Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Cereb Blood Flow Metab ; 19(6): 643-51, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10366194

RESUMO

The present study describes, for the first time, a temporal and spatial cellular expression of erythropoietin (Epo) and Epo receptor (Epo-R) with the evolution of a cerebral infarct after focal permanent ischemia in mice. In addition to a basal expression of Epo in neurons and astrocytes, a postischemic Epo expression has been localized specifically to endothelial cells (1 day), microglia/macrophage-like cells (3 days), and reactive astrocytes (7 days after occlusion). Under these conditions, the Epo-R expression always precedes that of Epo for each cell type. These results support the hypothesis that there is a continuous formation of Epo, with its corresponding receptor, during the active evolution of a focal cerebral infarct and that the Epo/Epo-R system might be implicated in the processes of neuroprotection and restructuring (such as angiogenesis and gliosis) after ischemia. To support this hypothesis, a significant reduction in infarct volume (47%; P < 0.0002) was found in mice treated with recombinant Epo 24 hours before induction of cerebral ischemia. Based on the above, we propose that the Epo/Epo-R system is an endogenous mechanism that protects the brain against damages consequent to a reduction in blood flow, a mechanism that can be amplified by the intracerebroventricular application of exogenous recombinant Epo.


Assuntos
Isquemia Encefálica/metabolismo , Eritropoetina/biossíntese , Eritropoetina/farmacologia , Receptores da Eritropoetina/biossíntese , Animais , Astrócitos/efeitos dos fármacos , Western Blotting , Encéfalo/citologia , Química Encefálica/fisiologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/metabolismo , Infarto Cerebral/patologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Eritropoetina/administração & dosagem , Imuno-Histoquímica , Hibridização In Situ , Injeções Intraventriculares , Camundongos , Neurônios/efeitos dos fármacos
2.
Thromb Haemost ; 82 Suppl 1: 44-52, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10695485

RESUMO

Angiogenic growth factors and their endothelial receptors function as major regulators of blood vessel formation. The VEGF/VEGFR and the Angiopoietin/Tie2 receptor systems represent key signal transduction pathways involved in the regulation of embryonic vascular development. Inactivation of any of the genes encoding these molecules results in defective vascular development and lethality between embryonic day 8.5 and 12.5. In addition, VEGF and its receptors are also critically involved in the regulation of pathological blood vessel growth in the adult during various angiogenesis-dependent diseases that are associated with tissue hypoxia, such as solid tumor growth and ischemic diseases. It is now well established that therapeutic angiogenesis can be achieved in animal models of hind limb and myocardial ischemia by exogenously adding VEGF and/or other angiogenic growth factors. Available clinical data from human trials also suggests that patients with severe cardiovascular diseases could potentially benefit from such therapies. However, much more work needs to be done to compare the potency of different angiogenic factors or the combination thereof, as well as the best way of delivery, either as recombinant proteins, as naked DNA or via adenoviral vectors. Nevertheless, the therapeutic efficacy of simply injecting naked plasmid DNA or proteins into ischemic tissue to deliver secreted angiogenic factors is an encouraging finding. Time will show whether the adverse side effects of therapeutic angiogenesis, mainly vascular permeability and edema formation, can be minimized and angiogenic factors can be used as an effective therapy in patients for the treatment of ischemic diseases such as arterial occlusive disease, myocardial infarction, and, eventually, also stroke.


Assuntos
Sistema Cardiovascular/fisiopatologia , Isquemia Miocárdica/fisiopatologia , Neovascularização Patológica , Adulto , Animais , Humanos
3.
Placenta ; 24(10): 941-50, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14580376

RESUMO

The transforming growth factor-beta 3 (TGF-beta 3) is involved in oxygen-dependent differentiation processes during placental development and pregnancy disorders. However, the importance of oxygen partial pressure for the regulation of TGF-beta 3 expression is presently unclear. We and others presented preliminary evidence that the hypoxia-inducible factor-1 (HIF-1) confers TGF-beta 3 transcription but it was unknown whether this occurred directly or indirectly. To analyze how HIF-1 regulates TGF-beta 3 gene transcription, we cloned and sequenced the mouse TGF-beta 3 promoter region. Multiple putative HIF-1 binding sites (HBSs) were identified, many of which co-localized with two G+C rich CpG islands 5' to the TGF-beta 3 transcription start site. A 6.8 kb fragment of the TGF-beta 3 promoter induced reporter gene expression under hypoxic conditions or when treated with an iron chelator known to stabilize and activate the HIF-1 alpha subunit. Deletion of a 2.4 kb fragment upstream of the distal CpG island abolished inducibility of reporter gene expression. Two HBSs (HBS1 and HBS6) that bound the HIF-1 protein could be identified within this 2.4 kb fragment. These results suggest that TGF-beta 3 gene expression is directly regulated by HIF-1.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas Nucleares/fisiologia , Fatores de Transcrição , Ativação Transcricional , Fator de Crescimento Transformador beta/genética , Animais , Sítios de Ligação , Diferenciação Celular , Hipóxia Celular , DNA/metabolismo , Feminino , Regulação da Expressão Gênica , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Camundongos , Gravidez , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Fator de Crescimento Transformador beta3 , Trofoblastos/citologia
4.
Int J Mol Med ; 2(3): 317-24, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9855704

RESUMO

Hypoxia is thought to be a common precursor of coronary artery disease and malignant tumors, both diseases representing the leading causes of death in industrial nations. So far, investigations of oxygen-regulated erythropoietin (EPO) gene expression in the human hepatoma cell lines Hep3B and HepG2 allowed many important insights into the mechanisms of oxygen-sensing, signalling and regulation of an increasing number of oxygen-responsive genes. To differentiate the various signalling pathways involved in EPO production by these two cell lines, we examined several factors that positively influenced EPO expression. The results demonstrate a keen differential effect of cell density and oxygen concentration on EPO induction in Hep3B compared to HepG2 cells. Using optimized cell culture conditions, EPO production rates as high as 1 U EPO per 10(6) Hep3B cells in 24 h could be achieved. We also found a moderate but reproducible positive effect of CoCl2 on hypoxia-induced EPO expression in Hep3B but a negative CoCl2 effect on hypoxic induction in HepG2 cells. CoCl2 inhibited cell growth in a concentration-dependent manner. Interleukin-6 was synergistic with hypoxia on EPO induction in Hep3B as well as HepG2 cells, and dexamethasone enhanced this effect in Hep3B but not in HepG2 cells. The moderate CoCl2-dependent increase of EPO production observed in hypoxic Hep3B cells might indicate that CoCl2 and hypoxia do not necessarily act via, identical signalling pathways.


Assuntos
Eritropoetina/biossíntese , Carcinoma Hepatocelular , Contagem de Células , Hipóxia Celular , Cobalto/farmacologia , Dexametasona/farmacologia , Humanos , Interleucina-6/farmacologia , Neoplasias Hepáticas , Oxigênio/metabolismo , Transdução de Sinais
5.
Proc Natl Acad Sci U S A ; 95(26): 15809-14, 1998 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-9861052

RESUMO

Vascular endothelial growth factor (VEGF) plays a key role in physiological blood vessel formation and pathological angiogenesis such as tumor growth and ischemic diseases. Hypoxia is a potent inducer of VEGF in vitro. Here we demonstrate that VEGF is induced in vivo by exposing mice to systemic hypoxia. VEGF induction was highest in brain, but also occurred in kidney, testis, lung, heart, and liver. In situ hybridization analysis revealed that a distinct subset of cells within a given organ, such as glial cells and neurons in brain, tubular cells in kidney, and Sertoli cells in testis, responded to the hypoxic stimulus with an increase in VEGF expression. Surprisingly, however, other cells at sites of constitutive VEGF expression in normal adult tissues, such as epithelial cells in the choroid plexus and kidney glomeruli, decreased VEGF expression in response to the hypoxic stimulus. Furthermore, in addition to VEGF itself, expression of VEGF receptor-1 (VEGFR-1), but not VEGFR-2, was induced by hypoxia in endothelial cells of lung, heart, brain, kidney, and liver. VEGF itself was never found to be up-regulated in endothelial cells under hypoxic conditions, consistent with its paracrine action during normoxia. Our results show that the response to hypoxia in vivo is differentially regulated at the level of specific cell types or layers in certain organs. In these tissues, up- or down-regulation of VEGF and VEGFR-1 during hypoxia may influence their oxygenation after angiogenesis or modulate vascular permeability.


Assuntos
Fatores de Crescimento Endotelial/genética , Regulação da Expressão Gênica , Hipóxia/fisiopatologia , Linfocinas/genética , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento/genética , Animais , Encéfalo/metabolismo , Plexo Corióideo/metabolismo , Fatores de Crescimento Endotelial/biossíntese , Hibridização In Situ , Glomérulos Renais/metabolismo , Túbulos Renais/metabolismo , Linfocinas/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Especificidade de Órgãos , Receptores Proteína Tirosina Quinases/biossíntese , Receptores de Fatores de Crescimento/biossíntese , Receptores de Fatores de Crescimento do Endotélio Vascular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Sertoli/metabolismo , Testículo/metabolismo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
6.
Blood ; 92(9): 3388-93, 1998 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9787178

RESUMO

Hemangioblastomas are highly vascular tumors of the central nervous system that overexpress the hypoxia-inducible gene, vascular endothelial growth factor (VEGF), as a consequence of mutational inactivation of the von Hippel-Lindau tumor suppressor gene (VHL). Previous reports showed that hemangioblastomas can also express erythropoietin (Epo), which is also hypoxia-inducible. However, Epo expression in hemangioblastomas was observed only in individual cases, and the analyses were mainly based on indirect determination of erythropoiesis-stimulating activity. Therefore, we analyzed a series of 11 hemangioblastomas for Epo, VEGF, and VHL expression by Northern blot analysis and compared the results with normal brain and glioblastomas. Surprisingly, we observed Epo mRNA expression in all hemangioblastoma specimens analyzed, but in none of four glioblastomas. In contrast, VEGF mRNA was expressed in all hemangioblastomas and all glioblastomas. In situ hybridization revealed neoplastic stromal cells as Epo- and VEGF-producing cells in hemangioblastomas. These results suggest that in the nonhypoxic microenvironment of hemangioblastoma, Epo, similar to VEGF, might be negatively regulated by the VHL gene product.


Assuntos
Neoplasias do Sistema Nervoso Central/genética , Fatores de Crescimento Endotelial/biossíntese , Eritropoetina/biossíntese , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Hemangioblastoma/genética , Hormônios Ectópicos/biossíntese , Ligases , Linfocinas/biossíntese , Proteínas de Neoplasias/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Biossíntese de Proteínas , Proteínas Supressoras de Tumor , Ubiquitina-Proteína Ligases , Doença de von Hippel-Lindau/genética , Adulto , Idoso , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Hipóxia Celular , Neoplasias do Sistema Nervoso Central/metabolismo , Neoplasias do Sistema Nervoso Central/patologia , Fatores de Crescimento Endotelial/genética , Eritropoese , Eritropoetina/genética , Feminino , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Hemangioblastoma/metabolismo , Hemangioblastoma/patologia , Hormônios Ectópicos/genética , Humanos , Hibridização In Situ , Linfocinas/genética , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Proteínas do Tecido Nervoso/genética , Proteínas/genética , Proteínas/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Células Estromais/metabolismo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Proteína Supressora de Tumor Von Hippel-Lindau
7.
Microvasc Res ; 63(1): 70-80, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11749074

RESUMO

In vivo, hypoxia is known to damage the blood-brain barrier (BBB) leading to the development of vasogenic brain edema. Primary cultures of porcine brain derived microvascular endothelial cells were used as an in vitro BBB model to evaluate the mechanisms by which hypoxia regulates paracellular permeability. Paracellular passage across endothelial cell monolayers is regulated by specialized intercellular structures like the tight junctions (TJ). Zonula occludens-1 (ZO-1), a protein of the TJ, lines the cytoplasmic face of intact TJ. The continuity of the ZO-1 expression was disrupted during 24 h of hypoxia which correlated with a decrease of the protein level to 32 +/- 8% and with a twofold increase in the phosphorylation of ZO-1 in comparison to values determined at the start of the experiment. The localization and expression level of ZO-1 were maintained during hypoxia in the presence of a polyclonal antibody to vascular endothelial growth factor (VEGF) demonstrating that hypoxia-induced changes of the ZO-1 expression are mediated by VEGF. The effect of hypoxia on the ZO-1 distribution probably is not tissue- or cell-specific because similar changes of ZO-1 distribution were observed when the rat brain endothelial cell line RBE4 or the murine epithelial cell line CSG was used. Furthermore, ZO-1 changes correlated with small changes in actin distribution. These results suggest that hypoxia increases the paracellular flux across the cell monolayer via the release of VEGF, which in turn leads to the dislocalization, decreased expression, and enhanced phosphorylation of ZO-1. Science.


Assuntos
Encéfalo/irrigação sanguínea , Fatores de Crescimento Endotelial/metabolismo , Endotélio Vascular/metabolismo , Hipóxia , Linfocinas/metabolismo , Proteínas de Membrana/biossíntese , Microcirculação/metabolismo , Fosfoproteínas/biossíntese , Actinas/biossíntese , Animais , Barreira Hematoencefálica , Western Blotting , Encéfalo/metabolismo , Linhagem Celular , Células Cultivadas , Citoplasma/metabolismo , Endotélio Vascular/citologia , Imuno-Histoquímica , Camundongos , Fosforilação , Testes de Precipitina , Ratos , Suínos , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Proteína da Zônula de Oclusão-1
8.
Kidney Int ; 51(2): 560-3, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9027739

RESUMO

The hypoxia-inducible factor-1 (HIF-1) is involved in the induction of oxygen regulated genes such as erythropoietin and vascular endothelial growth factor (VEGF). HIF-1 is a heterodimeric transcription factor consisting of an alpha and a beta subunit. The question of how HIF-1 itself is regulated remains to be elucidated. Studies performed in human Hep3B hepatoma cells suggested that the prevalent mode of HIF-1 action is an increase in HIF-1 alpha steady-state mRNA and protein levels after hypoxic exposure. In contrast to the reported very low basal HIF-1 alpha mRNA levels, however, we detected HIF-1 alpha mRNA in several cell lines cultured under normoxic conditions, although no HIF-1 DNA binding activity was observed. Following hypoxic induction, VEGF mRNA levels and HIF-1 DNA binding activity increased, but HIF-1 alpha mRNA levels remained largely unchanged. One possible explanation for this discrepancy might be that HIF-1 DNA binding activity does not follow HIF-1 alpha mRNA kinetics. We therefore incubated HeLaS3 cells in tonometers for 7.5 minutes up to four hours at either 20% O2 or 0.5% O2. Although there was some variation in HIF-1 alpha mRNA levels, we did not find significant changes over this time frame, suggesting that HIF-1 alpha is not transcriptionally regulated.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , RNA Mensageiro/genética , Fatores de Transcrição , Animais , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Linhagem Celular , DNA/metabolismo , Proteínas de Ligação a DNA/química , Fatores de Crescimento Endotelial/genética , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Cinética , Linfocinas/genética , Camundongos , Proteínas Nucleares/química , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
9.
Kidney Int ; 51(2): 564-6, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9027740

RESUMO

The hypoxia-inducible factor-1 (HIF-1) was first described as a DNA binding activity that specifically recognizes an 8 bp hypoxia response element (HRE) known to be essential for oxygen-regulated erythropoietin gene expression. In electrophoretic mobility shift assays (EMSAs) HIF-1 DNA binding activity is only detectable in nuclear extracts of cells cultivated in a low oxygen atmosphere. In addition to HIF-1, a constitutive DNA binding activity also specifically binds the HIF-1 probe. Based on EMSAs using competitor oligonucleotides, specific antibodies and recombinant proteins, we previously reported that the constitutive HRE binding factor is composed of ATF-1 and CREB-1. Here we show that this site is functionally responsive to the cAMP agonist 8Br-cAMP in a dose-dependent manner under hypoxic but not under normoxic conditions. These results were confirmed by using the protein kinase A (PKA) activator Sp-cAMPS and the PKA inhibitor Rp-cAMPS: while Sp-cAMPS was synergistic with hypoxia on the HIF-1 DNA recognition site, the Rp-cAMPS isomer showed no effect. Our findings suggest that the PKA-signaling pathway is enhancing oxygen-dependent gene expression via the HRE.


Assuntos
AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Sítios de Ligação , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , DNA/genética , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Eritropoetina/genética , Regulação da Expressão Gênica , Genes Reporter , Células HeLa , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Luciferases/genética , Transfecção
10.
FASEB J ; 13(11): 1315-24, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10428756

RESUMO

The tissue type plasminogen activator (t-PA) is a serine protease that is involved in neuronal plasticity and cell death induced by excitotoxins and ischemia in the brain. t-PA activity in the central nervous system is regulated through the activation of serine protease inhibitors (serpins) such as the plasminogen activator inhibitor (PAI-1), the protease nexin-1 (PN-1), and neuroserpin (NSP). Recently we demonstrated in vitro that PAI-1 produced by astrocytes mediates the neuroprotective effect of the transforming growth factor-beta1 (TGF-beta1) in NMDA-induced neuronal cell death. To investigate whether serpins may be involved in neuronal cell death after cerebral ischemia, we determined, by using semiquantitative RT-PCR and in situ hybridization, that focal cerebral ischemia in mice induced a dramatic overexpression of PAI-1 without any effect on PN-1, NSP, or t-PA. Then we showed that although the expression of PAI-1 is restricted to astrocytes, PN-1, NSP, and t-PA are expressed in both neurons and astrocytes. Moreover, by using semiquantitative RT-PCR and Western blotting, we observed that only the expression of PAI-1 was modulated by TGF-beta1 treatment via a TGF-beta-inducible element contained in the PAI-1 promoter (CAGA box). Finally, we compared the specificity of TGF-beta1 action with other members of the TGF-beta family by using luciferase reporter genes. These data show that TGF-beta and activin were able to induce the overexpression of PAI-1 in astrocytes, but that bone morphogenetic proteins, glial cell line-derived neutrophic factor, and neurturin did not. These results provide new insights into the regulation of the serpins/t-PA axis and the mechanism by which TGF-beta may be neuroprotective.


Assuntos
Isquemia Encefálica/metabolismo , Proteínas de Transporte/metabolismo , Neuropeptídeos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Serpinas/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Precursor de Proteína beta-Amiloide , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Proteínas de Transporte/genética , Morte Celular/genética , Células Cultivadas , Regulação da Expressão Gênica , Camundongos , Neurônios/patologia , Neuropeptídeos/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Nexinas de Proteases , Receptores de Superfície Celular , Serpinas/genética , Ativador de Plasminogênio Tecidual/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia , Neuroserpina
11.
Pflugers Arch ; 429(2): 216-22, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7892107

RESUMO

O2 plays a dominant role in the metabolism and viability of cells; changes in O2 supply lead to many physiological responses in the cell. Recent reports have shown that hypoxia induces the transcription of a number of genes, among them those for the glycolytic enzymes. We have investigated signalling events that may lead to enhanced activity of lactate dehydrogenase (LDH) in cultured vascular smooth muscle (VSM) cells derived from rat aorta, grown under hypoxic conditions (1% versus 20% O2). LDH was chosen because this enzyme exhibits one of the largest increases in activity among the glycolytic enzymes after hypoxic stimulation of cells. Hypoxic exposure of VSM cells for 24 h resulted in a 2-fold increase in LDH activity and in a 2.5-fold increase in intracellular cAMP levels. Agents that activate adenylate cyclase, such as forskolin, cholera toxin and 1-methyl-3-isobutylxanthine (IBMX), and thus increase cAMP production, significantly induced LDH activity. Moreover, induction of LDH activity by hypoxia was prevented in the presence of the protein kinase A inhibitor N-[2-(methyl-amino)ethyl]-5-isoquinolinsulphonamide dihydrochloride (H-8), and the cyclooxygenase inhibitor indomethacin. In contrast to the cAMP-stimulating agents, stable cGMP analogues (dibutyryl-cGMP, 8-bromo-cGMP), activators of protein kinase C [12-O-tetradecanoylphorbol-13-acetate (TPA), and 1-oleoyl-2-acetyl-glycerol (OAG), and the calcium ionophore ionomycin did not alter LDH activity in VSM cells kept at 20% O2. A dose-dependent increase in LDH activity was also observed in normoxic cells exposed to cobalt chloride (50-200 microM), indicating that a metal binding protein might be involved in this signalling cascade.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Hipóxia Celular/fisiologia , Cobalto/farmacologia , AMP Cíclico/metabolismo , L-Lactato Desidrogenase/metabolismo , Músculo Liso Vascular/enzimologia , Animais , Desferroxamina/farmacologia , Relação Dose-Resposta a Droga , Masculino , Músculo Liso Vascular/metabolismo , Ratos , Ratos Sprague-Dawley
12.
Biochem Biophys Res Commun ; 223(1): 54-9, 1996 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-8660378

RESUMO

The heterodimeric hypoxia-inducible transcription factor HIF-1 is involved in the oxygen-regulated transcription of several genes including erythropoietin. Cloning and sequencing of the alpha-subunit of mouse HIF-1 cDNA revealed a 90% overall homology to human HIF-l alpha but lack of any similarity in the 5' untranslated region and translational start site. Mouse HIF-1 alpha is encoded by an evolutionary conserved single-copy gene located on chromosome 12. We found a widespread constitutive expression of mouse HIf-1 alpha mRNA which was particularly high in lung and kidney. Despite a strong erythropoietin induction, HIF-1 alpha mRNA concentrations were not upregulated in hypoxic mouse tissues.


Assuntos
Evolução Biológica , Mapeamento Cromossômico , Proteínas de Ligação a DNA/biossíntese , Proteínas Nucleares/biossíntese , Transcrição Gênica , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Sequência Conservada , Primers do DNA , DNA Complementar , Proteínas de Ligação a DNA/química , Células HeLa , Humanos , Hipóxia , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Rim/metabolismo , Neoplasias Hepáticas Experimentais , Pulmão/metabolismo , Substâncias Macromoleculares , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/química , Reação em Cadeia da Polimerase , RNA Mensageiro/biossíntese , Homologia de Sequência do Ácido Nucleico , Fatores de Transcrição/biossíntese , Células Tumorais Cultivadas
13.
J Biol Chem ; 270(46): 27865-70, 1995 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-7499259

RESUMO

Transcriptional regulation of gene expression by hypoxia is an important, but yet only marginally characterized mechanism by which organisms adapt to low oxygen concentrations. The human hepatoma cell line HepG2 is a widely used model for studying hypoxic induction of the hematopoietic growth factor erythropoietin. In an attempt to identify additional genes expressed in HepG2 cells during hypoxia, we differentially screened a cDNA library derived from hypoxic (1% O2) HepG2 cells using probes isolated from either normoxic (21% O2) or hypoxic cells. Two genes were identified, one encoding aldolase, a member of the glycolytic enzymes, and the other encoding alpha 1-antitrypsin which belongs to the family of the acute phase (AP) responsive proteins. Whereas hypoxic induction of glycolytic enzymes is well established, oxygen-dependent regulation of AP genes has not been reported so far. AP proteins are liver-derived plasma proteins whose production during inflammation is either up-regulated (positive AP reactants) or down-regulated (negative AP reactants). In the present study, we demonstrate that on the mRNA level hypoxic stimulation of HepG2 cells led to (i) an induction of the positive AP reactants alpha 1-antitrypsin, alpha 1-antichymotrypsin, complement C3, haptoglobin, and alpha 1-acid glycoprotein; (ii) a down-regulation of the negative AP reactant albumin; (iii) an up-regulation of the negative AP reactant transferrin; and (iv) unchanged levels of the positive AP reactants alpha- and beta-fibrinogen as well as hemopexin. Cycloheximide inhibited hypoxic up-regulation of AP mRNAs demonstrating that de novo protein synthesis is required for hypoxic induction. Nuclear run-on assays indicate that the hypoxic increase in AP mRNAs is mainly due to transcriptional regulation. The hypoxic response was compared to AP stimulation by interleukin 6. The results suggest that the adaptive response to hypoxia overlaps with, but is not identical with, the AP response mediated by interleukin 6.


Assuntos
Proteínas de Fase Aguda/biossíntese , Hipóxia Celular , Expressão Gênica , Animais , Sequência de Bases , Northern Blotting , Carcinoma Hepatocelular , Linhagem Celular , Cicloeximida/farmacologia , Primers do DNA , DNA Complementar , Dexametasona/farmacologia , Fatores de Crescimento Endotelial/biossíntese , Eritropoetina/biossíntese , Frutose-Bifosfato Aldolase/biossíntese , Frutose-Bifosfato Aldolase/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Biblioteca Gênica , Glicólise , Humanos , Interleucina-6/farmacologia , Neoplasias Hepáticas , Linfocinas/biossíntese , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Transcrição Gênica , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , alfa 1-Antitripsina/biossíntese , alfa 1-Antitripsina/genética
14.
Nucleic Acids Res ; 23(22): 4542-50, 1995 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-8524640

RESUMO

The hypoxia-inducible factor-1 (HIF-1) was first described as a DNA binding activity that specifically recognizes an 8 bp motif known to be essential for hypoxia-inducible erythropoietin gene transcription. Subsequently HIF-1 activity has also been found in cell lines which do not express erythropoietin, suggesting that HIF-1 is part of a widespread oxygen sensing mechanism. In electrophoretic mobility shift assays HIF-1 DNA binding activity is only detectable in nuclear extracts of cells cultivated in a low oxygen atmosphere. In addition to HIF-1, a constitutive DNA binding activity also specifically binds the HIF1 probe. Here we report that CRE and AP1 oligonucleotides efficiently competed for binding of the HIF1 probe to this constitutive factor, whereas HIF-1 activity itself remained unaffected. Monoclonal antibodies raised against the CRE binding factors ATF-1 and CREB-1 supershifted the constitutive factors ATF-1 and CREB-1 supershifted the constitutive factor, while Jun and Fos family members, which constitute the AP-1 factor, were immunologically undetectable. Recombinant ATF-1 and CREB-1 proteins bound HIF1 probes either as homodimers or as heterodimers, indicating a new binding specificity for ATF-1/CREB-1. Finally, reporter gene assays in HeLa cells treated with either a cAMP analogue or a phorbol ester suggest that the PKA, but not the PKC signalling pathway is involved in oxygen sensing.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , DNA/química , DNA/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Fator 1 Ativador da Transcrição , Fator 2 Ativador da Transcrição , Animais , Sequência de Bases , Sítios de Ligação , Núcleo Celular/metabolismo , Sequência Consenso , Células HeLa , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Células L , Zíper de Leucina , Luciferases/análise , Luciferases/biossíntese , Camundongos , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Homologia de Sequência do Ácido Nucleico , Fatores de Transcrição/isolamento & purificação , Transfecção
15.
Am J Physiol ; 271(4 Pt 1): C1172-80, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8897823

RESUMO

Hypoxia-inducible factor 1 (HIF-1) is a heterodimeric basic helix-loop-helix protein implicated in the transcriptional activation of genes encoding erythropoietin, glycolytic enzymes, and vascular endothelial growth factor in hypoxic mammalian cells. In this study, we have quantitated HIF-1 DNA-binding activity and protein levels of the HIF-1 alpha and HIF-1 beta subunits in human HeLa cells exposed to O2 concentrations ranging from 0 to 20% in the absence or presence of 1 mM KCN to inhibit oxidative phosphorylation and cellular O2 consumption. HIF-1 DNA-binding activity, HIF-1 alpha protein and HIF-1 beta protein each increased exponentially as cells were subjected to decreasing O2 concentrations, with a half maximal response between 1.5 and 2% O2 and a maximal response at 0.5% O2, both in the presence and absence of KCN. The HIF-1 response was greatest over O2 concentrations associated with ischemic/hypoxic events in vivo. These results provide evidence for the involvement of HIF-1 in O2 homeostasis and represent a functional characterization of the putative O2 sensor that initiates hypoxia signal transduction leading to HIF-1 expression.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Oxigênio/metabolismo , Fatores de Transcrição/metabolismo , Western Blotting , Núcleo Celular/metabolismo , Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Cianeto de Potássio/farmacologia
16.
Glia ; 30(3): 271-8, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10756076

RESUMO

Erythropoietin (Epo), the major hormone controlling the hypoxia-induced increase in the number of erythrocytes, has also a functional role in the brain. However, few data exist as to the cellular source of brain-derived Epo as well as to the molecular mechanisms that control Epo expression in the central nervous system. Using patch-clamp and RT-PCR methods, we provide direct evidence that, besides astrocytes, neurons are a source of Epo in the brain. Both the astrocytic and neuronal expression of Epo mRNA are induced not only by hypoxia, but also by desferrioxamine (DFX) and cobalt chloride (CoCl(2)), two agents known to mimic the hypoxic induction of Epo in hepatoma cells. This induction is blocked by cycloheximide suggesting that de novo protein synthesis is required. Furthermore, the addition of H(2)O(2) decreases the hypoxia-induced Epo mRNA levels. These data indicate that, following hypoxia, a common oxygen sensing and signaling pathway leads to increased Epo gene expression in both nervous and hepatoma cells; this pathway would be dependent on the redox-state of the brain. Furthermore, we show that the in vivo administration of CoCl(2) and DFX to mice induces an increased Epo mRNA level in the neocortex. As Epo protects the brain against ischemia, our in vivo experiments suggest that the use of molecules such as CoCl(2) or DFX, that provoke an increased Epo gene expression in the brain, could be useful in the development of potential therapeutic strategies for the treatment of hypoxic or ischemic brain injury.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Eritropoetina/genética , Hipóxia/metabolismo , Neurônios/metabolismo , Oxirredução , Oxigênio/metabolismo , RNA Mensageiro/metabolismo , Animais , Células Cultivadas , Hipóxia/genética , Masculino , Camundongos , Camundongos Endogâmicos , Reação em Cadeia da Polimerase , Fatores de Tempo
17.
Proc Natl Acad Sci U S A ; 93(7): 2867-72, 1996 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-8610133

RESUMO

Blastocyst-derived pluripotent mouse embryonic stem cells can differentiate in vitro to form so-called embryoid bodies (EBs), which recapitulate several aspects of murine embryogenesis. We used this in vitro model to study oxygen supply and consumption as well as the response to reduced oxygenation during the earliest stages of development. EBs were found to grow equally well when cultured at 20% (normoxia) or 1% (hypoxia) oxygen during the first 5 days of differentiation. Microelectrode measurements of pericellular oxygen tension within 13- to 14-day-old EBs (diameter 510-890 micron) done at 20% oxygen revealed efficient oxygenation of the EBs' core region. Confocal laser scanning microscopy analysis of EBs incubated with fluorescent dyes that specifically stain living cells confirmed that the cells within an EB were viable. To determine the EBs' capability to sense low oxygen tension and to specifically respond to low ambient oxygen by modulating gene expression we quantified aldolase A and vascular endothelial growth factor (VEGF) mRNAs, since expression of these genes is upregulated by hypoxia in a variety of cells. Compared with the normoxic controls, we found increased aldolase A and VEGF mRNA levels after exposing 8- to 9-day-old EBs to 1% oxygen. We propose that EBs represent a powerful tool to study oxygen-regulated gene expression during the early steps of embryogenesis, where the preimplantation conceptus resides in a fluid environment with low oxygen tension until implantation and vascularization allow efficient oxygenation.


Assuntos
Blastocisto/fisiologia , Diferenciação Celular , Regulação da Expressão Gênica , Consumo de Oxigênio , Células-Tronco/fisiologia , Aerobiose , Animais , Sequência de Bases , Blastocisto/citologia , Hipóxia Celular , Células Cultivadas , Primers do DNA , Fatores de Crescimento Endotelial/biossíntese , Corantes Fluorescentes , Frutose-Bifosfato Aldolase/biossíntese , Isoenzimas/biossíntese , Cinética , Linfocinas/biossíntese , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Células-Tronco/citologia , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
18.
Pediatr Res ; 43(3): 349-54, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9505273

RESUMO

The fetal liver is the main hematopoietic organ during intrauterine life. Morphometrical studies were performed on liver sections to detect changes occurring with intrauterine growth retardation and preeclampsia. Compared with the controls (n = 10), fetuses from preeclamptic mothers showed a severe reduction of erythroid cells by 60% on average (n = 18). Closer examination revealed that the erythroid cells at early stages of differentiation were more affected (80% reduction) than at later stages (55%). Seven out of 18 fetuses from preeclamptic mothers did not show growth retardation but exhibited severely reduced hepatic erythropoiesis. We suggest that the prime factor for impaired red blood cell production is preeclampsia itself rather than intrauterine growth retardation. Regulation of erythropoiesis in utero might depend on the interaction of many hematopoietic growth factors, and preeclampsia might alter the balance. To test this notion, we quantitated erythropoietin in fetal blood and various cytokines in the amniotic fluid. An elevation of erythropoietin and interleukin (IL)-3 levels was seen in babies born under the conditions of preeclampsia, whereas the concentrations of granulocyte/macrophage-colony-stimulating factor (CSF), granulocyte-CSF, and IL-1 beta were reduced, and the levels of IL-6 and IL-8 remained constant. With preeclampsia, a discrepancy between elevation of erythrocyte numbers in peripheral blood and depression of hematopoiesis at the main production site, the fetal liver, is seen. Concomitantly, there is elevation of some but reduction of other hematopoietic cytokines. We envision that during the course of preeclampsia quantitation of hematopoietic growth factors might allow to predict the deterioration of in utero life conditions.


Assuntos
Eritropoese , Feto/patologia , Fígado/patologia , Pré-Eclâmpsia/patologia , Líquido Amniótico/metabolismo , Estudos de Casos e Controles , Citocinas/metabolismo , Eritropoese/fisiologia , Eritropoetina/sangue , Feminino , Sangue Fetal/metabolismo , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/patologia , Retardo do Crescimento Fetal/fisiopatologia , Feto/fisiopatologia , Fatores de Crescimento de Células Hematopoéticas/metabolismo , Humanos , Fígado/fisiopatologia , Troca Materno-Fetal , Pré-Eclâmpsia/complicações , Pré-Eclâmpsia/fisiopatologia , Gravidez
19.
Proc Natl Acad Sci U S A ; 92(9): 3717-20, 1995 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-7731971

RESUMO

The main physiological regulator of erythropoiesis is the hematopoietic growth factor erythropoietin (EPO), which is induced in response to hypoxia. Binding of EPO to the EPO receptor (EPO-R), a member of the cytokine receptor superfamily, controls the terminal maturation of red blood cells. So far, EPO has been reported to act mainly on erythroid precursor cells. However, we have detected mRNA encoding both EPO and EPO-R in mouse brain by reverse transcription-PCR. Exposure to 0.1% carbon monoxide, a procedure that causes functional anemia, resulted in a 20-fold increase of EPO mRNA in mouse brain as quantified by competitive reverse transcription-PCR, whereas the EPO-R mRNA level was not influenced by hypoxia. Binding studies on mouse brain sections revealed defined binding sites for radioiodinated EPO in distinct brain areas. The specificity of EPO binding was assessed by homologous competition with an excess of unlabeled EPO and by using two monoclonal antibodies against human EPO, one inhibitory and the other noninhibitory for binding of EPO to EPO-R. Major EPO binding sites were observed in the hippocampus, capsula interna, cortex, and midbrain areas. Functional expression of the EPO-R and hypoxic upregulation of EPO suggest a role of EPO in the brain.


Assuntos
Encéfalo/metabolismo , Eritropoetina/metabolismo , Expressão Gênica , Receptores da Eritropoetina/biossíntese , Animais , Autorradiografia , Sequência de Bases , Sítios de Ligação , Encéfalo/citologia , Primers do DNA , Eritropoetina/biossíntese , Lobo Frontal/metabolismo , Radioisótopos do Iodo , Rim/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , Reação em Cadeia da Polimerase , Receptores da Eritropoetina/análise , Receptores da Eritropoetina/metabolismo , Células Tumorais Cultivadas
20.
Blood ; 98(9): 2872-4, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11675366

RESUMO

Kidney and liver are the major organs of erythropoietin (Epo) synthesis. However, Epo messenger RNA (mRNA) has been detected in several organs, such as brain, lung, and testis. Furthermore, functional Epo receptors have been demonstrated on different cell types, including rat Leydig cells. The aim of the study was to identify testicular cells expressing Epo mRNA and to quantitate its levels by competitive reverse transcriptase-polymerase chain reaction (RT-PCR). Besides whole testis, Epo transcripts were found in Sertoli and peritubular myoid cells, while no signal was detected in Leydig cells. Exposure of Sertoli cells to CoCl(2) led to an increase of Epo mRNA level. Semiquantitative competitive RT-PCR presented an increase in the level of Epo mRNA in Sertoli cells stimulated by follicle-stimulating hormone, while exposure of peritubular myoid cells cultures to testosterone reduced Epo mRNA expression. Due to the blood-testis barrier, basal expression of Epo suggests a not yet defined function of this hormone in testis.


Assuntos
Eritropoetina/metabolismo , Células de Sertoli/metabolismo , Animais , Eritropoetina/genética , Células Intersticiais do Testículo/metabolismo , Masculino , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/citologia , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA