Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Endocrinol ; 2021: 3670499, 2021.
Article in English | MEDLINE | ID: mdl-34367277

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.

2.
J Clin Endocrinol Metab ; 97(3): 978-86, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22259067

ABSTRACT

CONTEXT: Mitochondrial capacity and metabolic potential are under the control of hormones, such as thyroid hormones. The most proximal regulator of the hypothalamic-pituitary-thyroid (HPT) axis, TRH, is the key hypothalamic integrator of energy metabolism via its impact on thyroid hormone secretion. OBJECTIVE: Here, we asked whether TRH directly modulates mitochondrial functions in normal, TRH-receptor-positive human epidermis. METHODS: Organ-cultured human skin was treated with TRH (5-100 ng/ml) for 12-48 h. RESULTS: TRH significantly increased epidermal immunoreactivity for the mitochondria-selective subunit I of respiratory chain complex IV (MTCO1). This resulted from an increased MTCO1 transcription and protein synthesis and a stimulation of mitochondrial biogenesis as demonstrated by transmission electron microscopy and TRH-enhanced mitochondrial DNA synthesis. TRH also significantly stimulated the transcription of several other mitochondrial key genes (TFAM, HSP60, and BMAL1), including the master regulator of mitochondrial biogenesis (PGC-1α). TRH significantly enhanced mitochondrial complex I and IV enzyme activity and enhanced the oxygen consumption of human skin samples, which shows that the stimulated mitochondria are fully vital because the main source for cellular oxygen consumption is mitochondrial endoxidation. CONCLUSIONS: These findings identify TRH as a potent, novel neuroendocrine stimulator of mitochondrial activity and biogenesis in human epidermal keratinocytes in situ. Thus, human epidermis offers an excellent model for dissecting neuroendocrine controls of human mitochondrial biology under physiologically relevant conditions and for exploring corresponding clinical applications.


Subject(s)
Epidermis/metabolism , Mitochondria/metabolism , Thyrotropin-Releasing Hormone/metabolism , Adult , Energy Metabolism , Humans , Middle Aged , Organ Culture Techniques , Receptors, Thyrotropin-Releasing Hormone/metabolism
3.
Proc Natl Acad Sci U S A ; 107(45): 19502-7, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20974954

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.


Subject(s)
Chlamydia Infections/immunology , Fallopian Tubes/immunology , Fallopian Tubes/microbiology , Hypoxia/microbiology , Interferon-gamma/immunology , Urinary Tract Infections/immunology , 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
4.
J Surg Res ; 151(1): 153-62, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18621399

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.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation/physiology , Genetic Therapy/methods , Ischemia/metabolism , Vascular Endothelial Growth Factor A/metabolism , 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
5.
Cell Microbiol ; 9(9): 2181-91, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17490410

ABSTRACT

Chlamydiaceae are obligate intracellular bacteria that cause endemic trachoma, sexually transmitted diseases and respiratory infections. The course of the diseases is determined by local inflammatory immune responses and the propensity of the pathogen to replicate within infected host cells. Both features require energy which is inseparably coupled to oxygen availability in the microenvironment. Hypoxia-inducible factor-1 (HIF-1) regulates crucial genes involved in the adaptation to low oxygen concentrations, cell metabolism and the innate immune response. Here we report that Chlamydia pneumoniae directly interferes with host cell HIF-1alpha regulation in a biphasic manner. In hypoxia, C. pneumoniae infection had an additive effect on HIF-1alpha stabilization resulting in enhanced glucose uptake during the early phase of infection. During the late phase of intracellular chlamydial replication, host cell adaptation to hypoxia was actively silenced by pathogen-induced HIF-1alpha degradation. HIF-1alpha was targeted by the chlamydial protease-like activity factor, which was secreted into the cytoplasm of infected cells. Direct interference with HIF-1alpha stabilization was essential for efficient C. pneumoniae replication in hypoxia and highlights a novel strategy of adaptive pathogen-host interaction in chlamydial diseases.


Subject(s)
Chlamydophila pneumoniae/metabolism , Host-Pathogen Interactions , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell Hypoxia , Cell Line , Chlamydia Infections/metabolism , Chlamydophila pneumoniae/cytology , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
6.
FEBS Lett ; 580(13): 3153-60, 2006 May 29.
Article in English | MEDLINE | ID: mdl-16684536

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.


Subject(s)
Cyclic AMP/pharmacology , Erythropoietin/genetics , Gene Expression Regulation , Interleukin-1/antagonists & inhibitors , NF-kappa B/metabolism , Blotting, Western , Cells, Cultured , Cyclic AMP/metabolism , Cyclic CMP/analogs & derivatives , Cyclic CMP/pharmacology , Electrophoretic Mobility Shift Assay , Erythropoietin/biosynthesis , GATA2 Transcription Factor/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , 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
7.
J Interferon Cytokine Res ; 25(6): 297-310, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15957953

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.


Subject(s)
DNA-Binding Proteins/immunology , Nuclear Proteins/immunology , Transcription Factors/immunology , Animals , Cytokines/immunology , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , MAP Kinase Signaling System/immunology , Proteasome Endopeptidase Complex/immunology
8.
Proc Natl Acad Sci U S A ; 102(9): 3447-52, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15722416

ABSTRACT

Atherosclerosis is characterized by inflammation and proliferation of vascular cells. The intracellular bacterium Chlamydia (Chlamydophila) pneumoniae uses blood monocytes [peripheral blood mononuclear cells (PBMCs)] for dissemination, has been found to persist in atherosclerotic lesions, and has been implicated in atherogenesis by small GTPase activation and T lymphocyte recruitment. Infection of human coronary artery smooth muscle cells with C. pneumoniae significantly induced mRNA and protein for the angiogenic transcription factor Egr-1, resulting in enhanced coronary artery smooth muscle cell proliferation, which was reduced by transfection with small interfering RNA duplexes targeted at Egr-1 mRNA. These effects required viable chlamydiae and depended on p44/42 mitogen-activated protein kinase activity but not on the p38 mitogen-activated protein kinase pathway. Postinfectious Egr-1 mRNA up-regulation in arterial vessels was confirmed ex vivo in a rat aortic ring model of focal vascular chlamydial infection. An in vivo model based on the injection of C. pneumoniae-infected PBMCs into mice confirmed Egr-1 mRNA up-regulation within 24 h of endovascular infection. Arterial injury from repeated direct chlamydial infections and cell-to-cell contact with C. pneumoniae-infected PBMCs might represent a chronic focus of proliferative activity linked to the media proliferation seen in advanced atherosclerosis. Overall, chlamydial infection induces a proliferative phenotype in vascular cells via transcription factor Egr-1 activation in vitro, ex vivo, and in vivo.


Subject(s)
Blood Vessels/pathology , Cell Division , Chlamydia Infections/pathology , Chlamydophila pneumoniae/pathogenicity , DNA-Binding Proteins/metabolism , Immediate-Early Proteins/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , Chlamydia Infections/metabolism , Chlamydia Infections/microbiology , DNA Probes , Early Growth Response Protein 1 , Electrophoretic Mobility Shift Assay , Humans , In Vitro Techniques , Mice , Phenotype , Polymerase Chain Reaction , Rats , Rats, Wistar
9.
Cell Physiol Biochem ; 15(1-4): 99-108, 2005.
Article in English | MEDLINE | ID: mdl-15665520

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.


Subject(s)
DNA-Binding Proteins/metabolism , Kidney Tubules, Proximal/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , Vascular Endothelial Growth Factors/biosynthesis , 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 , Signal Transduction/drug effects , Vascular Endothelial Growth Factors/genetics
10.
Biochem J ; 387(Pt 3): 711-7, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15563275

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.


Subject(s)
Gene Expression Regulation/physiology , Immediate-Early Proteins/metabolism , Procollagen-Proline Dioxygenase/metabolism , Promoter Regions, Genetic/physiology , 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
11.
Free Radic Biol Med ; 38(1): 117-24, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15589380

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.


Subject(s)
Deferoxamine/pharmacology , Glycine/analogs & derivatives , Iron Chelating Agents/pharmacology , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/pathology , Superoxides/metabolism , 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
12.
J Cell Sci ; 116(Pt 7): 1319-26, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12615973

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.


Subject(s)
Cell Hypoxia/physiology , Cell Nucleus/enzymology , Cytoplasm/enzymology , Oxygen/metabolism , Procollagen-Proline Dioxygenase/metabolism , Transcription Factors/metabolism , 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
13.
FASEB J ; 16(13): 1811-3, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12223449

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.


Subject(s)
DNA-Binding Proteins/metabolism , Erythropoietin/genetics , NF-kappa B/metabolism , Transcription Factors/metabolism , 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
14.
Am J Clin Oncol ; 25(4): 325-36, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12151958

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.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/physiology , Breast Neoplasms , Hyperthermia, Induced , Hypoxia , Neovascularization, Pathologic , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , 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
15.
FEBS Lett ; 512(1-3): 157-62, 2002 Feb 13.
Article in English | MEDLINE | ID: mdl-11852072

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.


Subject(s)
Insulin/pharmacology , Interleukin-1/pharmacology , Oxygen/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Transcription Factors/biosynthesis , 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
SELECTION OF CITATIONS
SEARCH DETAIL
...