Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Placenta ; 24(10): 941-50, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14580376

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/fisiología , Factores de Transcripción , Activación Transcripcional , Factor de Crecimiento Transformador beta/genética , Animales , Sitios de Unión , Diferenciación Celular , Hipoxia de la Célula , ADN/metabolismo , Femenino , Regulación de la Expresión Génica , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Embarazo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Factor de Crecimiento Transformador beta3 , Trofoblastos/citología
2.
Microvasc Res ; 63(1): 70-80, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11749074

RESUMEN

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.


Asunto(s)
Encéfalo/irrigación sanguínea , Factores de Crecimiento Endotelial/metabolismo , Endotelio Vascular/metabolismo , Hipoxia , Linfocinas/metabolismo , Proteínas de la Membrana/biosíntesis , Microcirculación/metabolismo , Fosfoproteínas/biosíntesis , Actinas/biosíntesis , Animales , Barrera Hematoencefálica , Western Blotting , Encéfalo/metabolismo , Línea Celular , Células Cultivadas , Citoplasma/metabolismo , Endotelio Vascular/citología , Inmunohistoquímica , Ratones , Fosforilación , Pruebas de Precipitina , Ratas , Porcinos , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Proteína de la Zonula Occludens-1
3.
Blood ; 98(9): 2872-4, 2001 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11675366

RESUMEN

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.


Asunto(s)
Eritropoyetina/metabolismo , Células de Sertoli/metabolismo , Animales , Eritropoyetina/genética , Células Intersticiales del Testículo/metabolismo , Masculino , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/citología , Distribución Tisular
4.
Glia ; 30(3): 271-8, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10756076

RESUMEN

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.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Eritropoyetina/genética , Hipoxia/metabolismo , Neuronas/metabolismo , Oxidación-Reducción , Oxígeno/metabolismo , ARN Mensajero/metabolismo , Animales , Células Cultivadas , Hipoxia/genética , Masculino , Ratones , Ratones Endogámicos , Reacción en Cadena de la Polimerasa , Factores de Tiempo
5.
Eur J Appl Physiol ; 81(6): 497-503, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10774874

RESUMEN

In order to investigate whether vascular endothelial growth factor (VEGF) and inflammatory pathways are activated during acute hypobaric hypoxia in subjects who are susceptible to high-altitude pulmonary oedema (HAPE-S), seven HAPE-S and five control subjects were exposed to simulated altitude corresponding to 4000 m in a hypobaric chamber for 1 day. Peripheral venous blood was taken at 450 m (Zürich level) and at 4000 m, and levels of erythropoietin (EPO), VEGF, interleukin-6 (IL-6) and the acute-phase proteins complement C3 (C3), alpha1-antitrypsin (alpha1AT), transferrin (Tf) and C-reactive protein (CRP) were measured. Peripheral arterial oxygen saturation (SaO2) was recorded. Chest radiography was performed before and immediately after the experiment. EPO increased during altitude exposure, correlating with SaO2, in both groups (r = -0.86, P < 0.001). Venous serum VEGF did not show any elevation despite a marked decrease in SaO2 in the HAPE-S subjects [mean (SD) HAPE-S: 69.6 (9.1)%; controls: 78.7 (5.2)%]. C3 and alpha1AT levels increased in HAPE-S during hypobaric hypoxia [from 0.94 (0.11) g/l to 1.07 (0.13) g/l, and from 1.16 (0.08) g/l to 1.49 (0.27) g/l, respectively; P < 0.05], but remained within the clinical reference ranges. No significant elevations of IL-6, Tf or CRP were observed in either group. The post-exposure chest radiography revealed no signs of oedema. We conclude that VEGF is not up-regulated in HAPE-S and thus does not seem to increase critically pulmonary vascular permeability during the 1st day at high altitude. Furthermore, our data provide evidence against a clinically relevant inflammation in the initial phase of exposure to hypoxia in HAPE-S, although C3 and alpha1AT are mildly induced.


Asunto(s)
Reacción de Fase Aguda/etiología , Altitud , Presión Atmosférica , Factores de Crecimiento Endotelial/sangre , Hipoxia/sangre , Hipoxia/complicaciones , Linfocinas/sangre , Edema Pulmonar/etiología , Proteínas de Fase Aguda/análisis , Adulto , Susceptibilidad a Enfermedades , Eritropoyetina/sangre , Humanos , Hipoxia/etiología , Hipoxia/fisiopatología , Masculino , Persona de Mediana Edad , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
6.
FASEB J ; 13(11): 1315-24, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10428756

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/metabolismo , Proteínas Portadoras/metabolismo , Neuropéptidos/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Serpinas/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Precursor de Proteína beta-Amiloide , Animales , Astrocitos/metabolismo , Astrocitos/patología , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Proteínas Portadoras/genética , Muerte Celular/genética , Células Cultivadas , Regulación de la Expresión Génica , Ratones , Neuronas/patología , Neuropéptidos/genética , Inhibidor 1 de Activador Plasminogénico/genética , Nexinas de Proteasas , Receptores de Superficie Celular , Serpinas/genética , Activador de Tejido Plasminógeno/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología , Neuroserpina
7.
J Cereb Blood Flow Metab ; 19(6): 643-51, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10366194

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/metabolismo , Eritropoyetina/biosíntesis , Eritropoyetina/farmacología , Receptores de Eritropoyetina/biosíntesis , Animales , Astrocitos/efectos de los fármacos , Western Blotting , Encéfalo/citología , Química Encefálica/fisiología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/metabolismo , Infarto Cerebral/patología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Eritropoyetina/administración & dosificación , Inmunohistoquímica , Hibridación in Situ , Inyecciones Intraventriculares , Ratones , Neuronas/efectos de los fármacos
8.
Thromb Haemost ; 82 Suppl 1: 44-52, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10695485

RESUMEN

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.


Asunto(s)
Sistema Cardiovascular/fisiopatología , Isquemia Miocárdica/fisiopatología , Neovascularización Patológica , Adulto , Animales , Humanos
9.
Int J Mol Med ; 2(3): 317-24, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9855704

RESUMEN

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.


Asunto(s)
Eritropoyetina/biosíntesis , Carcinoma Hepatocelular , Recuento de Células , Hipoxia de la Célula , Cobalto/farmacología , Dexametasona/farmacología , Humanos , Interleucina-6/farmacología , Neoplasias Hepáticas , Oxígeno/metabolismo , Transducción de Señal
10.
Proc Natl Acad Sci U S A ; 95(26): 15809-14, 1998 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-9861052

RESUMEN

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.


Asunto(s)
Factores de Crecimiento Endotelial/genética , Regulación de la Expresión Génica , Hipoxia/fisiopatología , Linfocinas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/genética , Animales , Encéfalo/metabolismo , Plexo Coroideo/metabolismo , Factores de Crecimiento Endotelial/biosíntesis , Hibridación in Situ , Glomérulos Renales/metabolismo , Túbulos Renales/metabolismo , Linfocinas/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Especificidad de Órganos , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento Endotelial Vascular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/metabolismo , Testículo/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
11.
Blood ; 92(9): 3388-93, 1998 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9787178

RESUMEN

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.


Asunto(s)
Neoplasias del Sistema Nervioso Central/genética , Factores de Crecimiento Endotelial/biosíntesis , Eritropoyetina/biosíntesis , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Hemangioblastoma/genética , Hormonas Ectópicas/biosíntesis , Ligasas , Linfocinas/biosíntesis , Proteínas de Neoplasias/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Biosíntesis de Proteínas , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas , Enfermedad de von Hippel-Lindau/genética , Adulto , Anciano , Encéfalo/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Hipoxia de la Célula , Neoplasias del Sistema Nervioso Central/metabolismo , Neoplasias del Sistema Nervioso Central/patología , Factores de Crecimiento Endotelial/genética , Eritropoyesis , Eritropoyetina/genética , Femenino , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Hemangioblastoma/metabolismo , Hemangioblastoma/patología , Hormonas Ectópicas/genética , Humanos , Hibridación in Situ , Linfocinas/genética , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/genética , Proteínas/genética , Proteínas/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Células del Estroma/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau
12.
Pediatr Res ; 43(3): 349-54, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9505273

RESUMEN

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.


Asunto(s)
Eritropoyesis , Feto/patología , Hígado/patología , Preeclampsia/patología , Líquido Amniótico/metabolismo , Estudios de Casos y Controles , Citocinas/metabolismo , Eritropoyesis/fisiología , Eritropoyetina/sangre , Femenino , Sangre Fetal/metabolismo , Retardo del Crecimiento Fetal/etiología , Retardo del Crecimiento Fetal/patología , Retardo del Crecimiento Fetal/fisiopatología , Feto/fisiopatología , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Humanos , Hígado/fisiopatología , Intercambio Materno-Fetal , Preeclampsia/complicaciones , Preeclampsia/fisiopatología , Embarazo
13.
Kidney Int ; 51(2): 416-8, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9027715

RESUMEN

Until now, erythropoietin (EPO) was thought to be produced exclusively in fetal liver and adult kidney and to regulate mammalian erythropoiesis. However, we recently showed that steady state levels of EPO mRNA could be induced up to 100-fold in primary mouse astrocytes cultured under hypoxic conditions, and also reported the presence of mRNA for EPO and its receptor in the brain of mouse, monkey and human. In extending these studies on humans we now show that immunoreactive EPO is present in ventricular cerebrospinal fluid (CSF) of 5 patients with traumatic brain injuries: EPO was found in 15 out of 15 CSF samples. There was no correlation between the serum EPO concentration and the concentration in the CSF. However, EPO concentrations in CSF correlated with the degree of blood-brain-barrier dysfunction. This suggests that EPO does not cross the intact blood-brain-barrier, implying that EPO is produced in the brain itself, most probably by astrocytes in an oxygen-dependent manner. In view that neuronal cells carry the EPO receptor, we propose that EPO acts in a paracrine fashion in the central nervous system and might function as a protective factor against hypoxia-induced damage of neurons.


Asunto(s)
Encéfalo/metabolismo , Eritropoyetina/biosíntesis , Adulto , Animales , Barrera Hematoencefálica , Traumatismos Craneocerebrales/sangre , Traumatismos Craneocerebrales/líquido cefalorraquídeo , Eritropoyetina/líquido cefalorraquídeo , Eritropoyetina/genética , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
Kidney Int ; 51(2): 560-3, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9027739

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN Mensajero/genética , Factores de Transcripción , Animales , Hipoxia de la Célula/genética , Hipoxia de la Célula/fisiología , Línea Celular , ADN/metabolismo , Proteínas de Unión al ADN/química , Factores de Crecimiento Endotelial/genética , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Cinética , Linfocinas/genética , Ratones , Proteínas Nucleares/química , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
15.
Kidney Int ; 51(2): 564-6, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9027740

RESUMEN

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.


Asunto(s)
AMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Sitios de Unión , Hipoxia de la Célula/genética , Hipoxia de la Célula/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , ADN/genética , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Eritropoyetina/genética , Regulación de la Expresión Génica , Genes Reporteros , Células HeLa , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Luciferasas/genética , Transfección
16.
Am J Physiol ; 271(4 Pt 1): C1172-80, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8897823

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Oxígeno/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , Núcleo Celular/metabolismo , Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Cianuro de Potasio/farmacología
17.
Biochem Biophys Res Commun ; 223(1): 54-9, 1996 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-8660378

RESUMEN

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.


Asunto(s)
Evolución Biológica , Mapeo Cromosómico , Proteínas de Unión al ADN/biosíntesis , Proteínas Nucleares/biosíntesis , Transcripción Genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Secuencia Conservada , Cartilla de ADN , ADN Complementario , Proteínas de Unión al ADN/química , Células HeLa , Humanos , Hipoxia , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Riñón/metabolismo , Neoplasias Hepáticas Experimentales , Pulmón/metabolismo , Sustancias Macromoleculares , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/química , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/biosíntesis , Células Tumorales Cultivadas
18.
Eur J Neurosci ; 8(4): 666-76, 1996 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9081618

RESUMEN

The haematopoietic growth factor erythropoietin is the primary regulator of mammalian erythropoiesis and is produced by the kidney and the liver in an oxygen-dependent manner. We and others have recently demonstrated erythropoietin gene expression in the rodent brain. In this work, we show that cerebral erythropoietin gene expression is not restricted to rodents but occurs also in the primate brain. Erythropoietin mRNA was detected in biopsies from the human hippocampus, amygdala and temporal cortex and in various brain areas of the monkey Macaca mulatta. Exposure to a low level of oxygen led to elevated erythropoietin mRNA levels in the monkey brain, as did anaemia in the mouse brain. In addition, erythropoietin receptor mRNA was detected in all brain biopsies tested from man, monkey and mouse. Analysis of primary cerebral cells isolated from newborn mice revealed that astrocytes, but not microglia cells, expressed erythropoietin. When incubated at 1% oxygen, astrocytes showed >100-fold time-dependent erythropoietin mRNA accumulation, as measured with the quantitative reverse transcription-polymerase chain reaction. The specificity of hypoxic gene induction in these cells was confirmed by quantitative Northern blot analysis showing hypoxic up-regulation of mRNA encoding the vascular endothelial growth factor, but not of other genes. These findings demonstrate that erythropoietin and its receptor are expressed in the brain of primates as they are in rodents, and that, at least in mice, primary astrocytes are a source of cerebral erythropoietin expression which can be up-regulated by reduced oxygenation.


Asunto(s)
Encéfalo/fisiología , Eritropoyetina/genética , Expresión Génica , Macaca mulatta/genética , Ratones/genética , Adulto , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Factores de Crecimiento Endotelial/metabolismo , Eritropoyetina/metabolismo , Femenino , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Linfocinas/metabolismo , Masculino , Oxígeno/farmacología , ARN Mensajero/metabolismo , Receptores de Eritropoyetina/genética , Distribución Tisular , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
19.
Proc Natl Acad Sci U S A ; 93(7): 2867-72, 1996 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-8610133

RESUMEN

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.


Asunto(s)
Blastocisto/fisiología , Diferenciación Celular , Regulación de la Expresión Génica , Consumo de Oxígeno , Células Madre/fisiología , Aerobiosis , Animales , Secuencia de Bases , Blastocisto/citología , Hipoxia de la Célula , Células Cultivadas , Cartilla de ADN , Factores de Crecimiento Endotelial/biosíntesis , Colorantes Fluorescentes , Fructosa-Bifosfato Aldolasa/biosíntesis , Isoenzimas/biosíntesis , Cinética , Linfocinas/biosíntesis , Ratones , Microscopía Confocal , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Células Madre/citología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
20.
Blood ; 87(2): 756-61, 1996 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-8555500

RESUMEN

Reduced oxygenation of a variety of cells results in transcriptional upregulation of several genes, including the hematopoietic hormone erythropoietin, the angiogenic vascular endothelial growth factor (VEGF), and glycolytic enzymes such as aldolase. Recently, the heme protein cytochrome b558 of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex has been proposed as a key component of the oxygen-sensing mechanism. Cytochrome b558 consists of the p22phox and gp91phox subunits and is essential for superoxide generation in phagocytes and B lymphocytes. Mutations in these subunits result in cytochrome b558-negative chronic granulomatous disease (cytb- CGD), an inherited disorder in humans characterized by reduced microbicidal activity due to deficient superoxide generation. To test whether NADPH oxidase is involved in oxygen sensing, we exposed wild-type B-cell lines as well as cytb- CGD-derived B cell lines, deficient in either p22phox or gp91phox, to hypoxia (1% oxygen) or CoCl2 (100 mumol/L) and compared the mRNA levels of VEGF and aldolase with the untreated controls. Northern blot analysis revealed unimpaired basal and inducible expression of VEGF and aldolase mRNA in all four cytb- CGD-derived B-cell lines compared with wild-type cells. Furthermore, reconstitution of cytochrome b558 expression in cytb- CGD-derived B cells by transfection with p22phox or gp91phox expression vectors did not modify VEGF and aldolase mRNA expression. Thus, cytochrome b558 of the NADPH oxidase complex appears not to be essential for hypoxia-activated gene expression and can be excluded as a candidate for the putative universal oxygen sensor.


Asunto(s)
Linfocitos B/metabolismo , Hipoxia de la Célula , Grupo Citocromo b/fisiología , Factores de Crecimiento Endotelial/biosíntesis , Fructosa-Bifosfato Aldolasa/biosíntesis , Regulación de la Expresión Génica , Enfermedad Granulomatosa Crónica/patología , Linfocinas/biosíntesis , Glicoproteínas de Membrana/deficiencia , Proteínas de Transporte de Membrana , Complejos Multienzimáticos/fisiología , NADH NADPH Oxidorreductasas/fisiología , NADPH Deshidrogenasa/deficiencia , Fosfoproteínas/deficiencia , Secuencia de Bases , Biomarcadores , Línea Celular Transformada , Cobalto/farmacología , Factores de Crecimiento Endotelial/genética , Fructosa-Bifosfato Aldolasa/genética , Enfermedad Granulomatosa Crónica/enzimología , Enfermedad Granulomatosa Crónica/genética , Humanos , Peróxido de Hidrógeno/metabolismo , Linfocinas/genética , Glicoproteínas de Membrana/fisiología , Datos de Secuencia Molecular , NADPH Deshidrogenasa/fisiología , NADPH Oxidasa 2 , NADPH Oxidasas , Oxígeno/metabolismo , Presión Parcial , Fosfoproteínas/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA