22 results on '"Hellwig-Bürgel, T"'
Search Results
2. Interleukin-1β inhibits the hypoxic inducibility of the erythropoietin enhancer by suppressing hepatocyte nuclear factor-4α
- Author
-
Krajewski, J., Batmunkh, C., Jelkmann, W., and Hellwig-Bürgel, T.
- Published
- 2007
- Full Text
- View/download PDF
3. Interleukin-1beta and tumor necrosis factor-alpha stimulate DNA binding of hypoxia-inducible factor-1
- Author
-
Hellwig-Bürgel T, Rutkowski K, Metzen E, Joachim Fandrey, and Jelkmann W
- Subjects
Carcinoma, Hepatocellular ,Liver Neoplasms ,Nuclear Proteins ,DNA ,Hypoxia-Inducible Factor 1, alpha Subunit ,DNA-Binding Proteins ,Mice ,Liver Neoplasms, Experimental ,Gene Expression Regulation ,Tumor Cells, Cultured ,Animals ,Humans ,Hypoxia-Inducible Factor 1 ,Hypoxia ,Interleukin-1 ,Protein Binding ,Transcription Factors - Abstract
The rate of transcription of several genes encoding proteins involved in O(2) and energy homeostasis is controlled by hypoxia-inducible factor-1 (HIF-1), a heterodimeric DNA binding complex composed of alpha and beta subunits. HIF-1 is considered the primary trans-acting factor for the erythropoietin (EPO) and vascular endothelial growth factor (VEGF) genes. Since EPO gene expression is inhibited by the proinflammatory cytokines interleukin-1beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha), while no such effect has been reported with respect to the VEGF gene, we investigated the effects of IL-1beta and TNF-alpha on the activation of the HIF-1 DNA-binding complex and the amount of HIF-1alpha protein in human hepatoma cells in culture. Under normoxic conditions, both cytokines caused a moderate activation of HIF-1 DNA binding. In hypoxia, cytokines strongly increased HIF-1 activity compared with the effect of hypoxia alone. Only IL-1beta increased HIF-1alpha protein levels. In transient transfection experiments, HIF-1-driven reporter gene expression was augmented by cytokines only under hypoxic conditions. In contrast to their effect on EPO synthesis, neither IL-1beta nor TNF-alpha decreased VEGF production. The mRNA levels of HIF-1alpha and VEGF were unaffected. Thus, cytokine-induced inhibition of EPO production is not mediated by impairment of HIF-1 function. We propose that HIF-1 may be involved in modulating gene expression during inflammation.
- Published
- 1999
4. Dexamethasone impairs hypoxia-inducible factor-1 function
- Author
-
Wagner, A.E., primary, Huck, G., additional, Stiehl, D.P., additional, Jelkmann, W., additional, and Hellwig-Bürgel, T., additional
- Published
- 2008
- Full Text
- View/download PDF
5. Infektionen mit respiratorischen Bakterien verändern die Anpassung von Lungengewebszellen an Hypoxie
- Author
-
Hellwig-Bürgel, T, primary, Rupp, J, additional, Deiwick, J, additional, Solbach, W, additional, and Dalhoff, K, additional
- Published
- 2008
- Full Text
- View/download PDF
6. Hypoxia and interleukin-1beta stimulate vascular endothelial growth factor production in human proximal tubular cells.
- Author
-
El Awad, Baha, Kreft, Burkhard, Wolber, Eva-Maria, Hellwig-Burgel, Thomas, Metzen, Eric, Fandrey, Joachim, Jelkmann, Wolfgang, El Awad, B, Kreft, B, Wolber, E M, Hellwig-Bürgel, T, Metzen, E, Fandrey, J, and Jelkmann, W
- Subjects
- *
HYPOXEMIA , *INTERLEUKIN-1 , *INTERLEUKINS , *PHYSIOLOGY - Abstract
Background: Vascular endothelial growth factor (VEGF) promotes angiogenesis and inflammatory reactions. VEGF mRNA is detectable in the proximal tubules of inflamed kidneys but not in normals. In other organs VEGF gene expression is induced by hypoxia and cytokines such as interleukin 1 (IL-1). To identify the cellular mechanisms in control of tubular VEGF production, we studied effects of hypoxia and IL-1beta in VEGF mRNA levels, VEGF secretion, and activity of the hypoxia-inducible dimeric transcription factor 1 (HIF-1alpha/beta) in human proximal tubular epithelial cells (PTECs) in primary culture.Methods: PTECs were grown in monolayers from human kidneys. Hypoxia was induced by incubation at 3% O2. VEGF mRNA was quantitated by competitive polymerase chain reaction following reverse transcription. VEGF was measured by enzyme-linked immunoassay. HIF-1alpha was demonstrated by Western blot analysis and HIF-1 DNA binding by gel shift assay.Results: Significant amounts of VEGF mRNA and VEGF protein were measured in PTEC extracts and culture media, respectively. Stimulation of VEGF synthesis at low O2 tension and following IL-1beta treatment was detectable at the protein level only. Nuclear HIF-1alpha protein levels and HIF-1 binding to DNA were also increased under these conditions.Conclusions: PTECs in culture produce VEGF. One mechanism of induction appears to be increased DNA binding of HIF-1 to hypoxia-responsive elements in the VEGF gene promoter. In inflammatory diseases of the kidney, tubular cell-derived VEGF may contribute to microvascular leakage and monocyte extravasation. [ABSTRACT FROM AUTHOR]- Published
- 2000
- Full Text
- View/download PDF
7. miR663 Prevents Epo Inhibition Caused by TNF-Alpha in Normoxia and Hypoxia.
- Author
-
Ozkurt M, Hellwig-Bürgel T, Depping R, Kadabere S, Ozyurt R, Karadag A, and Erkasap N
- Abstract
Objective: In chronic inflammatory diseases, proinflammatory cytokines such as TNF- α are present in high amounts in the circulation and are associated with anemia in most cases. Experimental studies have shown that TNF- α inhibits the synthesis of erythropoietin (Epo), the main stimulant of hematopoiesis. Our aim was to figure out which microRNAs are involved in the Epo repression by TNF- α ., Methods: First, we determined the dose of TNF- α in HepG2 cells that has no cytotoxic effect by using MTT assays and that inhibits Epo synthesis by qRT-PCR and ELISA. Then, we performed the microRNA array study with TNF- α (20 ng/ml)-treated cells, and the array results were confirmed by qRT-PCR. We transfected the miR663 group with the mimic-miR663 (30 pmol) for 24 hrs; other groups were treated with a transfection reagent followed by treatment of TNF- α for 24 hrs; miR663 groups were treated with TNF- α for 24 hrs; and the control group was incubated with normal medium. We analyzed Epo mRNA levels by qRT-PCR. If mimic-miR663 prevents the Epo repression by TNF- α , more Epo-dependent UT-7 cells would survive. Therefore, we cocultured HepG2 cells with UT-7 cells. The percentage of apoptotic UT-7 cells was determined by TUNEL assays., Results: According to our array study, TNF- α significantly decreases miR663 expression. After transfection of miR663 mimics into HepG2 cells, TNF-alpha was unable to decrease Epo mRNA amounts. Furthermore, mimic-miR663 transfection resulted in a lower apoptosis rate of UT-7 cells in coculture experiments., Conclusions: miR663 is involved in Epo mRNA production and that is able to prevent or reverse the inhibitory effect of TNF- α . In our coculture study, transfecting HepG2 cells with miR663 mimics decreased the apoptosis of UT-7 cells., Competing Interests: The authors declare that there are no conflicts of interest., (Copyright © 2021 Mete Ozkurt et al.)
- Published
- 2021
- Full Text
- View/download PDF
8. Hypoxia abrogates antichlamydial properties of IFN-γ in human fallopian tube cells in vitro and ex vivo.
- Author
-
Roth A, König P, van Zandbergen G, Klinger M, Hellwig-Bürgel T, Däubener W, Bohlmann MK, and Rupp J
- Subjects
- Adaptive Immunity, Chlamydia Infections etiology, Chlamydia trachomatis growth & development, Chlamydia trachomatis immunology, Fallopian Tubes cytology, Female, Humans, Hypoxia immunology, Indoleamine-Pyrrole 2,3,-Dioxygenase genetics, Urinary Tract Infections etiology, Chlamydia Infections immunology, Fallopian Tubes immunology, Fallopian Tubes microbiology, Hypoxia microbiology, Interferon-gamma immunology, Urinary Tract Infections immunology
- Abstract
IFN-γ has an important role in the adaptive immune response against intracellular pathogens. In urogenital tract (UGT) infections with the obligate intracellular pathogen Chlamydia trachomatis, IFN-γ-mediated control of chlamydial growth implies the JAK-STAT signaling cascades and subsequent induction of the indoleamine 2,3-dioxygenase (IDO). As oxygen concentrations in the UGT are low under physiological conditions (O(2) < 5%) and further decrease during an inflammatory process, we wondered whether antibacterial properties of IFN-γ are maintained under hypoxic conditions. Using primary cells that were isolated from human fallopian tubes and an ex vivo human fallopian tube model (HFTM), we found that even high IFN-γ concentrations (200 units/mL) were not sufficient to limit growth of C. trachomatis under hypoxia. Reduced antibacterial activity of IFN-γ under hypoxia was restricted to the urogenital serovars D and L(2), but was not observed with the ocular serovar A. Impaired effectiveness of IFN-γ on chlamydial growth under hypoxia was accompanied by reduced phosphorylation of Stat-1 on Tyr701 and diminished IDO activity. This study shows that IFN-γ effector functions on intracellular C. trachomatis depend on the environmental oxygen supply, which could explain inadequate bacterial clearance and subsequent chronic infections eventually occurring in the UGT of women.
- Published
- 2010
- Full Text
- View/download PDF
9. Isolation and characterisation of mesenchymal stromal cells from adult mouse kidney.
- Author
-
Waldmann J, Lindner U, Hellwig-Bürgel T, Jelkmann W, Lehnert H, Rohwedel J, and Kramer J
- Published
- 2010
10. Timing and targeting of cell-based VEGF165 gene expression in ischemic tissue.
- Author
-
Spanholtz T, Maichle A, Niedworok C, Stoeckelhuber BM, Krüger S, Wedel T, Aach T, Middeler G, Hellwig-Bürgel T, Bader A, Krengel S, Müller OJ, Franz WM, Lindenmaier W, and Machens HG
- Subjects
- Adenoviridae, Animals, Cell Proliferation, Endothelium, Vascular cytology, Endothelium, Vascular physiology, Female, Fibroblasts cytology, Gene Transfer Techniques, Ischemia pathology, Models, Animal, Neovascularization, Physiologic physiology, Rats, Rats, Sprague-Dawley, Surgical Flaps blood supply, Time Factors, Transfection, Vascular Endothelial Growth Factor A genetics, Fibroblasts metabolism, Gene Expression Regulation physiology, Genetic Therapy methods, Ischemia metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
Background: Therapeutic angiogenesis has become a key technology in experimental and clinical medicine. Only few data are available on the effects of timing and targeting of therapeutic proteins after cell-based gene transfer. This work investigates such effects after temporary expression of vascular endothelial growth factor 165 (VEGF(165)), the most commonly used angiogenic protein for therapeutic purposes., Methods: We established a cell-based gene-transfer model using fibroblasts to temporarily produce VEGF(165). Cells were implanted into 40 rats. Protein expression and angiogenic effects were measured by PCR, immunohistology, and microangiography. To determine an improvement for survival of ischemically challenged tissue, cells were implanted in an ischemic flap model at different locations and time points., Results: After implantation of modified cells, a temporary increase was found in the target tissue for VEGF(165), endothelial cell counts, and capillary network formations. Four wk later, histological alterations in the target tissue area were not different from controls. Implantation of modified cells into flap plus wound margin 1 wk before surgery showed significant improvement of tissue survival demonstrated by planimetric measurements and blood vessels counting in the target tissue., Conclusion: In our model, temporary expression of VEGF(165) induces therapeutically relevant angiogenesis and improves blood supply only if applied 1 wk before ischemia. It is essential to include the surrounding area for induction of angiogenesis in this model. In contrast, the angiogenic effects are not effective in the target area and its surrounding tissue, if therapeutic gene expression is started during onset of ischemia or 2 wk before ischemia in this model.
- Published
- 2009
- Full Text
- View/download PDF
11. Erythropoietin production: Molecular mechanisms of the antagonistic actions of cyclic adenosine monophosphate and interleukin-1.
- Author
-
Batmunkh C, Krajewski J, Jelkmann W, and Hellwig-Bürgel T
- Subjects
- Blotting, Western, Cells, Cultured, Cyclic AMP metabolism, Cyclic CMP analogs & derivatives, Cyclic CMP pharmacology, Electrophoretic Mobility Shift Assay, Erythropoietin biosynthesis, GATA2 Transcription Factor metabolism, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Interleukin-1 metabolism, NF-kappa B antagonists & inhibitors, Promoter Regions, Genetic, RNA, Messenger genetics, RNA, Messenger metabolism, Cyclic AMP pharmacology, Erythropoietin genetics, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Interleukin-1 antagonists & inhibitors, NF-kappa B metabolism
- Abstract
Erythropoietin (Epo) mRNA expression is suppressed by interleukin 1 (IL-1). Cyclic adenosine monophosphate (cAMP) can increase Epo mRNA and Epo protein levels in IL-1 treated HepG2 cells to some extent. To identify molecular mechanisms of this reaction we investigated three transcription factors (NF-kappaB, GATA-2 and HIF-1) that control the Epo gene. Western blot analyses and electrophoretic mobility shift assays (EMSAs) revealed that IL-1 strongly activated NF-kappaB, which is a likely suppressor of the Epo promoter. Treatment of the cells with dibutyryl-cAMP (Bt2-cAMP) inhibited the activation of NF-kappaB by IL-1. Bt2-cAMP increased GATA-2 DNA binding. Since GATA-2 is a suppressor of the Epo promoter, GATA-2 activation was unlikely to cause the increase of Epo mRNA expression in IL-1 treated cells. Furthermore, Western blots, EMSAs and reporter gene studies showed that Bt2-cAMP was without effect on the hypoxia-inducible transcription factor HIF-1. Thus, NF-kappaB is probably the primary transcription factor by which cAMP counteracts the inhibition of Epo gene expression by IL-1.
- Published
- 2006
- Full Text
- View/download PDF
12. Review: hypoxia-inducible factor-1 (HIF-1): a novel transcription factor in immune reactions.
- Author
-
Hellwig-Bürgel T, Stiehl DP, Wagner AE, Metzen E, and Jelkmann W
- Subjects
- Animals, Cytokines immunology, Humans, Hypoxia-Inducible Factor 1, Hypoxia-Inducible Factor 1, alpha Subunit, MAP Kinase Signaling System immunology, Proteasome Endopeptidase Complex immunology, DNA-Binding Proteins immunology, Nuclear Proteins immunology, Transcription Factors immunology
- Abstract
Hypoxia-inducible factor-1 (HIF-1) is a dimeric transcriptional complex that has been recognized primarily for its role in the maintenance of oxygen and energy homoeostasis. The HIF-1alpha subunit is O(2) labile and is degraded by the proteasome following prolyl-hydroxylation and ubiquitination in normoxic cells. The present review summarizes evidence that HIF-1 is also involved in immune reactions. Immunomodulatory peptides, including interleukin-1 (IL-1) and tumor necrosis factor-alpha (TNF-alpha), stimulate HIF-1 dependent gene expression even in normoxic cells. Both the hypoxic and the cytokine-induced activation of HIF-1 involve the phosphatidylinositol- 3-kinase (PI3K) and the mitogen-activated protein kinase (MAPK) signaling pathways. In addition, heat shock proteins (HSP) and other cofactors interact with HIF-1 subunits. HIF-1 increases the transcription of several genes for proteins that promote blood flow and inflammation, including vascular endothelial growth factor (VEGF), heme oxygenase-1, endothelial and inducible nitric oxide synthase (NOS) and cyclooxygenase-2 (COX-2). The pharmacologic activation of the HIF-1 complex can be desirable in ischemic and inflammatory disorders. In contrast, HIF-1 blockade may be beneficial to prevent tumor angiogenesis and tumor growth.
- Published
- 2005
- Full Text
- View/download PDF
13. Regulation of the prolyl hydroxylase domain protein 2 (phd2/egln-1) gene: identification of a functional hypoxia-responsive element.
- Author
-
Metzen E, Stiehl DP, Doege K, Marxsen JH, Hellwig-Bürgel T, and Jelkmann W
- Subjects
- Base Sequence, Binding Sites, Cell Line, Tumor, CpG Islands physiology, Humans, Hypoxia-Inducible Factor-Proline Dioxygenases, Molecular Sequence Data, Sequence Homology, Nucleic Acid, Transcription Initiation Site physiology, Gene Expression Regulation physiology, Immediate-Early Proteins metabolism, Procollagen-Proline Dioxygenase metabolism, Promoter Regions, Genetic physiology
- Abstract
The HIFs (hypoxia-inducible factors) are a family of heterodimeric transcription factors essential for the adaptation of cells to reduced oxygen supply. Three human PHDs (prolyl hydroxylase domain proteins, PHD1-PHD3) initiate oxygen-dependent degradation of HIF-alpha-subunits in normoxia. RNA interference directed against PHD2, but not PHD1 or PHD3, is sufficient to stabilize HIF-1alpha in normoxia. Therefore PHD2 is regarded as the main cellular oxygen sensor. PHD2 itself is up-regulated by hypoxia and may thus limit hypoxic signalling. By sequence analysis, we predicted a promoter approx. 3.5 kb 5' of the translation start codon and a second promoter located in a CpG island immediately upstream of the coding sequence. A consensus HIF-1-binding site that is conserved in the murine phd2 gene was detected in the CpG island. By electrophoretic mobility-shift assay, we demonstrated binding of HIF-1 to the putative HIF-1-binding site. In luciferase reporter vectors, the isolated upstream promoter was inactive in all cell lines tested unless 200 bp were deleted at the 3'-end. The downstream promoter was active and induced by hypoxia. In reporter vectors containing both promoter sequences, luciferase activity was equal to vectors containing only the downstream promoter. In cells transfected with a vector containing both promoters, a single luciferase transcript was detectable. This transcript had the same length as transcripts from a vector containing the downstream promoter only. We conclude that the phd2 gene is transcribed exclusively from the downstream promoter that contains a functional hypoxia-responsive, cis-regulatory element. Our results establish that PHD2 is a direct HIF target gene.
- Published
- 2005
- Full Text
- View/download PDF
14. VEGF production by primary human renal proximal tubular cells: requirement of HIF-1, PI3-kinase and MAPKK-1 signaling.
- Author
-
Hellwig-Bürgel T, Stiehl DP, Katschinski DM, Marxsen J, Kreft B, and Jelkmann W
- Subjects
- Blotting, Western, Cell Hypoxia drug effects, Cells, Cultured, Chromones pharmacology, Dose-Response Relationship, Drug, Enzyme Activation drug effects, Fluorescent Antibody Technique, Humans, Hypoxia-Inducible Factor 1, Hypoxia-Inducible Factor 1, alpha Subunit, Kidney Tubules, Proximal cytology, Kidney Tubules, Proximal enzymology, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Morpholines pharmacology, Oxygen metabolism, Phosphoinositide-3 Kinase Inhibitors, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt, Reverse Transcriptase Polymerase Chain Reaction, Vascular Endothelial Growth Factors genetics, DNA-Binding Proteins metabolism, Kidney Tubules, Proximal metabolism, Mitogen-Activated Protein Kinase Kinases metabolism, Nuclear Proteins metabolism, Phosphatidylinositol 3-Kinases metabolism, Signal Transduction drug effects, Transcription Factors metabolism, Vascular Endothelial Growth Factors biosynthesis
- Abstract
Renal proximal tubular epithelial cells (PTEC) respond to hypoxia exposure or interleukin-1beta (IL-1beta) treatment with increased vascular endothelial growth factor (VEGF) production. With respect to O2 deprivation, the hypoxia-inducible factor 1alpha/ beta (HIF-1) is the most important transcription factor driving VEGF mRNA expression. HIF-1 is also activated by IL-1beta and may thus be involved in the stimulation of VEGF production by this cytokine. However, the molecular mechanisms of HIF-1 dependent VEGF synthesis are poorly understood. Herein, human PTEC in primary culture were challenged by hypoxic incubation and/or IL-1beta treatment in absence or presence of specific phosphatidylinositol 3-kinase (PI3K) or mitogen activated protein kinase kinase-1 (MAPKK-1) inhibitors for assay of VEGF protein, VEGF mRNA and detection of HIF-1alpha by Western Blotting, EMSA and fluorescence microscopy. In addition, the activities of PI3K and MAPKK-1 were studied following hypoxia and IL-1beta treatment of the cultures. The study shows that PI3K but not MAPKK-1 inhibition resulted in the loss of hypoxic and IL-1beta induced HIF-1alpha accumulation, whereas VEGF synthesis was reduced by either intervention. Thus, PI3K signaling is required for HIF-1alpha accumulation and VEGF synthesis, whereas MAPKK-1 signaling is required for VEGF synthesis only. Furthermore, hypoxia alone was sufficient to activate PI3K in PTEC in contrast to MAPKK-1, whose activity was lowered in hypoxia., (Copyright 2005 S. Karger AG, Basel.)
- Published
- 2005
- Full Text
- View/download PDF
15. Deferoxamine induces prolonged cardiac preconditioning via accumulation of oxygen radicals.
- Author
-
Dendorfer A, Heidbreder M, Hellwig-Bürgel T, Jöhren O, Qadri F, and Dominiak P
- Subjects
- Aldehyde Reductase metabolism, Alkaloids, Animals, Aryl Hydrocarbon Receptor Nuclear Translocator, Basic Helix-Loop-Helix Transcription Factors, Benzophenanthridines, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Glucose Transporter Type 4, Glycine pharmacology, Hypoxia-Inducible Factor 1, alpha Subunit, Male, Monosaccharide Transport Proteins metabolism, Muscle Proteins metabolism, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Reperfusion Injury metabolism, Phenanthridines pharmacology, Protein Kinase C antagonists & inhibitors, Protein Kinase C metabolism, Rats, Rats, Wistar, Receptors, Aryl Hydrocarbon genetics, Receptors, Aryl Hydrocarbon metabolism, Sulfhydryl Compounds pharmacology, Trans-Activators genetics, Trans-Activators metabolism, Transcription Factors genetics, Transcription Factors metabolism, Deferoxamine pharmacology, Glycine analogs & derivatives, Iron Chelating Agents pharmacology, Ischemic Preconditioning, Myocardial, Myocardial Reperfusion Injury pathology, Superoxides metabolism
- Abstract
Iron chelation by deferoxamine (DFO) blocks the Fenton reaction, but also inhibits prolyl hydroxylases and thereby activates certain hypoxia-inducible transcription factors (HIFs) that trigger cellular adaptation to hypoxia. Because both mechanisms may alleviate tissue damage in ischemia and reperfusion, we tried to differentiate their contribution to DFO-induced cardioprotection. Myocardial ischemia and reperfusion were induced in anesthetized Wistar rats. Infarct size was related to the ischemic area. Myocardial mRNA expression was determined by real-time PCR. Radical reactivity was probed in myocardial tissue slices with the redox-sensitive dye CM-H(2)DCFDA. Single ip applications of DFO (200 mg/kg) administered 2 h to 3 days before infarction reduced infarct size from 55 +/- 7% to 22-26%. Protection was abolished by the radical scavenger N-(2-mercaptopropionyl)glycine and the protein kinase C inhibitor chelerythrine when either was given 30 min before DFO, whereas subsequent application was ineffective. DFO did not alter the expression of various HIF target genes, whereas mRNAs of HIF-independent genes, aldose reductase and glucose transporter-4, were increased in infarcted myocardium 2 days after DFO treatment. Enhancement of superoxide activity by DFO could be demonstrated in vitro. Acute and prolonged myocardial preconditioning is triggered by DFO in response to accumulation of oxygen radicals and activation of protein kinase C.
- Published
- 2005
- Full Text
- View/download PDF
16. Intracellular localisation of human HIF-1 alpha hydroxylases: implications for oxygen sensing.
- Author
-
Metzen E, Berchner-Pfannschmidt U, Stengel P, Marxsen JH, Stolze I, Klinger M, Huang WQ, Wotzlaw C, Hellwig-Bürgel T, Jelkmann W, Acker H, and Fandrey J
- Subjects
- Cell Compartmentation physiology, Genes, Reporter genetics, Green Fluorescent Proteins, Humans, Hypoxia-Inducible Factor 1, alpha Subunit, Luminescent Proteins, Mixed Function Oxygenases, Osteosarcoma metabolism, Procollagen-Proline Dioxygenase genetics, Protein Isoforms metabolism, RNA, Messenger metabolism, Recombinant Fusion Proteins, Repressor Proteins genetics, Repressor Proteins metabolism, Transcription Factors genetics, Transcriptional Activation genetics, Tumor Cells, Cultured, Cell Hypoxia physiology, Cell Nucleus enzymology, Cytoplasm enzymology, Oxygen metabolism, Procollagen-Proline Dioxygenase metabolism, Transcription Factors metabolism
- Abstract
Hypoxia-inducible factor1 (HIF-1) is an essential transcription factor for cellular adaptation to decreased oxygen availability. In normoxia the oxygen-sensitive alpha-subunit of HIF-1 is hydroxylated on Pro564 and Pro402 and thus targeted for proteasomal degradation. Three human oxygen-dependent HIF-1 alpha prolyl hydroxylases (PHD1, PHD2, and PHD3) function as oxygen sensors in vivo. Furthermore, the asparagine hydroxylase FIH-1 (factor inhibiting HIF) has been found to hydroxylate Asp803 of the HIF-1 C-terminal transactivation domain, which results in the decreased ability of HIF-1 to bind to the transcriptional coactivator p300/CBP. We have fused these enzymes to the N-terminus of fluorescent proteins and transiently transfected the fusion proteins into human osteosarcoma cells (U2OS). Three-dimensional 2-photon confocal fluorescence microscopy showed that PHD1 was exclusively present in the nucleus, PHD2 and FIH-1 were mainly located in the cytoplasm and PHD3 was homogeneously distributed in cytoplasm and nucleus. Hypoxia did not influence the localisation of any enzyme under investigation. In contrast to FIH-1, each PHD inhibited nuclear HIF-1 alpha accumulation in hypoxia. All hydroxylases suppressed activation of a cotransfected hypoxia-responsive luciferase reporter gene. Endogenous PHD2mRNA and PHD3mRNA were hypoxia-inducible, whereas expression of PHD1mRNA and FIH-1mRNA was oxygen independent. We propose that PHDs and FIH-1 form an oxygen sensor cascade of distinct subcellular localisation.
- Published
- 2003
- Full Text
- View/download PDF
17. Inhibition of erythropoietin gene expression signaling involves the transcription factors GATA-2 and NF-kappaB.
- Author
-
La Ferla K, Reimann C, Jelkmann W, and Hellwig-Bürgel T
- Subjects
- Blotting, Northern, Cell Hypoxia physiology, Electrophoretic Mobility Shift Assay, GATA2 Transcription Factor, Gene Expression Regulation drug effects, Humans, Interleukin-1 pharmacology, Oligonucleotides metabolism, Protein Binding drug effects, RNA, Messenger drug effects, RNA, Messenger genetics, RNA, Messenger metabolism, Signal Transduction drug effects, Tumor Cells, Cultured, Tumor Necrosis Factor-alpha pharmacology, DNA-Binding Proteins metabolism, Erythropoietin genetics, NF-kappa B metabolism, Transcription Factors metabolism
- Abstract
The anemia of chronic inflammatory and malignant diseases is partly due to impaired synthesis of the hormone erythropoietin (Epo). The proinflammatory cytokines interleukin-1 (IL-1) and tumor necrosis factor a (TNF-alpha) suppress in vitro Epo gene expression and Epo protein secretion. However, the molecular mechanisms of this inhibition are poorly understood. The human Epo promoter and the 5' flanking region contain several recognition sequences for transcription factors acting either positively or negatively. Herein, we investigated the roles of the transcription factors GATA-2 and NF-kappaB in the modulation of Epo gene expression by IL-1beta and TNF-alpha in the human hepatoma cell line HepG2. Electrophoretic mobility shift assays revealed increased GATA-2 and NF-kappaB DNA binding in cells treated with IL-1beta or TNF-alpha. Reporter gene assays with a sequence from the Epo promoter in front of the firefly luciferase gene showed that the cytokines reduced Epo reporter gene activity. Functional inactivation of GATA-2 and NF-kappaB by oligo-decoy techniques prevented the inhibition of Epo production by IL-1beta and TNF-alpha. In HepG2 cells stably transfected with a dominant-negative form of IkappaBalpha, the activation of NF-kappaB was inhibited, while Epo mRNA levels and Epo secretion increased. Thus, both GATA-2 and NF-kappaB seem to be involved in the suppression of Epo gene expression by IL-1beta and TNF-alpha in vitro and may be responsible for impaired Epo synthesis in inflammatory diseases in vivo.
- Published
- 2002
- Full Text
- View/download PDF
18. Preferential topography of proteins regulating vascularization and apoptosis in a MX1 xenotransplant after treatment with hypoxia, hyperthermia, ifosfamide, and irradiation.
- Author
-
Schmitt O, Schubert C, Feyerabend T, Hellwig-Bürgel T, Weiss C, and Kühnel W
- Subjects
- Animals, Cell Adhesion Molecules metabolism, Combined Modality Therapy, Cytokines metabolism, Endothelial Growth Factors metabolism, Endothelins metabolism, Extracellular Matrix Proteins metabolism, Factor VIII metabolism, Female, Ifosfamide pharmacology, Immunohistochemistry, Lymphokines metabolism, Mice, Mice, Nude, Nerve Growth Factor metabolism, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Xenograft Model Antitumor Assays, Antineoplastic Agents, Alkylating pharmacology, Apoptosis physiology, Breast Neoplasms blood supply, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Breast Neoplasms pathology, Breast Neoplasms radiotherapy, Hyperthermia, Induced, Hypoxia, Neovascularization, Pathologic
- Abstract
The MX1 xenotransplant growing in nude mice was used as a model for estrogen- and progesterone-receptor-negative breast cancer. The effects of different therapeutic regimens-combinations of hyperthermia, chemotherapy, and irradiation-on the expression of proteins playing a role in tumor vascularization and apoptosis were investigated. Additionally, MX-1 tumors were exposed to hypoxia to investigate changes in protein expression related to angiogenesis. This is of particular importance with respect to antiangiogenic therapies that may be combined with the treatments mentioned before. Endothelial and adhesion factors, extracellular matrix (ECM) factors, apoptosis-regulating factors, and neuronal factors were examined by immunohistochemical techniques. Concerning vascularization, the most prominent changes were seen in the expression of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), which increased strongly after hypoxia. The other cytokines, adhesion and ECM molecules, were either little affected or unaffected by the therapy. At the ultrastructural level, the walls of the tumor vessels are of the sinusoidal type, possessing many fenestrations. With regard to the second focus of this investigation, apoptosis, tumor cells again exerted the strongest differences after hypoxia where c-myc was clearly enhanced, whereas the effects on p53, bcl-2, and CD95 were extremely weak or not detectable. Furthermore, the neurotransmitter somatostatin, a possible "external" regulator of apoptosis, did not show treatment-related changes. In summary, it was shown that 1) within the group of apoptosis-regulating proteins c-myc was particularly affected by hypoxia, indicating a possible role for an activation-induced pathway of apoptosis in this context; 2) minor changes seen after treatment combined with hyperthermia point to a more acute vascular reaction (=dilatation), causing an increase of tissue pO2 rather than angiogenesis; and 3) the concentrations of the angiogenic factors VEGF and bFGF rose strongly under hypoxia, thereby possibly exerting counterproductive effects to antiangiogenic therapy but not to thermochemotherapy or irradiation. This supports the concept of a combined antiangiogenic, hyperthermia, chemo- and irradiation therapy.
- Published
- 2002
- Full Text
- View/download PDF
19. Normoxic induction of the hypoxia-inducible factor 1alpha by insulin and interleukin-1beta involves the phosphatidylinositol 3-kinase pathway.
- Author
-
Stiehl DP, Jelkmann W, Wenger RH, and Hellwig-Bürgel T
- Subjects
- Cell Nucleus metabolism, Endothelial Growth Factors biosynthesis, Erythropoietin biosynthesis, Gene Expression Regulation, Humans, Hypoxia-Inducible Factor 1, alpha Subunit, Lymphokines biosynthesis, Phosphoinositide-3 Kinase Inhibitors, Tumor Cells, Cultured, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Insulin pharmacology, Interleukin-1 pharmacology, Oxygen pharmacology, Phosphatidylinositol 3-Kinases metabolism, Transcription Factors biosynthesis
- Abstract
Hypoxia-inducible factor 1 (HIF-1) is a heterodimeric DNA-binding complex of the subunits alpha and beta with relevance in O(2) and energy homeostasis. The labile component, HIF-1alpha, is not only activated by hypoxia but also by peptides such as insulin and interleukin-1 (IL-1) in normoxia. We investigated whether inhibitors of mitogen-activated protein kinase kinases (MAPKKs: PD 98059, U0126) and phosphatidylinositol 3-kinase (PI3K: LY 294002) do not only lower the hypoxia-induced, but also the insulin- and IL-1-induced HIF-1alpha accumulation and HIF-1 DNA-binding in human hepatoma cell cultures (line HepG2). The results show that LY 294002 suppressed HIF-1 activation in a dose-dependent manner irrespective of the stimulus. With respect to target proteins controlled by HIF-1, the production of erythropoietin was fully blocked and that of vascular endothelial growth factor reduced following inhibition of the PI3K pathway. The role of MAPKKs in this process remained in question, because PD 98059 and U0126 did not significantly reduce HIF-1alpha levels at non-toxic doses. We propose that PI3K signaling is not only important in the hypoxic induction of HIF-1 but it is also crucially involved in the response to insulin and IL-1.
- Published
- 2002
- Full Text
- View/download PDF
20. Vascular endothelial growth factor gene expression in the human breast cancer cell line MX-1 is controlled by O2 availability in vitro and in vivo.
- Author
-
Marxsen JH, Schmitt O, Metzen E, Jelkmann W, and Hellwig-Bürgel T
- Subjects
- Adult, Animals, Breast Neoplasms pathology, Endothelial Growth Factors analysis, Enzyme-Linked Immunosorbent Assay, Female, Humans, Immunohistochemistry, Lymphokines analysis, Mice, Mice, Nude, Oxygen pharmacology, Partial Pressure, Polymerase Chain Reaction, Transplantation, Heterologous, Tumor Cells, Cultured, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Breast Neoplasms genetics, Cell Hypoxia, Endothelial Growth Factors genetics, Gene Expression Regulation, Neoplastic drug effects, Lymphokines genetics, Oxygen physiology
- Abstract
The vascular endothelial growth factor (VEGF) plays an important role in angiogenesis. Mediated by the hypoxia-inducible transcription factor HIF-1alpha/beta, a reduction in O2 tension (pO2) leads to increased VEGF gene expression in nonmalignant tissues. In tumor cells VEGF mRNA levels are often constitutively elevated. We examined pO2-dependent VEGF mRNA expression and VEGF protein formation in the human breast cancer cell line MX-1 in vitro and in vivo. For in vitro study MX-1 cultures were grown on dishes with a gas-permeable bottom to expose the cells to defined O2 concentrations (from 95% to 0%) for 4 h. Northern blot analysis showed significant VEGF mRNA in MX-1 cultures under normoxic conditions which was further increased by hypoxia. The amount of secreted VEGF was also elevated in hypoxic cultures. Western blot analysis revealed a correlation between the severity of hypoxia and HIF-1alpha protein amounts in the nucleus. Furthermore, DNA-binding activity of HIF-1 could be demonstrated by gel-shift assays. For in vivo study immunodeficient nude mice bearing MX-1 tumor transplants were exposed to inspiratory hypoxia (10% O2). Northern blot and immunohistochemical analyses of MX-1 tumor transplants showed that VEGF mRNA and VEGF protein levels were increased in mice 17 h after the induction of inspiratory hypoxia. Thus, pO2-dependence of VEGF gene expression can be maintained in cancer cells, even in vivo, which may be relevant in regard to therapeutic attempts to inhibit tumor angiogenesis by increasing tumor oxygenation.
- Published
- 2001
- Full Text
- View/download PDF
21. Biology of erythropoietin.
- Author
-
Jelkmann W and Hellwig-Bürgel T
- Subjects
- Anemia drug therapy, Animals, Aryl Hydrocarbon Receptor Nuclear Translocator, Erythropoietin genetics, Erythropoietin metabolism, Erythropoietin therapeutic use, Humans, Hypoxia-Inducible Factor 1, alpha Subunit, Recombinant Proteins, Transcription Factors metabolism, DNA-Binding Proteins, Erythropoietin physiology, Receptors, Aryl Hydrocarbon
- Abstract
Hypoxia induces tissue-specific gene products such as erythropoietin (EPO) and vascular endothelial growth factor (VEGF), which improve the peripheral O2 supply, and glucose transporters and glycolytic enzymes, which adapt cells to reduced O2 availability. EPO has been the fountainhead in research on pO2-dependent synthesis of proteins. The EPO gene enhancer (like the flanking DNA-elements of several other pO2-controlled genes) contains a consensus sequence (CGTG) that binds the trans-acting dimeric hypoxia-inducible factor 1 (HIF-1alpha/beta). The alpha-subunit of HIF-1 is rapidly degraded by the proteasome under normoxic conditions, but it is stabilized on occurrence of hypoxia. HIF-1 DNA-binding is also increased by insulin, and by interleukin-1 and tumor necrosis factor. Thus, in some aspects there is synergy in the cellular responses to hypoxia, glucose deficiency and inflammation. In viewing clinical medicine recombinant human EPO (rHu-EPO) has become the mainstay of treatment for renal anemia. Endogenous EPO and rHu-EPO are similar except for minor differences in the pattern of their 4 carbohydrate chains. RHu-EPO is also administered to patients suffering from non-renal anemias, such as in autoimmune diseases or malignancies. The correction of anemia in patients with solid tumors is not merely considered a palliative intervention. Hypoxia promotes tumor growth. However, the benefits of the administration of rHu-EPO to tumor patients with respect to its positive effects on tumor oxygenation, tumor growth inhibition and support of chemo- and radiotherapy is still debatable ground.
- Published
- 2001
- Full Text
- View/download PDF
22. Transcription and proper splicing of a mammalian gene in yeast.
- Author
-
Kunze B, Hellwig-Bürgel T, Weichenhan D, and Traut W
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Blotting, Northern, Chromosomes, Artificial, Yeast, DNA, Complementary genetics, Gene Expression Regulation, Mice, Mice, Inbred C57BL, Molecular Sequence Data, RNA genetics, RNA metabolism, Repetitive Sequences, Nucleic Acid, Antigens, Nuclear, Nuclear Proteins, Proteins genetics, RNA Splicing, Saccharomyces cerevisiae genetics, Transcription, Genetic
- Abstract
The house mouse strain C57BL/6 harbours 64 copies of the multicopy gene Sp100-rs. Three of these are contained in the yeast artificial chromosome (YAC) clone yMm75. Four Sp100-rs transcripts of 3.0, 2.6, 1.6 and 1.3kb were detected by Northern hybridization in the yMm75-harbouring line of Saccharomyces cerevisiae. Additional and less abundant transcripts were detected by RT-PCR. With one exception, the YAC-derived Sp100-rs transcripts were a subset of those found in the C57BL/6 mouse. This indicates transcription and proper splicing of murine pre-mRNAs in yeast. Analysis of the splice sites shows that the yeast splicing machinery accepts splice sites that deviate from the standard yeast consensus sequences. It may be feasible, therefore, at least in a fair proportion of cases, to exploit the mammalian mRNAs present in transgenic yeast for gene recognition of YAC-inserts.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.