Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
2.
Vision Res ; 110(Pt A): 76-86, 2015 May.
Article in English | MEDLINE | ID: mdl-25805148

ABSTRACT

The sampling of our visual environment through saccadic eye movements is an essential function of the brain, allowing us to overcome the limits of peripheral vision. Understanding which parts of a scene attract overt visual attention is subject to intense research, and considerable progress has been made in unraveling the underlying cortical mechanisms. In contrast to spatial aspects, however, relatively little is understood about temporal aspects of overt visual sampling. At every fixation, the oculomotor system faces the decision whether to keep exploring different aspects of an object or scene or whether to remain fixated to allow for in-depth cortical processing - a situation that can be understood in terms of an exploration-exploitation dilemma. To improve our understanding of the factors involved in these decisions, we here investigate how the level of visual information, experimentally manipulated by scene context and stimulus ambiguity, changes the sampling behavior preceding the recognition of centrally presented ambiguous and disambiguated objects. Behaviorally, we find that context, although only presented until the first voluntary saccade, biases the perceptual outcome and significantly reduces reaction times. Importantly, we find that increased information about an object significantly alters its visual exploration, as evident through increased fixation durations and reduced saccade amplitudes. These results demonstrate that the initial sampling of an object, preceding its recognition, is subject to change based on the amount of information available in the system: increased evidence for its identity biases the exploration-exploitation strategy towards in-depth analyses.


Subject(s)
Attention/physiology , Fixation, Ocular/physiology , Form Perception/physiology , Adult , Analysis of Variance , Eye Movements , Female , Humans , Male , Pattern Recognition, Visual/physiology , Photic Stimulation/methods , Reaction Time , Saccades/physiology , Young Adult
3.
HNO ; 54(9): 689-97, 2006 Sep.
Article in German | MEDLINE | ID: mdl-16479386

ABSTRACT

BACKGROUND: Transcription factor HIF-1 (hypoxia-inducible factor-1) regulates the expression of genes which are involved in glucose supply, growth, metabolism, redox reactions and blood supply. Hypoxia and ischemia play an important role in the pathogenesis of tinnitus and hearing loss. Therefore, HIF-1 activity and the expression of HIF-1 dependent genes in the cochlea were examined under ischemic and hypoxic conditions. MATERIAL AND METHODS: For the HIF-1 analysis, single-cell cultures of the organ of Corti (OC), stria vascularis (SV) and modiolus (MOD) were used. mRNA expression was analyzed in the organotypic culture using a microarray technique (RN U34-chip, Affymetrix). RESULTS: Ischemia (hypoxia without glucose) and pure hypoxia increase the HIF-1 activity identically, with the highest increase found in MOD and OC. The HIF-1 alpha mRNA levels were found to be higher in SV than in the OC and MOD. During culturing, there is a clear increase in HIF-1 alpha mRNA and the expression of a number of HIF-1 dependent genes, such as Gapdh/glyceraldehyde-3-phosphate dehydrogenase, Slc2a1/solute carrier family 2 (facilitated glucose transporter), member 1, Tf/transferrin and Tfrc/transferrin receptor, in all three regions. In SV, MOD and OC, increase in the expression of Hmox1/hemoxygenase 1, Nos2/nitric oxide synthase, inducible and Tfrc is particularly high. Hypoxia (5 h) results in an increased expression of Igf2/Insulin-like growth factor 2. CONCLUSION: The present data underline the contribution of radical forming processes to the pathogenesis of inner ear diseases. For experimental research, it is important to note that organotypic culture may be coupled with hypoxia.


Subject(s)
Cochlea/metabolism , Hypoxia-Inducible Factor 1/metabolism , Hypoxia/metabolism , Ischemia/metabolism , Animals , Animals, Newborn , Cell Hypoxia , Cells, Cultured , Gene Expression Regulation , Rats , Rats, Wistar
4.
Z Gastroenterol ; 44(1): 67-76, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16397842

ABSTRACT

Oxygen has important functions as substrate for biochemical reactions and as modulator of gene expression. In the liver, the physiologically occurring oxygen gradient is a major effector of metabolic zonation. In addition, cross-talks between the O2 signaling and nutrient signaling chains initiate a dynamic zonation pattern. Under pathological situations, hypoxia appears to be a major determinant for liver diseases and cancer. Thereby transcription factors of the HIF family are activated whereas USF proteins have the potential to counteract HIFs. In addition, feedback mechanisms between hypoxia, HIF and the IGF axes appear to exist. Thus, the knowledge of these mechanisms may help to initiate new therapies in diseases with disturbed O2 availability.


Subject(s)
Carbohydrate Metabolism , Hypoxia-Inducible Factor 1/metabolism , Hypoxia/metabolism , Liver Diseases/metabolism , Liver/metabolism , Oxygen/metabolism , Somatomedins/metabolism , Animals , Humans , Hypoxia/complications , Liver Diseases/etiology
5.
Anat Embryol (Berl) ; 207(6): 503-12, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14770308

ABSTRACT

The expression patterns of erythropoietin (EPO) and its receptor (EPOR) were investigated in the midbrain and in adjacent parts of the synencephalon and hindbrain of embryonic C57Bl mice. On embryonic (E) day 8 (E8), virtually all neuroepithelial cells expressed EPOR. After neural tube closure, subsets of these cells downregulated EPOR. In contrast, radial glial cells were EPOR-immunolabeled from E11 onwards. Simultaneously, subpopulations of early developing neurons upregulated EPO and expressed HIF-1, known to transcriptionally activate EPO. Three-dimensional reconstructions revealed subpopulations of EPO-expressing neurons: (1) in the trigeminal mesencephalic nucleus (TMN), (2) at the rostral transition of the midbrain and synencephalon, (3) in the basal plate of the midbrain, (4) in the trigeminal motor nucleus, and (5) in the trigeminal principal sensory nucleus. In the rostral midbrain and synencephalon, EPO-immunoreactive neurons were attached to EPOR-expressing radial glial cells. The identity of radial glial cells was proven by their immunoreactivity for antibodies against astrocyte-specific glutamate transporter, brain lipid-binding protein, and nestin. From E12.5 onwards EPOR was downregulated in radial glial cells. Viable neurons of the TMN continued to express EPO and upregulated EPOR. Our findings provide new evidence that components of the EPO system are present in distinct locations of the embryonic brain and, by interactions between neurons and radial glial cells as well as among clustered TMN neurons, may contribute to its morphogenesis. Whether the observed expression patterns of EPO and EPOR may reflect EPO-mediated trophic and/or antiapoptotic effects on neurons is discussed.


Subject(s)
Erythropoietin/metabolism , Mesencephalon/metabolism , Organogenesis , Receptors, Erythropoietin/metabolism , Animals , DNA-Binding Proteins/biosynthesis , Female , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Mesencephalon/cytology , Mesencephalon/embryology , Mice , Mice, Inbred C57BL , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/biosynthesis , Pregnancy , Transcription Factors/biosynthesis , Up-Regulation
6.
Prague Med Rep ; 105(3): 291-300, 2004.
Article in English | MEDLINE | ID: mdl-15782555

ABSTRACT

The aim of the present study was to determine hypoxia-induced changes in the long-term expression of tyrosine hydroxylase (TH) mRNA and the steady-state dopamine (DA) levels in rat mesencephalic cell cultures. The cultures were exposed to hypoxia during the early developmental period, and DA content and TH mRNA expression were determined on day in vitro (DIV) 14. Hypoxic exposure of 5-day-old cultures resulted in increased DA (control 89.9+/-8.9, hypoxia 135.8+/-23.7 pg/microg protein) and TH mRNA (control 37.3+/-4.7, hypoxia 143.1+/-49.4 pg/microg RNA) levels. To analyze the involvement of hypoxia-inducible factor-1 (HIF-1) in these changes, we studied its activation using reporter gene. Hypoxia caused a 3-fold increase in HIF-1 activity. Our data suggest that hypoxia/ischemia during the putative critical developmental period of neurons may determine the tyrosine hydroxylase gene expression and, consequently, the development of the dopaminergic system.


Subject(s)
Dopamine/metabolism , Hypoxia/metabolism , Mesencephalon/metabolism , RNA, Messenger/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Cells, Cultured , DNA-Binding Proteins/metabolism , Dopamine/genetics , Gene Expression , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Nuclear Proteins/metabolism , Rats , Rats, Wistar , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/genetics
7.
Hear Res ; 183(1-2): 73-83, 2003 Sep.
Article in English | MEDLINE | ID: mdl-13679140

ABSTRACT

Hypoxia/ischemia is a major pathogenetic factor in the development of hearing loss. An important transcription factor involved in the signaling and adaptation to hypoxia/ischemia is the hypoxia-inducible factor-1 (HIF-1). To study HIF-1 expression we used an in vitro hypoxia model of explant and dissociated cultures of the stria vascularis, the organ of Corti with limbus and the modiolus from the cochlea of 3-5-day-old Wistar rats. Hypoxia differentially increased HIF-1 activity as measured by a reporter gene. Twenty-four hour hypoxia increased HIF-1 activity 14.1+/-3.5-fold in the modiolus, 9.4+/-3.0-fold in the organ of Corti with limbus, and 6.4+/-1.5-fold in the stria vascularis. The HIF-1alpha mRNA level was measured by quantitative reverse transcription polymerase chain reaction and showed a lower expression in the modiolus (1.3+/-0.2 pg/microg RNA) than in both the organ of Corti with limbus and the stria vascularis (2.7-3.2+/-1.3, P<0.01). Hypoxia had no effect on the HIF-1alpha mRNA levels. The region-specific regulation of HIF-1 expression on the transcriptional and posttranslational levels may expand the possibilities for adaptation of the cochlea to hypoxia.


Subject(s)
Cochlea/metabolism , DNA-Binding Proteins/biosynthesis , Hearing Loss/metabolism , Hypoxia/metabolism , Nuclear Proteins/biosynthesis , Animals , Cells, Cultured , Cochlea/cytology , DNA-Binding Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression , Genes, Reporter , Hearing Loss/etiology , Histocytochemistry , Hypoxia/complications , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Immunohistochemistry , Luciferases/metabolism , Nuclear Proteins/genetics , Organ Culture Techniques , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/analysis , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection
8.
Eur Arch Psychiatry Clin Neurosci ; 251(4): 170-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11697581

ABSTRACT

Hypoxia, due to impaired cerebral blood flow, has hazardous effects on brain structure and function. Therefore, mechanisms should exist to meet the needs for hypoxic adaptation via regulation of gene expression. Signaling between the O2 sensor and the regulator(s) of transcription is only partially characterized and requires regulatory transcription factors. Among these regulatory proteins, hypoxia-inducible factor-1 (HIF-1) appears to have a key role. HIF-1 modulates gene activity in response to low O2 tensions in the developing and in the adult brain. Moderate hypoxia may elicit autoprotective mechanisms or hypoxia-induced regulators can contribute to mechanisms leading to cell death. Moreover, reactivation of embryonic gene expression may occur after injury-induced hypoxia. Thus, analyses of embryonic and pathogenic models should help to understand how hypoxia-mediated proliferative/cell death processes are involved in brain development and in the pathogenesis of acute or chronic neurodegenerative brain diseases.


Subject(s)
Brain/metabolism , DNA-Binding Proteins/metabolism , Hypoxia/physiopathology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Adaptation, Physiological , Adult , Animals , Apoptosis , Brain/embryology , Brain/physiopathology , Cell Death , Gene Expression Regulation , Gene Expression Regulation, Developmental , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Neurons/metabolism , Signal Transduction
9.
J Biol Chem ; 276(43): 39805-11, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11514583

ABSTRACT

Hypoxic/ischemic conditions provoke activation of the hypoxia-inducible factor-1 (HIF-1), which functions as a transcription factor. HIF-1 is composed of the HIF-1alpha and -beta subunits, and stability regulation occurs via accumulation/degradation of HIF-1alpha with the notion that a prolyl hydroxylase accounts for changes in protein level. In addition, there is evidence that HIF-1 is up-regulated by diverse agonists during normoxia. We investigated the impact of inflammatory mediators nitric oxide (NO) and tumor necrosis factor-alpha (TNF-alpha) on HIF-1alpha regulation. For comparison, LLC-PK(1) cells were exposed to hypoxia, stimulated with desferroxamine (DFX, known to mimic hypoxia), and the thiol-cross-linking agent phenylarsine oxide (PAO). Although all stimuli elicited HIF-1alpha stabilization with differences in the time-dependent accumulation pattern, significant variations appeared with regard to signaling. With the use of a superoxide anion (O(2-)) generator, we established an O(2-)-sensitive pathway that blocked HIF-1alpha stabilization in response to NO and TNF-alpha while DFX- and PAO-evoked HIF-1alpha stabilization appeared O(2-)-insensitive. NO and TNF-alpha signaling required phosphorylation events, especially activation of the phosphatidylinositol 3-kinase/Akt, which is in contrast to DFX and PAO. Based on HIF-1-dependent luciferase reporter gene analysis, it was found that, in contrast to NO and TNF-alpha, PAO resembled a stimulus that induced a dysfunctional HIF-1 complex. These data indicate that diverse agonists activate HIF-1alpha under normoxic conditions by employing different signaling pathways.


Subject(s)
Arsenicals/pharmacology , Deferoxamine/pharmacology , Inflammation Mediators/pharmacology , Nitric Oxide/pharmacology , Protein Serine-Threonine Kinases , Transcription Factors/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Androstadienes/pharmacology , Genistein/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit , Kidney/blood supply , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Oxygen/pharmacology , Phosphorylation , Protein Subunits , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species , Reperfusion Injury , Signal Transduction , Up-Regulation , Wortmannin
10.
Circ Res ; 89(1): 47-54, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11440977

ABSTRACT

The heterodimeric transcription factor hypoxia-inducible factor-1 (HIF-1) is activated under hypoxic conditions, resulting in the upregulation of its target genes plasminogen activator inhibitor-1 (PAI-1) and vascular endothelial growth factor (VEGF). PAI-1 and VEGF are also induced in response to vascular injury, which is characterized by the activation of platelets and the coagulation cascade as well as the generation of reactive oxygen species (ROS). However, it is not known whether HIF-1 is also stimulated by thrombotic factors. We investigated the role of thrombin, platelet-associated growth factors, and ROS derived from the p22(phox)-containing NADPH oxidase in the activation of HIF-1 and the induction of its target genes PAI-1 and VEGF in human vascular smooth muscle cells (VSMCs). Thrombin, platelet-derived growth factor-AB (PDGF-AB), and transforming growth factor-beta(1) (TGF-beta(1)) upregulated HIF-1alpha protein in cultured and native VSMCs. This response was accompanied by nuclear accumulation of HIF-1alpha as well as by increased HIF-1 DNA-binding and reporter gene activity. The thrombin-induced expression of HIF-1alpha, PAI-1, and VEGF was attenuated by antioxidant treatment as well as by transfection of p22(phox) antisense oligonucleotides. Inhibition of p38 mitogen-activated protein kinase and phosphatidylinositol-3-kinase significantly decreased thrombin-induced HIF-1alpha, PAI-1, and VEGF expression. These findings demonstrate that the HIF-1 signaling pathway can be stimulated by thrombin and platelet-associated growth factors and that a redox-sensitive cascade activated by ROS derived from the p22(phox)-containing NADPH oxidase is crucially involved in this response.


Subject(s)
DNA-Binding Proteins/metabolism , Membrane Transport Proteins , Muscle, Smooth, Vascular/metabolism , NADPH Dehydrogenase/physiology , NADPH Oxidases/physiology , Nuclear Proteins/metabolism , Phosphoproteins/physiology , Signal Transduction , Thrombin/pharmacology , Transcription Factors , Antioxidants/pharmacology , Cells, Cultured , DNA-Binding Proteins/physiology , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Lymphokines/biosynthesis , Lymphokines/genetics , Mitogen-Activated Protein Kinases/physiology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Nuclear Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Platelet-Derived Growth Factor/pharmacology , RNA, Messenger/biosynthesis , Reactive Oxygen Species/physiology , Transcriptional Activation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , p38 Mitogen-Activated Protein Kinases
11.
Endocrinology ; 142(6): 2707-18, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11356723

ABSTRACT

The signals oxygen and glucose play an important role in metabolism, angiogenesis, tumorigenesis, and embryonic development. Little is known about an interaction of these two signals. We demonstrate here the cross-talk between oxygen and glucose in the regulation of L-type pyruvate kinase (L-PK) gene expression in the liver. In the liver the periportal to perivenous drop in O(2) tension was proposed to be an endocrine key regulator for the zonated gene expression. In primary rat hepatocyte cultures the expression of the L-PK gene on mRNA and on protein level was induced by venous pO(2), whereas its glucose-dependent induction occurred predominantly under arterial pO(2). It was shown by transient transfection of L-PK promoter luciferase and glucose response element (Glc(PK)RE) SV40 promoter luciferase gene constructs that the modulation by O(2) of the glucose-dependent induction occurred at the Glc(PK)RE in the L-PK gene promoter. The reduction of the glucose-dependent induction of the L-PK gene expression under venous pO(2) appeared to be mediated via an interference between hypoxia inducible factor-1 (HIF-1) and upstream stimulating factor at the Glc(PK)RE. The glucose response element also functioned as an hypoxia response element which was confirmed in cotransfection assays with Glc(PK)RE luciferase gene constructs and HIF-1alpha expression vectors. Furthermore, it was found by gel shift and supershift assay that HIF-1alpha and USF-1 or USF-2 could bind to the Glc(PK)RE. Our findings implicate that the cross-talk between oxygen and glucose might have a fundamental role in the regulation of several physiological and pathophysiological processes.


Subject(s)
Cell Hypoxia , Glucose/pharmacology , Pyruvate Kinase/genetics , Response Elements , Signal Transduction , Transcription Factors , Animals , Arteries , Cells, Cultured , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Gene Expression Regulation/drug effects , Hepatocytes/enzymology , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Immunohistochemistry , Kinetics , Liver/blood supply , Luciferases/genetics , Male , Nuclear Proteins/metabolism , Nuclear Proteins/pharmacology , Oxygen/blood , Oxygen/pharmacology , Promoter Regions, Genetic , Protein Biosynthesis , Pyruvate Kinase/analysis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Simian virus 40/genetics , Transcription, Genetic , Transfection , Veins/enzymology
12.
Blood ; 97(9): 2657-66, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11313255

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) expression is induced by hypoxia (8% O(2)) via the PAI-1 promoter region -175/-159 containing a hypoxia response element (HRE-2) binding the hypoxia-inducible factor-1 (HIF-1) and an adjacent response element (HRE-1) binding a so far unknown factor. The aim of the present study was to identify this factor and to investigate its role in the regulation of PAI-1 expression. It was found by supershift assays that the upstream stimulatory factor-2a (USF-2a) bound mainly to the HRE-1 of the PAI-1 promoter and to a lesser extent to HRE-2. Overexpression of USF-2a inhibited PAI-1 messenger RNA and protein expression and activated L-type pyruvate kinase expression in primary rat hepatocytes under normoxia and hypoxia. Luciferase (Luc) gene constructs driven by 766 and 276 base pairs of the 5'-flanking region of the PAI-1 gene were transfected into primary hepatocytes together with expression vectors encoding wild-type USF-2a and a USF-2a mutant lacking DNA binding and dimerization activity (DeltaHU2a). Cotransfection of the wild-type USF-2a vector reduced Luc activity by about 8-fold, whereas cotransfection of DeltaHU2a did not influence Luc activity. Mutation of the HRE-1 (-175/-168) in the PAI-1 promoter Luc constructs decreased USF-dependent inhibition of Luc activity. Mutation of the HRE-2 (-165/-158) was less effective. Cotransfection of a HIF-1alpha vector could compete for the binding of USF at HRE-2. These results indicated that the balance between 2 transcriptional factors, HIF-1 and USF-2a, which can bind adjacent HRE sites, appears to be involved in the regulation of PAI-1 expression in many clinical conditions.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation , Nuclear Proteins/genetics , Plasminogen Activator Inhibitor 1/genetics , Transcription Factors/genetics , Animals , Cell Hypoxia , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Liver , Male , Promoter Regions, Genetic , Rats , Upstream Stimulatory Factors
13.
Biochem J ; 354(Pt 3): 531-7, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11237857

ABSTRACT

The cDNAs of three hypoxia-inducible factor (HIF) alpha-subunits were cloned from RNA of primary rat hepatocytes by reverse transcriptase PCR. All three cDNAs encoded functionally active proteins, of 825, 874 and 662 amino acids. After transfection they were able to activate luciferase activity of a luciferase gene construct containing three HIF-responsive elements. The mRNAs of the rat HIF alpha-subunits were expressed predominantly in the perivenous zone of rat liver tissue; the nuclear HIFalpha proteins, however, did not appear to be zonated.


Subject(s)
DNA-Binding Proteins/genetics , Liver/metabolism , Nuclear Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Antigens, Polyomavirus Transforming/genetics , Apoptosis Regulatory Proteins , Basic Helix-Loop-Helix Transcription Factors , Cell Nucleus/metabolism , Cells, Cultured , Cloning, Molecular , DNA-Binding Proteins/biosynthesis , Erythropoietin/genetics , Hepatocytes/metabolism , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Liver/blood supply , Luciferases/genetics , Molecular Sequence Data , Nuclear Proteins/biosynthesis , RNA, Messenger/biosynthesis , Rats , Repressor Proteins , Sequence Homology, Amino Acid , Trans-Activators/biosynthesis , Transcription Factors/biosynthesis , Transfection
14.
Eur J Neurosci ; 14(12): 1981-91, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11860493

ABSTRACT

Under severe oxygen deprivation, all cells are able to express the transcription factor HIF-1, which activates a wide range of genes. Under tolerable hypoxia, chemosensory inputs are integrated in brainstem areas, which control cardiorespiratory responses. However, the molecular mechanisms of this functional acclimatization are unknown. We investigated when and where the inducible HIF-1alpha subunit is expressed in the rat brainstem in vivo, under physiological hypoxia. The regional localization of HIF-1alpha mRNA and protein was determined by in situ hybridization and immunocytochemistry in adult male rats exposed to moderate hypoxia (10% O2) for 1-6 h. HIF-1alpha protein was found in cell types identified by immunocytochemistry as catecholaminergic neurons. Hypoxia induced HIF-1alpha mRNA and protein in only some parts of the brainstem located dorsomedially and ventrolaterally, which are those involved in the cardiorespiratory control. No labelling was detected under normoxia. The protein was detected in glia and neurons after 1 and 6 h of hypoxia, respectively. A subset of A2C2 and A1C1 catecholaminergic neurons colocalized tyrosine hydroxylase and HIF-1alpha proteins under hypoxia, but no HIF-1alpha was detected in more rostral catecholaminergic areas. In contrast to cardiorespiratory areas, HIF-1alpha protein was already present under normoxia in glial cells of brainstem tracts but was not overexpressed under hypoxia, although HIF-1alpha mRNA was up-regulated. In conclusion, there appear to be two regulatory mechanisms for HIF-1alpha expression in the brainstem: hypoxic induction of HIF-1alpha protein in cardiorespiratory-related areas and constitutive protein expression unaffected by hypoxia in brainstem tracts.


Subject(s)
Catecholamines/metabolism , Hypoxia/metabolism , Neurons/metabolism , Respiratory Center/metabolism , Transcription Factors/metabolism , Animals , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit , Immunohistochemistry , Male , Neurons/cytology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Respiratory Center/cytology , Transcription Factors/genetics , Tyrosine 3-Monooxygenase/metabolism
15.
Life Sci ; 67(21): 2595-603, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-11104361

ABSTRACT

Perinatal hypoxia is known to induce long-lasting changes in the central dopaminergic system. In order to understand the cellular mechanism of these changes, we studied the effects of hypoxia on the levels of dopamine (DA) and tyrosine hydroxylase (TH) mRNA in untreated and NGF treated PC12 cells. On the second day after plating (DAP), cells were exposed to a hypoxic episode (pO2 = 10-20 mm Hg, 24 h), and the levels of DA and TH mRNA were examined on DAP 4 and DAP 8. In untreated cells, hypoxia induced a two fold increase both in DA and TH mRNA levels on DAP 4 which normalized up to DAP 8. This increase correlated with an activation of the hypoxia inducible factor (HIF-1alpha), measured with a reporter gene. In contrast, NGF treated cells responded to hypoxia with an increase of DA level on DAP 8. In these cells neither an increase of the HIF-1alpha activity measured immediately after hypoxia nor a significant increase of the TH mRNA level on DAP 8 were found. The findings indicate that NGF shifts the hypoxia induced changes of DA levels from a short-term to a long-term mode. The long-term increase of dopamine levels is the most likely result of changes connected with cell growth and differentiation and not the result of a long-term TH mRNA level increase.


Subject(s)
Dopamine/genetics , Hypoxia/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells/drug effects , RNA, Messenger/biosynthesis , Transcription Factors , Tyrosine 3-Monooxygenase/genetics , Animals , Cell Count , Cell Hypoxia , DNA Primers/chemistry , DNA-Binding Proteins/biosynthesis , Dopamine/biosynthesis , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Luminescent Measurements , Nuclear Proteins/biosynthesis , PC12 Cells/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tyrosine 3-Monooxygenase/biosynthesis
16.
Biochem J ; 350 Pt 2: 443-51, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10947958

ABSTRACT

P-Glycoprotein transporters encoded by mdr1 (multidrug resistance) genes mediate extrusion of an array of lipophilic xenobiotics from the cell. In rat liver, mdr transcripts have been shown to be expressed mainly in hepatocytes of the periportal region. Since gradients in oxygen tension (pO(2)) may contribute towards zonated gene expression, the influence of arterial and venous pO(2) on mRNA expression of the mdr1b isoform was examined in primary rat hepatocytes cultured for up to 3 days. Maximal mdr1b mRNA levels (100%) were observed under arterial pO(2) after 72 h, whereas less than half-maximal mRNA levels (40%) were attained under venous pO(2). Accordingly, expression of mdr protein and extrusion of the mdr1 substrate rhodamine 123 were maximal under arterial pO(2) and reduced under venous pO(2). Oxygen-dependent modulation of mdr1b mRNA expression was prevented by actinomycin D, indicating transcriptional regulation. Inhibition of haem synthesis by 25 microM CoCl(2) blocked mdr1b mRNA expression under both oxygen tensions, whereas 80 microM desferrioxamine abolished modulation by O(2). Haem (10 microM) increased mdr1b mRNA levels under arterial and venous pO(2). In hepatocytes treated with 50 microM H(2)O(2), mdr1b mRNA expression was elevated by about 1.6-fold at venous pO(2) and 1.5-fold at arterial pO(2). These results support the conclusion that haem proteins are crucial for modulation of mdr1b mRNA expression by O(2) in hepatocyte cultures and that reactive oxygen species may participate in O(2)-dependent signal transduction. Furthermore, the present study suggests that oxygen might be a critical modulator for zonated secretion of mdr1 substrates into the bile.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Hepatocytes/metabolism , Oxygen/metabolism , Animals , Arteries/metabolism , Blotting, Northern , Cells, Cultured , Chelating Agents/pharmacology , Cobalt/pharmacology , Dactinomycin/pharmacology , Deferoxamine/pharmacology , Fluorescent Dyes/pharmacology , Heme/metabolism , Immunoblotting , Male , Nucleic Acid Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Rhodamine 123/pharmacology , Signal Transduction , Time Factors , Transcription, Genetic , Veins/metabolism , ATP-Binding Cassette Sub-Family B Member 4
17.
Biochem J ; 348 Pt 2: 433-8, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10816439

ABSTRACT

Insulin stimulates glucose utilization in the liver, which occurs mainly in the less aerobic, perivenous, zone. Accordingly, the insulin receptor protein was predominantly expressed in this area, although the insulin receptor mRNA was homogeneously distributed. In hepatocyte cultures venous O(2) partial pressure (pO(2)) induced insulin receptor protein expression. High glucose concentrations enhanced insulin receptor protein under arterial and venous pO(2). The induction of insulin receptor protein by venous pO(2) would explain its zonated expression.


Subject(s)
Gene Expression Regulation/physiology , Glucose/pharmacology , Liver/metabolism , Oxygen/pharmacology , Receptor, Insulin/analysis , Receptor, Insulin/genetics , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Hepatic Veins/cytology , Liver/blood supply , Liver/cytology , Male , Partial Pressure , RNA, Messenger/genetics , Rats , Rats, Wistar , Transcription, Genetic/drug effects
18.
Biochem Biophys Res Commun ; 269(1): 46-53, 2000 Mar 05.
Article in English | MEDLINE | ID: mdl-10694475

ABSTRACT

Since human hepatocytes are available only in limited number, the development of a serum-free culture system for long-term cultivation of differentiated and functional hepatocytes is of great importance. Here we describe the culture of human hepatocytes in a chemically defined serum-free medium for up to 5 weeks. Cell morphology was assayed by light and electron microscopy and revealed a well-preserved cellular morphology. Marker proteins for epithelial and bile duct cells, cytokeratin (CK) 18 and 19, and liver-specific proteins, like phosphoenolpyruvate carboxykinase-2 (PCK2) and serum proteins, were expressed. Liver-enriched transcription factors CCAAT/enhancer binding protein alpha (C/EBPalpha) and hepatocyte nuclear factor-4 (HNF-4), cytokine and mitogen activated factors (nuclear factor kappa B) NFkappaB, and activator protein-1 (AP-1) were maintained and active for several weeks in our cultures. In summary, our serum-free culture system allows the culture of differentiated human hepatocytes for several weeks. It may serve as a model system for metabolic, pharmacologic-toxicologic studies, and studies on human pathogens under defined chemical conditions.


Subject(s)
Liver/cytology , Liver/physiology , Transcription Factors/metabolism , Blood Proteins/genetics , Blood Proteins/metabolism , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Culture Media, Serum-Free , Gene Expression , Humans , Liver/metabolism , Microscopy, Electron , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Transcription Factors/genetics
19.
Mol Pharmacol ; 57(3): 610-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10692503

ABSTRACT

Heme oxygenase (HO) catalyzes the rate-limiting enzymatic step of heme degradation and regulates the cellular heme content. The gene expression of the inducible isoform of HO, HO-1, is up-regulated in response to various agents causing oxidative stress. To investigate the regulatory role of protein phosphatases in the hepatic regulation of HO-1 gene expression, primary cultures of rat hepatocytes were treated with okadaic acid (OA), which specifically inhibits the serine threonine protein phosphatases 1 and 2A. Both protein synthesis and mRNA expression of HO-1 were induced by OA in cultured hepatocytes, but not in cultured tissue macrophages of rat liver. The HO-1 mRNA induction by OA occurred in a time- and concentration-dependent manner. Simultaneous treatment with OA plus dibutyryl cAMP caused a synergistic up-regulation of steady-state levels of HO-1 mRNA, and the specific protein kinase A inhibitor KT5720 markedly reduced the OA-dependent HO-1 mRNA induction. In contrast, the dibutyryl cAMP-dependent induction of the phosphoenolpyruvate carboxykinase mRNA expression and enzyme activity was inhibited by simultaneous treatment with OA in hepatocytes. The induction of the HO-1 gene expression by OA was transcriptional as determined by studies with actinomycin D, nuclear run-off assay, and measurement of the half-life of HO-1 mRNA. Luciferase reporter constructs containing DNA sequences of the rat HO-1 promoter 5'-flanking region were up-regulated by OA in transiently transfected hepatocytes. Mutation of the cAMP response element/activator protein-1 (-665/-654) site obliterated the OA-dependent induction, suggesting that this element is involved in the transcriptional induction of the rat HO-1 gene by OA.


Subject(s)
Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase (Decyclizing)/genetics , Liver/drug effects , Okadaic Acid/pharmacology , Animals , Cyclic AMP/metabolism , Enzyme Induction , Heme Oxygenase (Decyclizing)/biosynthesis , Heme Oxygenase-1 , In Vitro Techniques , Liver/cytology , Liver/enzymology , Male , Promoter Regions, Genetic , Protein Kinase C/biosynthesis , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Transcriptional Activation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...