kb of the human DSCR1s promoter coupled to the lacZ reporter to the hypoxanthine guanine phosphoribosyl transferase (Hprt) locus of mice. We determined that lacZ was uniformly expressed in the endothelium of transgenic embryos but was markedly downregulated postnatally. Systemic administration of VEGF or LPS in adult mice resulted in cyclosporine A-sensitive reactivation of the DSCR1s promoter and endogenous gene expression in a subset of organs, including the heart and brain. The DSCR1s promoter was similarly induced in the endothelium of tumor xenografts. In a mouse model of endotoxemia, DSCR-1s-deficient mice demonstrated increased sepsis mortality, whereas adenovirus-mediated DSCR-1s overexpression protected against LPS-induced lethality. Collectively, these data suggest that the DSCR1s promoter directs vascular bed-specific expression in activated endothelium and that DSCR-1s serves to dampen the host response to infection.


Subject(s)
Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Muscle Proteins/genetics , Age Factors , Animals , Calcium-Binding Proteins , Cells, Cultured , Endothelial Cells/metabolism , Female , GATA2 Transcription Factor/physiology , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/physiology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/physiology , NFATC Transcription Factors/physiology , Promoter Regions, Genetic , Vascular Endothelial Growth Factor A/pharmacology
11.
Arterioscler Thromb Vasc Biol ; 26(12): 2652-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17008592

ABSTRACT

OBJECTIVE: Tumor necrosis factor (TNF)-alpha initiates numerous changes in endothelial cell (EC) gene expression that contributes to the pathology of various diseases including inflammation. We hypothesized that TNF-alpha-mediated gene induction involves multiple signaling pathways, and that inhibition of one or more of these pathways may selectively target subsets of TNF-alpha-responsive genes and functions. METHODS AND RESULTS: Human umbilical vein endothelial cells (ECs) were preincubated with inhibitors of PI3 kinase (LY294002), histone deacetylases (HDAC) (trichostatin A [TSA]), de novo protein synthesis (CHX), proteasome (MG-132), and GATA factors (K-11430) before exposure to TNF-alpha at 4 hours and analyzed by microarray. TNF-alpha-mediated induction of vascular cell adhesion molecule-1 (VCAM-1) was attenuated by all of these inhibitors, whereas in contrast, stimulation of intercellular adhesion molecule-1 (ICAM-1) was blocked by MG-132 alone. Moreover TSA blocked TNF-alpha-mediated induction of monocyte adhesion both in vitro and in vivo through the suppression of VCAM-1. Further analysis demonstrated that HDAC3 plays a significant role in the regulation of TNF-alpha-mediated VCAM-1 expression. CONCLUSIONS: TNF-alpha activates ECs via multiple signaling pathways, and these pathways may be selectively targeted to modulate EC function. Moreover, TSA treatment reduced monocyte adhesion via VCAM-1 suppression in vitro and in vivo, suggesting that TSA might be useful for the attenuation of the inflammatory response in EC.


Subject(s)
Endothelium, Vascular/metabolism , Histone Deacetylase Inhibitors , Monocytes/cytology , Monocytes/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Azepines/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Chromones/pharmacology , E-Selectin/genetics , E-Selectin/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/physiology , Humans , Hydroxamic Acids/pharmacology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Leupeptins/pharmacology , Mice , Mice, Transgenic , Monocytes/drug effects , Morpholines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Transcriptional Activation , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/genetics
12.
J Biol Chem ; 281(29): 20503-20, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16627481

ABSTRACT

Activation and dysfunction of the endothelium underlie many vascular disorders including atherosclerosis, tumor growth, and inflammation. We recently reported that thrombin and vascular endothelial growth factor, but not tumor necrosis factor-alpha, results in dramatic up-regulation of Down syndrome critical region (DSCR)-1 gene in endothelial cells, a negative feedback regulator of calcineurin-NFAT signaling. Constitutive expression of DSCR-1 in activated endothelial cells markedly impaired NFAT nuclear localization, proliferation, tube formation, and tumor growth. The goal of the present study was to elucidate the relative roles of NFAT/DSCR-1 and NF-kappaB/I-kappaB in mediating thrombin-responsive gene expression in endothelial cells. DNA microarrays of thrombin-treated human umbilical vein endothelial cells overexpressing DSCR-1 or constitutive active IkappaBalpha revealed genes that were dependent on NFAT and/or NF-kappaB activity. Vascular cell adhesion molecule-1 was inhibited both by DSCR-1 and I-kappaB at the level of mRNA, protein, promoter activity, and function (monocyte adhesion). Using a combination of transient transfections, electrophoretic mobility shift assays, and chromatin immunoprecipitation, thrombin was shown to induce time-dependent coordinate binding of RelA and NFATc to a tandem NF-kappaB element in the upstream promoter region of vascular cell adhesion molecule-1. Together, these findings suggest that thrombin-mediated activation of endothelial cells involves an interplay between NFAT and NF-kappaB signaling pathways and their negative feedback inhibitors, DSCR-1 and I-kappaB, respectively. As natural brakes in the inflammatory process, DSCR-1 and I-kappaB may lend themselves to therapeutic manipulation in vasculopathic disease states.


Subject(s)
Endothelium, Vascular/physiopathology , Inflammation/physiopathology , Intracellular Signaling Peptides and Proteins/genetics , Muscle Proteins/genetics , NFATC Transcription Factors/physiology , Thrombin/physiology , Vascular Cell Adhesion Molecule-1/genetics , Cell Adhesion/physiology , Cell Line , Cells, Cultured , DNA/genetics , DNA-Binding Proteins , Down Syndrome/genetics , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/physiology , Monocytes/physiology , NF-KappaB Inhibitor alpha , Oligonucleotide Array Sequence Analysis , Plasmids , RNA/genetics , RNA, Messenger/genetics , Transcription Factor RelA/metabolism , Umbilical Veins
13.
J Biol Chem ; 279(48): 50537-54, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15448146

ABSTRACT

Activation and dysfunction of the endothelium underlie many vascular disorders including atherosclerosis, tumor growth, and inflammation. Endothelial cell activation is mediated by many different extra-cellular signals, which result in overlapping yet distinct patterns of gene expression. Here we show, in DNA microarray analyses, that vascular endothelial growth factor (VEGF) and thrombin result in dramatic and rapid upregulation of Down syndrome critical region (DSCR)-1 gene encoding exons 4-7, a negative feedback regulator of calcium-calcineurin-NF-AT signaling. VEGF- and thrombin-mediated induction of DSCR-1 involves the cooperative binding of NF-ATc and GATA-2/3 to neighboring consensus motifs in the upstream promoter. Constitutive expression of DSCR-1 in endothelial cells markedly impaired NF-ATc nuclear localization, proliferation, and tube formation. Under in vivo conditions, overexpression of DSCR-1 reduced vascular density in matrigel plugs and melanoma tumor growth in mice. Taken together, these findings support a model in which VEGF- and thrombin-mediated induction of endothelial cell proliferation triggers a negative feedback loop consisting of DSCR-1 gene induction and secondary inhibition of NF-AT signaling. As a natural brake in the angiogenic process, this negative pathway may lend itself to therapeutic manipulation in pathological states.


Subject(s)
Muscle Proteins/metabolism , Neovascularization, Pathologic/metabolism , Thrombin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Division/physiology , Chromosomes, Human, Pair 21 , DNA-Binding Proteins , Down Syndrome/metabolism , Endothelial Cells/metabolism , Exons , Humans , Intracellular Signaling Peptides and Proteins , Mice , Muscle Proteins/genetics , Promoter Regions, Genetic , Protein Isoforms , RNA, Messenger/metabolism
14.
J Biol Chem ; 279(20): 20626-35, 2004 May 14.
Article in English | MEDLINE | ID: mdl-15016828

ABSTRACT

Recent evidence supports a role for GATA transcription factors as important signal intermediates in differentiated endothelial cells. The goal of this study was to identify proteins that interact with endothelial-derived GATA transcription factors. Using yeast two-hybrid screening, we identified hematopoietically expressed homeobox (Hex) as a GATA-binding partner in endothelial cells. The physical association between Hex and GATA was confirmed with immunoprecipitation in cultured cells. Hex overexpression resulted in decreased flk-1/KDR expression, both at the level of the promoter and the endogenous gene, and attenuated vascular endothelial growth factor-mediated tube formation in primary endothelial cell cultures. In electrophoretic mobility shift assays, Hex inhibited the binding of GATA-2 to the flk-1/KDR 5'-untranslated region GATA motif. Finally, in RNase protection assays, transforming growth factor beta1, which has been previously shown to decrease flk-1 expression by interfering with GATA binding activity, was shown to increase Hex expression in endothelial cells. Taken together, the present study provides evidence for a novel association between Hex and GATA and suggests that transforming growth factor beta-mediated repression of flk-1/KDR and vascular endothelial growth factor signaling involves the inducible formation of inhibitory Hex-GATA complexes.


Subject(s)
DNA-Binding Proteins/metabolism , Endothelium, Vascular/physiology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/physiology , Cell Line , Endothelium, Vascular/drug effects , GATA4 Transcription Factor , Humans , Signal Transduction/drug effects , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL