Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
J Med Chem ; 65(2): 1505-1524, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-34818008

RESUMEN

Restoring lost heart muscle is an attractive goal for cardiovascular regenerative medicine. One appealing strategy is the therapeutic stimulation of cardiomyocyte proliferation, which inter alia remains challenging due to available assay technologies capturing the complex biology. Here, a high-throughput-formatted phenotypic assay platform was established using rodent whole heart-derived cells to preserve the cellular environment of cardiomyocytes. Several readouts allowed the quantification of cycling cardiomyocytes, including a transgenic H2B-mCherry system for unequivocal, automated detection of cardiomyocyte nuclei. A chemical genetics approach revealed pronounced species differences and furnished pan-kinase inhibitors 5 and 36 as potent and robust inducers of endoreplication and acytokinetic mitosis. Combined profiling of the commonly used p38 MAPK inhibitors SB203580 (1), SB239063 (2) and a novel set of skepinone-L (6) derivatives pointed to off-target effects beyond p38 that might be critical for effective cardiomyocyte cytokinesis. Kinome-focused screening eventually furnished TG003 (38) as a novel candidate for stimulating cardiomyocyte proliferation.


Asunto(s)
Ciclo Celular , Proliferación Celular , Corazón , Ensayos Analíticos de Alto Rendimiento , Sondas Moleculares , Miocitos Cardíacos , Inhibidores de Proteínas Quinasas , Animales , Ratones , Ratas , Animales Recién Nacidos , Corazón/efectos de los fármacos , Corazón/crecimiento & desarrollo , Ensayos Analíticos de Alto Rendimiento/métodos , Ratones Endogámicos C57BL , Mitosis , Sondas Moleculares/química , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología
3.
Sci Rep ; 8(1): 7145, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29739982

RESUMEN

Ventricular tachycardia (VT) is the most common and potentially lethal complication following myocardial infarction (MI). Biological correction of the conduction inhomogeneity that underlies re-entry could be a major advance in infarction therapy. As minimal increases in conduction of infarcted tissue markedly influence VT susceptibility, we reasoned that enhanced propagation of the electrical signal between non-excitable cells within a resolving infarct might comprise a simple means to decrease post-infarction arrhythmia risk. We therefore tested lentivirus-mediated delivery of the gap-junction protein Connexin 43 (Cx43) into acute myocardial lesions. Cx43 was expressed in (myo)fibroblasts and CD45+ cells within the scar and provided prominent and long lasting arrhythmia protection in vivo. Optical mapping of Cx43 injected hearts revealed enhanced conduction velocity within the scar, indicating Cx43-mediated electrical coupling between myocytes and (myo)fibroblasts. Thus, Cx43 gene therapy, by direct in vivo transduction of non-cardiomyocytes, comprises a simple and clinically applicable biological therapy that markedly reduces post-infarction VT.


Asunto(s)
Arritmias Cardíacas/genética , Cicatriz/genética , Conexina 43/genética , Terapia Genética , Infarto del Miocardio/genética , Animales , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/patología , Arritmias Cardíacas/terapia , Cicatriz/patología , Cicatriz/terapia , Conexina 43/administración & dosificación , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Vectores Genéticos/uso terapéutico , Células HEK293 , Humanos , Lentivirus/genética , Ratones , Células Musculares/metabolismo , Células Musculares/patología , Mioblastos/metabolismo , Mioblastos/patología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Taquicardia Ventricular/complicaciones , Taquicardia Ventricular/genética , Taquicardia Ventricular/patología , Taquicardia Ventricular/terapia
5.
Blood ; 117(18): 4905-14, 2011 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-21406725

RESUMEN

Erythropoietin (EPO), the key hormone in red blood cell renewal, is mainly produced in the adult kidney. Anemia and hypoxia substantially enhance EPO expression to increase erythropoiesis. Investigations of the cellular physiology of renal EPO production have been hampered by the lack of an adequate human cell line. In the present study, we present the human kidney cell line REPC (for renal Epo-producing cells), established from an explanted human kidney exhibiting EPO gene expression and release of the EPO protein in an oxygen-dependent manner. Hypoxic induction of EPO mRNA showed the typical transient increase and peak in expression after 36 hours under continuous conditions of hypoxia. Bioactive EPO protein accumulated in the culture supernatant. The induction of EPO gene expression in REPCs critically depended on the activation of hypoxia-inducible transcription factors (HIFs). SiRNA treatment revealed that the expression of EPO was largely dependent on the activation of the transcription factor complex HIF-2. In addition, hepatic nuclear factor 4α was shown to be critically involved in hypoxia-induced renal EPO expression. Using the human kidney cell line REPC, we provide for the first time a powerful tool with which to study the cellular and molecular regulation of renal EPO production.


Asunto(s)
Eritropoyetina/genética , Riñón/metabolismo , Oxígeno/metabolismo , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia de la Célula/genética , Línea Celular , Cartilla de ADN/genética , Dioxigenasas/genética , Eritropoyesis/genética , Eritropoyesis/fisiología , Eritropoyetina/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Riñón/citología , Modelos Biológicos , Procolágeno-Prolina Dioxigenasa/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Activación Transcripcional
6.
J Immunol ; 182(10): 6470-6, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19414801

RESUMEN

The oxygen-sensitive transcription factor hypoxia-inducible factor 1 (HIF-1) is known as the key regulator of hypoxia-induced gene expression. In addition to hypoxia, endotoxins such as bacterial LPS as well as proinflammatory cytokines have been shown to induce HIF-1, suggesting an integrative role for HIF-1 in conditions of hypoxia and inflammation. Cells can become tolerant to endotoxins by repetitive exposure to LPS. Herein, we studied the effect of endotoxin tolerance on HIF-1alpha accumulation and expression of HIF target genes in human monocytic cells and primary mouse peritoneal macrophages. Tolerant cells had reduced levels of HIF-1alpha under hypoxia, which was due to lowered levels of HIF-1alpha mRNA. HIF-1alpha expression is under control of NF-kappaB and increased DNA binding of the p52 subunit of NF-kappaB was found in tolerant cells. Knock down of p52 abolished the effects of endotoxin tolerance on HIF-1alpha expression, which suggest a negative regulatory role of p52 on HIF-1alpha transcription during endotoxin tolerance. Endotoxin tolerant cells showed diminished expression of the HIF target genes phosphoglycerate kinase 1 and adrenomedullin and reduced viability under hypoxic conditions, as well as a significantly reduced invasion. Peritoneal macrophages from endotoxin-tolerant mice made showed significantly reduced HIF-1alpha protein accumulation and subsequent HIF target gene expression. We conclude that endotoxin tolerance impairs HIF-1alpha induction which reduces the ability of monocytic cells to survive and function under hypoxic conditions.


Asunto(s)
Hipoxia de la Célula/inmunología , Endotoxinas/inmunología , Expresión Génica/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Tolerancia Inmunológica/inmunología , Monocitos/inmunología , Animales , Western Blotting , Línea Celular , Supervivencia Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Subunidad p52 de NF-kappa B/inmunología , Subunidad p52 de NF-kappa B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
BMC Gastroenterol ; 9: 26, 2009 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-19383129

RESUMEN

BACKGROUND: Recombinant human erythropoietin alpha (rHu-EPO) has been reported to protect the liver of rats and mice from ischemia-reperfusion injury. However, direct protective effects of rHu-EPO on hepatocytes and the responsible signalling pathways have not yet been described. The aim of the present work was to study the protective effect of rHu-EPO on warm hypoxia-reoxygenation and cold-induced injury to hepatocytes and the rHu-EPO-dependent signalling involved. METHODS: Loss of viability of isolated rat hepatocytes subjected to hypoxia/reoxygenation or incubated at 4 degrees C followed by rewarming was determined from released lactate dehydrogenase activity in the absence and presence of rHu-EPO (0.2-100 U/ml). Apoptotic nuclear morphology was assessed by fluorescence microscopy using the nuclear fluorophores H33342 and propidium iodide. Erythropoietin receptor (EPOR), EPO and Bcl-2 mRNAs were quantified by real time PCR. Activation of JAK-2, STAT-3 and STAT-5 in hepatocytes and rat livers perfused in situ was assessed by Western blotting. RESULTS: In contrast to previous in vivo studies on ischemia-reperfusion injury to the liver, rHu-EPO was without any protective effect on hypoxic injury, hypoxia-reoxygenation injury and cold-induced apoptosis to isolated cultured rat hepatocytes. EPOR mRNA was identified in these cells but specific detection of the EPO receptor protein was not possible due to the lack of antibody specificity. Both, in the cultured rat hepatocytes (10 U/ml for 15 minutes) and in the rat liver perfused in situ with rHu-EPO (8.9 U/ml for 15 minutes) no evidence for EPO-dependent signalling was found as indicated by missing effects of rHu-EPO on phosphorylation of JAK-2, STAT-3 and STAT-5 and on the induction of Bcl-2 mRNA. CONCLUSION: Together, these results indicate the absence of any protective EPO signalling in rat hepatocytes. This implies that the protection provided by rHu-EPO in vivo against ischemia-reperfusion and other causes of liver injury is most likely indirect and does not result from a direct effect on hepatocytes.


Asunto(s)
Eritropoyetina/farmacología , Hepatocitos/efectos de los fármacos , Daño por Reperfusión/prevención & control , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Células Cultivadas , Frío/efectos adversos , Eritropoyetina/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Janus Quinasa 2/metabolismo , Masculino , Modelos Animales , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/metabolismo , Ratas , Proteínas Recombinantes , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo
8.
Naunyn Schmiedebergs Arch Pharmacol ; 378(1): 27-32, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18488199

RESUMEN

We have previously shown that 2 weeks of hypoxia protect the right ventricle of the rat heart from subsequent ischemia and reperfusion (I/R). In the present study, we examined the following: (1) Do shorter periods of hypoxia protect from subsequent I/R? (2) Does intermittent normoxia increase the cardioprotective effect? (3) Is hypoxia-inducible factor-1alpha (HIF-1alpha), erythropoietin (EPO), or vascular endothelial growth factor (VEGF) involved in the protective effects? Preischemic cardiac work was followed by global ischemia, reperfusion, and postischemic cardiac work (15 min each). External heart work was determined at the end of both work phases. Four groups of hearts were investigated: hearts from normoxic rats (n=8), hearts from rats after 24 h of continuous hypoxia (10.5% inspired oxygen, n=7), hearts from rats after 24 h hypoxia with a single intermission of 30 min normoxia (n=9), and hearts from rats after 24 h hypoxia and multiple intermissions of 30 min normoxia (n=7). Protein levels of HIF-1alpha and mRNA levels of EPO and VEGF were determined in right ventricular tissue of normoxic and hypoxic hearts. Postischemic right heart recovery was better in all three hypoxic groups compared with normoxic hearts (61.8 +/- 5.9%, 65.6 +/- 3.0%, and 75.7 +/- 2.6% vs. 46.0 +/- 3.9%, p < 0.01). Hypoxia with multiple normoxic intermissions further improved right heart recovery compared to continuous hypoxia (p < 0.05). HIF-1alpha protein levels were 80.3 +/- 2.5 pg/microg in normoxic hearts and 108.0 +/- 10.3 pg/microg in hypoxic hearts (p = 0.02). No differences in EPO and VEGF mRNA levels were found between normoxic and hypoxic hearts. Twenty-four hours of continuous hypoxia protect the isolated working right heart from subsequent ischemia and reperfusion. When preceding hypoxia is interrupted by multiple reoxygenation periods, there is a further significant increase in cardiac functional recovery. HIF-1alpha may be involved in the protective effect.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Precondicionamiento Isquémico Miocárdico/métodos , Daño por Reperfusión/prevención & control , Animales , Eritropoyetina/metabolismo , Ventrículos Cardíacos/fisiopatología , Masculino , Oxígeno/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Daño por Reperfusión/fisiopatología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Biochem J ; 396(3): 517-27, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16533170

RESUMEN

Inflammatory mediators activate the transcriptional complex HIF-1 (hypoxia-inducible factor-1), the key regulator of hypoxia-induced gene expression. Here we report that bacterial LPS (lipopolysaccharide) induces HIF-1alpha mRNA expression and HIF-1alpha protein accumulation in human monocytes as well as in non-differentiated and differentiated cells of the human monocytic cell line THP-1 under normoxic conditions. LPS and hypoxia synergistically activated HIF-1. Whereas LPS increased HIF-1alpha mRNA expression through activation of a NF-kappaB (nuclear factor kappaB) site in the promoter of the HIF-1alpha gene, hypoxia post-translationally stabilized HIF-1alpha protein. HIF-1alpha activation was followed by increased expression of the HIF-1 target gene encoding ADM (adrenomedullin). Knocking down HIF-1alpha by RNA interference significantly decreased ADM expression, which underlines the importance of HIF-1 for the LPS-induced ADM expression in normoxia. Simultaneously with HIF-1 activation, an increase in p44/42 MAPK (mitogen-activated protein kinase) phosphorylation was observed after incubation with LPS. In cells pretreated with the p44/42 MAPK inhibitor PD 98059 or with RNAi (interfering RNA) directed against p44/42 MAPK, LPS-induced HIF-1alpha accumulation and ADM expression were significantly decreased. From these results we conclude that LPS critically involves the p44/42 MAPK and NF-kappaB pathway in the activation of HIF-1, which is an important transcription factor for LPS-induced ADM expression.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Factor 1 Inducible por Hipoxia/metabolismo , Lipopolisacáridos/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Monocitos/metabolismo , FN-kappa B/metabolismo , Adrenomedulina , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Flavonoides/farmacología , Humanos , Hipoxia/fisiopatología , Receptores de Lipopolisacáridos/biosíntesis , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Péptidos/metabolismo , Interferencia de ARN
10.
J Interferon Cytokine Res ; 25(7): 407-13, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16022585

RESUMEN

Clinical and laboratory studies indicate that thrombopoietin (TPO) gene expression increases during inflammation. To clarify the role of interleukin 6 (IL-6) in this process, blood cell counts, plasma TPO concentrations, and hepatic and renal TPO mRNA levels were investigated in wild-type and IL-6 knockout mice, with sterile abscesses produced by subcutaneous injection of turpentine oil. Treatment did not cause a change in blood cell counts during the 72 h period of observation. The numbers of thrombocytes and erythrocytes were slightly lower in the IL-6 knockout mice than in the wild-type littermates under all conditions. Plasma IL-6 and TPO concentrations increased on turpentine injection only in the wild-type mice. In addition, turpentine treatment of these caused an increase in hepatic TPO mRNA levels as assessed by competitive polymerase chain reaction (RT-PCR) and real-time PCR, whereas renal TPO mRNA levels were unaltered. TPO mRNA levels did not increase in the livers of IL-6 knockout mice on turpentine treatment. These results support the concept that TPO behaves like an acute-phase protein in that its synthesis is induced by IL-6 in the liver.


Asunto(s)
Inflamación/metabolismo , Interleucina-6/fisiología , Trombopoyetina/biosíntesis , Enfermedad Aguda , Animales , Recuento de Células Sanguíneas , Inflamación/genética , Inflamación/inmunología , Interleucina-6/biosíntesis , Interleucina-6/genética , Riñón/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Trombopoyetina/sangre , Trombopoyetina/genética
11.
Cancer Res ; 65(11): 4690-7, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15930287

RESUMEN

Adrenomedullin (ADM) is a potent hypotensive peptide produced by macrophages and endothelial cells during ischemia and sepsis. The molecular mechanisms that control ADM gene expression in tumor cells are still poorly defined. It is known, however, that hypoxia potently increases ADM expression by activation of the transcription factor complex hypoxia inducible factor 1 (HIF-1). Proinflammatory cytokines produced by tumor invading macrophages likewise activate expression of ADM. Herein, we show that apart from hypoxia, the proinflammatory cytokine interleukin 1beta (IL-1beta) induced the expression of ADM mRNA through activation of HIF-1 under normoxic conditions and enhanced the hypoxia-induced expression in the human ovarian carcinoma cell line OVCAR-3. IL-1beta significantly increased accumulation and nuclear translocation of HIF-1alpha under normoxic conditions and amplified hypoxic HIF-1 activation. IL-1beta treatment affected neither HIF-1alpha mRNA levels nor the hydroxylation status of HIF-1alpha and, thus, stability of the protein. Instead cycloheximide effectively prevented the increase in HIF-1alpha protein, indicating a stimulatory effect of IL-1beta on HIF-1alpha translation. Finally, treatment of HIF-1alpha with short interfering RNA revealed a significant role for HIF-1 in the IL-1beta-dependent stimulation of ADM expression.


Asunto(s)
Interleucina-1/fisiología , Neoplasias Ováricas/metabolismo , Péptidos/genética , Factores de Transcripción/metabolismo , Adrenomedulina , Hipoxia de la Célula , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , FN-kappa B/fisiología , Neoplasias Ováricas/genética , Péptidos/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
12.
J Cell Biochem ; 93(1): 153-61, 2004 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-15352172

RESUMEN

The glycoprotein hormone Erythropoietin (EPO) stimulates red cell production and maturation. EPO is produced by the kidneys and the fetal liver in response to hypoxia (HOX). Recently, EPO expression has also been observed in the central nervous system where it may be neuroprotective. It remained unclear, however, whether EPO is expressed in the peripheral nervous system and, if so, whether a neuronal phenotype is required for its regulation. Herein, we report that EPO expression was induced by HOX and a HOX mimetic in two cell lines derived from neuroblastoma (NB), a tumor of the peripheral nervous system. Both cell lines with inducible EPO expression, SH-SY5Y and Kelly cells, expressed typical neuronal markers like neuropeptide Y (NPY), growth-associated protein-43 (GAP-43), and neuron-specific enolase (ENO). NB cells with a more epithelial phenotype like SH-SHEP and LAN-5 did not show HOX inducible EPO gene regulation. Still, oxygen sensing and up-regulation of hypoxia-inducible factor-1 (HIF-1) were intact in all cell lines. We found that CpG methylation of the HIF binding site (HBS) in the EPO gene 3' enhancer was only present in the SH-SHEP and LAN-5 cells but not in SH-SY5Y and Kelly cells with regulated EPO expression. The addition of recombinant EPO to all NB cells, both under normoxic and hypoxic conditions, had no effect on cell proliferation. We conclude that the ability to respond to HOX with an increase in EPO expression in human NB may depend on CpG methylation and the differentiation status of these embryonic tumor cells but does not affect the proliferative characteristics of the cells.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Eritropoyetina/genética , Hipoxia/metabolismo , Neuroblastoma/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias del Sistema Nervioso Periférico/metabolismo , Factores de Transcripción/metabolismo , Sitios de Unión , Western Blotting , Diferenciación Celular , Islas de CpG , Proteínas de Unión al ADN/genética , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica , Secuencias Hélice-Asa-Hélice , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neuroblastoma/patología , Proteínas Nucleares/genética , Neoplasias del Sistema Nervioso Periférico/patología , Receptores de Eritropoyetina/metabolismo , Factores de Transcripción/genética , Células Tumorales Cultivadas
13.
J Biol Chem ; 279(43): 44976-86, 2004 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-15322093

RESUMEN

Hypoxia-Inducible Factor-1 (HIF-1) is the key transcription factor in control of the expression of hypoxia-inducible genes needed by cells to adapt to decreased oxygen availability. Herein, we investigated the HIF-1alpha-mediated gene expression of carbonic anhydrase 9 (CA9) in response to hypoxia and changes of intracellular calcium levels in the neuroblastoma cell line SH-SY5Y. Decreasing the intracellular calcium level by BAPTA (1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid) induced HIF-1alpha nuclear accumulation and enhanced HIF-1 DNA binding within 1 h of incubation. Like hypoxia, BAPTA stimulated HIF-1-dependent transcription by increasing the activity of the C-terminal transactivation domain of HIF-1alpha and greatly enhanced expression of the HIF-1 target gene CA9. Detailed analysis of HIF-1alpha accumulation revealed that BAPTA attenuated the interaction of HIF-1alpha with von-Hippel-Lindau protein thus decreasing proteasomal degradation of HIF-1alpha. Knock down of HIF-1alpha mRNA and protein by small interference RNA for HIF-1alpha revealed that both hypoxia and the BAPTA-induced gene expression of CA9 were strictly dependent on HIF-1alpha. In contrast, elevation of cytosolic calcium level by thapsigargin reduced the BAPTA-mediated effects. Measurements of intracellular calcium under hypoxia revealed a change in the cellular calcium distribution. BAPTA-dependent induction of HIF-1 activity was not caused by its in vitro capability to chelate iron. Instead, effective chelation of cellular calcium caused the accumulation of HIF-1alpha protein through inhibition of HIF-prolyl hydroxylases and activated HIF-1-dependent gene expression under normoxic conditions.


Asunto(s)
Calcio/metabolismo , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Factores de Transcripción/metabolismo , Western Blotting , Anhidrasas Carbónicas/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular , Sistema Libre de Células , Relación Dosis-Respuesta a Droga , Ácido Egtácico/farmacología , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Iones , Hierro/química , Hierro/metabolismo , Rayos Láser , Ligandos , Microscopía Confocal , Microscopía Fluorescente , Plásmidos/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tapsigargina/farmacología , Factores de Tiempo
14.
Am J Physiol Regul Integr Comp Physiol ; 287(3): R600-7, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15155277

RESUMEN

Oxygen is of vital importance for the metabolism and function of all cells in the human body. Hypoxia, the reduction of oxygen supply, results in adaptationally appropriate alterations in gene expression through the activation of hypoxia-inducible factor 1 (HIF-1) to overcome any shortage of oxygen. Thyroid hormones are required for normal function of nearly all tissues, with major effects on oxygen consumption and metabolic rate. Thyroid hormones have been found to augment the oxygen capacity of the blood by increasing the production of erythropoietin (EPO) and to improve perfusion by vasodilation through the augmented expression of adrenomedullin (ADM). Because the hypoxic expression of both genes depends on HIF-1, we studied the influence of thyroid hormone on HIF-1 activation in the human hepatoma cell line HepG2 under normoxic and hypoxic conditions. We found that thyroid hormones increased HIF-1alpha protein accumulation by increasing HIF-1alpha protein synthesis rather than attenuating its proteasomal degradation. HIF-1alpha expression directly correlated with augmented HIF-1 DNA binding and transcriptional activity of luciferase reporter plasmids, whereas HIF-1beta levels remained unaffected. Knocking down HIF-1alpha by short interfering RNA (siRNA) clearly demonstrated that thyroid hormone-induced target gene expression required the presence of HIF-1. Although an increased association of the two known coactivators of HIF-1, p300 and SRC-1, was found, thyroid hormone did not affect the activity of the isolated COOH-terminal transactivating domain of HIF-1alpha. Increased synthesis of HIF-1alpha may contribute to the adaptive response of increased oxygen demand under hyperthyroid conditions.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Eritropoyetina/biosíntesis , Proteínas Nucleares/metabolismo , Tiroxina/farmacología , Factores de Transcripción , Triyodotironina/farmacología , Adrenomedulina , Línea Celular Tumoral , Eritropoyetina/genética , Expresión Génica/efectos de los fármacos , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Péptidos/genética , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología , Transcripción Genética/efectos de los fármacos
15.
Brain Res Dev Brain Res ; 143(2): 217-23, 2003 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-12855193

RESUMEN

Thrombopoietin gene expression in the human adult central nervous system (CNS) appears to be locally restricted. The aim of this study was to identify areas of thrombopoietin expression in the developing human CNS, and to compare the thrombopoietin mRNA content in the CNS to that in liver and kidneys as major sites of thrombopoietin production. Thrombopoietin protein concentrations in the cerebrospinal fluid (CSF) were measured by ELISA. In 14 fetuses and neonates with perinatal death, thrombopoietin mRNA expression was measured by competitive RT-PCR. Thrombopoietin mRNA was expressed in 29 of 32 specimens taken from the CNS. The following ranking of the intensity of expression in the CNS was possible: Spinal cord=cerebellum=cortex>>pituitary gland>>>brain stem=corpora amygdala=hippocampus. Whereas in the latter three tissues only trace amounts of thrombopoietin transcripts were detectable, thrombopoietin mRNA levels in the spinal cord were comparable to levels in liver and kidney. Thrombopoietin protein concentrations in CSF ranged between 41 and 75 pg/ml. In the developing human CNS, the thrombopoietin gene is abundantly expressed. Considering that thrombopoietin contains a neurotrophic sequence, it may well play a role in neuronal cell biology.


Asunto(s)
Sistema Nervioso Central/embriología , Regulación del Desarrollo de la Expresión Génica , Trombopoyetina/genética , Sistema Nervioso Central/fisiología , Ensayo de Inmunoadsorción Enzimática , Feto , Humanos , Recién Nacido , Riñón/embriología , Hígado/embriología , Especificidad de Órganos , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombopoyetina/biosíntesis , Trombopoyetina/sangre , Trombopoyetina/líquido cefalorraquídeo
16.
Br J Haematol ; 119(2): 510-5, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12406094

RESUMEN

Regulation of gene expression during the ontogeny of haematopoiesis in the human fetal bone marrow is poorly understood. Studies in mice demonstrated that GATA-1, -2 and -3 play pivotal roles in haematopoiesis. In this study, we identified GATA-1-, GATA-2- and GATA-3-expressing cells in bone marrow sections and analysed the expression of GATA-transcription factors during the development of human fetal bone marrow haematopoiesis using semiquantitative reverse transcription-polymerase chain reaction (RT-PCR). We showed that GATA-1, -2 and -3 were expressed only in haematopoietic cells in the bone marrow. RT-PCR analysis demonstrated that (1) GATA-1 expression significantly increased during gestation; (2) GATA-2 expression peaked at the onset of medullary haematopoiesis, declined thereafter, and remained at a constant level after 30 weeks post conception; and (3) GATA-3 expression revealed no changes during development. The results indicated that the onset of medullary haematopoiesis in humans is accompanied by high expression of GATA-2, reflecting high proliferation rates of early haematopoietic progenitor cells, whereas expression of GATA-1 mirrors haematopoietic activity.


Asunto(s)
Médula Ósea/embriología , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis , ARN Mensajero/análisis , Factores de Transcripción/genética , División Celular/fisiología , Proteínas de Unión al ADN/genética , Factores de Unión al ADN Específico de las Células Eritroides , Factor de Transcripción GATA1 , Factor de Transcripción GATA2 , Factor de Transcripción GATA3 , Expresión Génica , Edad Gestacional , Células Madre Hematopoyéticas/citología , Humanos , Recién Nacido , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/genética
17.
Blood ; 100(7): 2623-8, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12239177

RESUMEN

Two human neuroblastoma (NB) cell lines, SH-SY5Y and Kelly, were found to express the gene for erythropoietin (EPO) in an oxygen (O(2))-dependent manner. However, NB cells had maximal production of EPO with lower partial pressure of O(2) values than the well-characterized hepatoma cell line HepG2. This maximal EPO expression was preceded by accumulation of the O(2)-sensitive alpha subunit of the heterodimeric transcription-factor complex hypoxia-inducible factor 1 (HIF-1). Western blot analysis revealed that the amount of the beta subunit of HIF-1, identical to aryl hydrocarbon receptor nuclear translocator 1 (ARNT1), and the homolog ARNT2 increased in nuclear extracts from SH-SY5Y cells exposed to anoxia. In neuronal cells, ARNT1 and ARNT2 can form a heterodimer with HIF-1alpha, generating a functional HIF-1 complex. Using the hypoxia response element of the human EPO enhancer, we conducted electrophoretic mobility shift assays that showed accumulation and binding of HIF-1 complexes containing both ARNT1 and ARNT2 in NB cells. In addition to the HIF-1 complex, hepatocyte nuclear factor 4alpha (HNF4alpha) was found to be indispensable for hypoxia-induced EPO gene expression in hepatoma cells. Western blot analysis and polymerase chain reaction assessment showed that NB cells express neither HNF4alpha nor the splicing variant HNF4alpha7 and thus express EPO in an HNF4alpha-independent manner. Together, SH-SY5Y and Kelly cells may provide a new in vitro model for studying the mechanism of tissue-specific, hypoxia-inducible EPO gene expression.


Asunto(s)
Hipoxia de la Célula/fisiología , Eritropoyetina/genética , Regulación Neoplásica de la Expresión Génica , Neuroblastoma/genética , Consumo de Oxígeno/fisiología , Secuencia de Bases , Carcinoma Hepatocelular , Cartilla de ADN , ADN Complementario/genética , Humanos , Neoplasias Hepáticas , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...