Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Vet Intern Med ; 38(1): 381-387, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37994487

RESUMEN

BACKGROUND: Inhibition of hypoxia-inducible factor prolyl hydroxylase (HIF-PH) stimulates erythropoiesis in rats, dogs, monkeys, and humans. HYPOTHESIS/OBJECTIVE: Determine if molidustat, a novel HIF-PH inhibitor, stimulates erythropoiesis in healthy cats. ANIMALS: Seventeen healthy adult laboratory cats. METHODS: Randomized, placebo-controlled study. Cats were treated PO once daily with suspensions of 0 (Group 1; n = 6), 5 (Group 2; n = 6), or 10 (Group 3; n = 5) mg/kg of molidustat. Effects on red blood cell parameters, reticulocyte indices and plasma erythropoietin (EPO) concentrations were evaluated. Molidustat treatment was stopped when hematocrit (HCT) exceeded 60%. RESULTS: Compared to placebo, a significant increase in mean HCT was evident starting on Day 14 (Group 2:54.4% vs 40.3%, P < .001, 95% confidence interval [CI] for the difference [8.95-19.28]; Group 3:61.2% vs 40.3%, P < .001, 95% CI [15.48-26.43]) and remained significantly higher for the entire treatment period. In molidustat-treated groups, HCT exceeded 60% on Day 21 (Group 2) and Day 14 (Group 3). Mean HCT in molidustat-treated cats returned to within the reference range (29%-45%) after Day 56 and was numerically comparable to placebo from Day 70 onwards. Red blood cell count and hemoglobin concentrations followed a similar pattern as HCT. Mean EPO concentrations significantly increased after molidustat administration on all assessment days. Molidustat treatments were well tolerated. CONCLUSIONS AND CLINICAL IMPORTANCE: Marked erythropoietic effects were identified after daily administration of molidustat to healthy cats and additional studies are warranted to evaluate the effects in anemic cats.


Asunto(s)
Anemia , Enfermedades de los Gatos , Animales , Gatos , Anemia/veterinaria , Enfermedades de los Gatos/tratamiento farmacológico , Enfermedades de los Gatos/inducido químicamente , Eritropoyesis , Pirazoles , Triazoles/farmacología
2.
J Pept Sci ; 29(12): e3530, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37423610

RESUMEN

The peptide hormone adrenomedullin (ADM) consists of 52 amino acids with a disulfide bond and an amidated C-terminus. Due to the vasodilatory and cardioprotective effects, the agonistic activity of the peptide on the adrenomedullin 1 receptor (AM1 R) is of high pharmacological interest. However, the wild-type peptide shows low metabolic stability leading to rapid degradation in the cardiovascular system. Previous work by our group has identified proteolytic cleavage sites and demonstrated stabilization of ADM by lipidation, cyclization, and N-methylation. Nevertheless, these ADM analogs showed reduced activity and subtype selectivity toward the closely related calcitonin gene-related peptide receptor (CGRPR). Here, we report on the rational development of ADM derivatives with increased proteolytic stability and high receptor selectivity. Stabilizing motifs, including lactamization and lipidation, were evaluated regarding AM1 R and CGRPR activation. Furthermore, the central DKDK motif of the peptide was replaced by oligoethylene glycol linkers. The modified peptides were synthesized by Fmoc/t-Bu solid-phase peptide synthesis and receptor activation of AM1 R and CGRPR was measured by cAMP reporter gene assay. Peptide stability was tested in human blood plasma and porcine liver homogenate and analyzed by RP-HPLC and MALDI-ToF mass spectrometry. Combination of the favorable lactam, lipidation, ethylene glycol linker, and previously described disulfide mimetic resulted in highly stabilized analogs with a plasma half-life of more than 144 h. The compounds display excellent AM1 R activity and wild-type-like selectivity toward CGRPR. Additionally, dose-dependent vasodilatory effects of the ADM derivatives lasted for several hours in rodents. Thus, we successfully developed an ADM analog with long-term in vivo activity.


Asunto(s)
Adrenomedulina , Disulfuros , Humanos , Animales , Porcinos , Adrenomedulina/genética , Receptores de Adrenomedulina/metabolismo
3.
ChemMedChem ; 13(10): 988-1003, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29485740

RESUMEN

Small-molecule inhibitors of hypoxia-inducible factor prolyl hydroxylases (HIF-PHs) are currently under clinical development as novel treatment options for chronic kidney disease (CKD) associated anemia. Inhibition of HIF-PH mimics hypoxia and leads to increased erythropoietin (EPO) expression and subsequently increased erythropoiesis. Herein we describe the discovery, synthesis, structure-activity relationship (SAR), and proposed binding mode of novel 2,4-diheteroaryl-1,2-dihydro-3H-pyrazol-3-ones as orally bioavailable HIF-PH inhibitors for the treatment of anemia. High-throughput screening of our corporate compound library identified BAY-908 as a promising hit. The lead optimization program then resulted in the identification of molidustat (BAY 85-3934), a novel small-molecule oral HIF-PH inhibitor. Molidustat is currently being investigated in clinical phase III trials as molidustat sodium for the treatment of anemia in patients with CKD.


Asunto(s)
Anemia/tratamiento farmacológico , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Enfermedades Renales/complicaciones , Pirazoles/farmacología , Triazoles/farmacología , Anemia/etiología , Animales , Sitios de Unión , Línea Celular Tumoral , Descubrimiento de Drogas , Humanos , Ratones , Estructura Molecular , Unión Proteica , Conformación Proteica , Pirazoles/uso terapéutico , Relación Estructura-Actividad , Triazoles/uso terapéutico
4.
PLoS One ; 12(10): e0186979, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29073196

RESUMEN

Plasma levels of FGF23 are increased in patients with chronic kidney disease. Beside its role in phosphate homeostasis, iron deficiency and anemia are associated with increased FGF23 plasma levels. Recently, FGF23 plasma levels were shown to be increased in mice after treatment with hypoxia inducible factor-proline hydroxylase (HIF-PH) inhibitors which are strong inducers of erythropoietin and erythropoiesis and are known to modulate iron uptake and availability. Therefore we investigated a potential context between expression of FGF23 and stimulation of erythropoiesis using a HIF-PH inhibitor and erythropoietin in rats. FGF23 plasma levels are induced at peak levels 2 h after intravenous injection of recombinant human Erythropoietin (rhEPO). Likewise induction of endogenous EPO using a HIF-PH inhibitor (BAY 85-3934) is followed by an increase of FGF23 plasma levels. In contrast to rhEPO the HIF-PH inhibitor induces lower peak levels of FGF23 applying equivalent hematopoietic doses. Bone and bone marrow were identified as sources of EPO-induced FGF23. Immediate induction of FGF23 mRNA was also detected in EPO receptor positive murine hematopoietic BAF3 cells after treatment with rhEPO but not after treatment with the HIF-PH inhibitor. Pretreatment of mice with a neutralizing anti-EPO antibody abrogated FGF23 induction by the HIF-PH inhibitor. Thus, direct impact on FGF23 expression by HIF-PH inhibition in vivo via hypoxia mimicking and modulation of iron metabolism appears unlikely. Collectively, the findings point to an EPO dependent regulation pathway of FGF23 gene expression which might be important in the context of erythropoiesis stimulating therapies in patients with renal anemia.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Eritropoyetina/farmacología , Factores de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Animales , Factor-23 de Crecimiento de Fibroblastos , Humanos , Masculino , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
5.
PLoS One ; 9(11): e111838, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25392999

RESUMEN

Oxygen sensing by hypoxia-inducible factor prolyl hydroxylases (HIF-PHs) is the dominant regulatory mechanism of erythropoietin (EPO) expression. In chronic kidney disease (CKD), impaired EPO expression causes anemia, which can be treated by supplementation with recombinant human EPO (rhEPO). However, treatment can result in rhEPO levels greatly exceeding the normal physiological range for endogenous EPO, and there is evidence that this contributes to hypertension in patients with CKD. Mimicking hypoxia by inhibiting HIF-PHs, thereby stabilizing HIF, is a novel treatment concept for restoring endogenous EPO production. HIF stabilization by oral administration of the HIF-PH inhibitor BAY 85-3934 (molidustat) resulted in dose-dependent production of EPO in healthy Wistar rats and cynomolgus monkeys. In repeat oral dosing of BAY 85-3934, hemoglobin levels were increased compared with animals that received vehicle, while endogenous EPO remained within the normal physiological range. BAY 85-3934 therapy was also effective in the treatment of renal anemia in rats with impaired kidney function and, unlike treatment with rhEPO, resulted in normalization of hypertensive blood pressure in a rat model of CKD. Notably, unlike treatment with the antihypertensive enalapril, the blood pressure normalization was achieved without a compensatory activation of the renin-angiotensin system. Thus, BAY 85-3934 may provide an approach to the treatment of anemia in patients with CKD, without the increased risk of adverse cardiovascular effects seen for patients treated with rhEPO. Clinical studies are ongoing to investigate the effects of BAY 85-3934 therapy in patients with renal anemia.


Asunto(s)
Anemia/tratamiento farmacológico , Eritropoyetina/biosíntesis , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Pirazoles/farmacología , Triazoles/farmacología , Anemia/etiología , Animales , Eritropoyetina/genética , Femenino , Humanos , Macaca fascicularis , Masculino , Pirazoles/uso terapéutico , Ratas , Ratas Wistar , Insuficiencia Renal Crónica/complicaciones , Triazoles/uso terapéutico , Regulación hacia Arriba
6.
Mol Cancer Res ; 11(11): 1337-48, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24048703

RESUMEN

UNLABELLED: Solid tumor growth is intimately associated with angiogenesis, a process that is efficiently triggered by hypoxia. Therefore, oxygen-sensitive signaling pathways are thought to play a critical role in tumor angiogenesis and progression. Here, the function of prolyl hydroxylase-4 (PHD4), a relative of the prolyl hydroxylase domain proteins 1-3 that promote the degradation of hypoxia-inducible factors (HIF), was interrogated. To test the hypothesis that PHD4 might inhibit tumor angiogenesis, it was overexpressed in osteosarcoma cells, and unexpectedly, this manipulation led to increased tumor blood vessel density. However, the newly formed blood vessels were smaller than normal and appeared to be partially nonfunctional, as indicated by poor vessel perfusion. PHD4 overexpression in tumor cells stimulated the expression of TGF-α, which was necessary and sufficient to promote angiogenic sprouting of endothelial cells. On the other hand, PHD4 overexpression reduced HIF-2α protein levels, which in turn inhibited in vivo tumor growth. Combined, elevated PHD4 levels deregulate angiogenesis via increased TGF-α expression in vitro and in vivo. These data support the hypothesis that tumor growth can be uncoupled from vessel density and that the individual PHD family members exert distinct functions in tumors. IMPLICATIONS: PHD4 influences tumor growth and vascularization through discrete mechanisms and molecular pathways that likely have therapeutic potential.


Asunto(s)
Neovascularización Patológica/metabolismo , Osteosarcoma/patología , Prolil Hidroxilasas/metabolismo , Sarcoma Experimental/patología , Factor de Crecimiento Transformador alfa/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Proliferación Celular , Células Endoteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C3H , Neovascularización Patológica/genética , Osteosarcoma/metabolismo , Prolil Hidroxilasas/genética , Sarcoma Experimental/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador alfa/genética
7.
Cancer Med ; 2(5): 611-24, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24403227

RESUMEN

The activation of the transcription factor hypoxia-inducible factor-1 (HIF-1) plays an essential role in tumor development, tumor progression, and resistance to chemo- and radiotherapy. In order to identify compounds targeting the HIF pathway, a small molecule library was screened using a luciferase-driven HIF-1 reporter cell line under hypoxia. The high-throughput screening led to the identification of a class of aminoalkyl-substituted compounds that inhibited hypoxia-induced HIF-1 target gene expression in human lung cancer cell lines at low nanomolar concentrations. Lead structure BAY 87-2243 was found to inhibit HIF-1α and HIF-2α protein accumulation under hypoxic conditions in non-small cell lung cancer (NSCLC) cell line H460 but had no effect on HIF-1α protein levels induced by the hypoxia mimetics desferrioxamine or cobalt chloride. BAY 87-2243 had no effect on HIF target gene expression levels in RCC4 cells lacking Von Hippel-Lindau (VHL) activity nor did the compound affect the activity of HIF prolyl hydroxylase-2. Antitumor activity of BAY 87-2243, suppression of HIF-1α protein levels, and reduction of HIF-1 target gene expression in vivo were demonstrated in a H460 xenograft model. BAY 87-2243 did not inhibit cell proliferation under standard conditions. However under glucose depletion, a condition favoring mitochondrial ATP generation as energy source, BAY 87-2243 inhibited cell proliferation in the nanomolar range. Further experiments revealed that BAY 87-2243 inhibits mitochondrial complex I activity but has no effect on complex III activity. Interference with mitochondrial function to reduce hypoxia-induced HIF-1 activity in tumors might be an interesting therapeutic approach to overcome chemo- and radiotherapy-resistance of hypoxic tumors.


Asunto(s)
Complejo I de Transporte de Electrón/antagonistas & inhibidores , Neoplasias Pulmonares/metabolismo , Oxadiazoles/farmacología , Pirazoles/farmacología , Animales , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/biosíntesis , Anhidrasas Carbónicas/genética , Hipoxia de la Célula/genética , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/métodos , Complejo I de Transporte de Electrón/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Relacionados con las Neoplasias , Genes Reporteros , Humanos , Factor 1 Inducible por Hipoxia/biosíntesis , Factor 1 Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Terapia Molecular Dirigida/métodos , Oxadiazoles/administración & dosificación , Oxadiazoles/sangre , Oxadiazoles/uso terapéutico , Pirazoles/administración & dosificación , Pirazoles/sangre , Pirazoles/uso terapéutico , ARN Interferente Pequeño/genética , Bibliotecas de Moléculas Pequeñas , Carga Tumoral/efectos de los fármacos , Células Tumorales Cultivadas , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
J Med Chem ; 53(15): 5629-38, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20684604

RESUMEN

Selective inhibitors of Jumonji domain-containing protein (JMJD) histone demethylases are candidate anticancer agents as well as potential tools for elucidating the biological functions of JMJDs. On the basis of the crystal structure of JMJD2A and a homology model of JMJD2C, we designed and prepared a series of hydroxamate analogues bearing a tertiary amine. Enzyme assays using JMJD2C, JMJD2A, and prolyl hydroxylases revealed that hydroxamate analogue 8 is a potent and selective JMJD2 inhibitor, showing 500-fold greater JMJD2C-inhibitory activity and more than 9100-fold greater JMJD2C-selectivity compared with the lead compound N-oxalylglycine 2. Compounds 17 and 18, prodrugs of compound 8, each showed synergistic growth inhibition of cancer cells in combination with an inhibitor of lysine-specific demethylase 1 (LSD1). These findings suggest that combination treatment with JMJD2 inhibitors and LSD1 inhibitors may represent a novel strategy for anticancer chemotherapy.


Asunto(s)
Antineoplásicos/síntesis química , Ácidos Hidroxámicos/síntesis química , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Profármacos/síntesis química , beta-Alanina/análogos & derivados , Aminoácidos Dicarboxílicos/química , Antineoplásicos/química , Antineoplásicos/farmacología , Dominio Catalítico , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Histona Demetilasas/antagonistas & inhibidores , Humanos , Ácidos Hidroxámicos/química , Ácidos Hidroxámicos/farmacología , Histona Demetilasas con Dominio de Jumonji/química , Modelos Moleculares , Estructura Molecular , Profármacos/química , Profármacos/farmacología , Relación Estructura-Actividad , beta-Alanina/síntesis química , beta-Alanina/química , beta-Alanina/farmacología
9.
Cancer Res ; 70(1): 357-66, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20028863

RESUMEN

Adaptive responses to hypoxia in tumors are transcriptionally regulated by the hypoxia inducible factors (HIF-1alpha/HIF-2alpha), which are tightly controlled by the HIF-prolyl hydroxylases (PHD). Hypoxia induces expression of the PHD2 and PHD3 proteins in tumors but the pathobiological significance of these events is uncertain. Here, we show that PHD2 and PHD3 induction acts within a negative feedback loop to limit the hypoxic HIF response. In glioblastomas, PHD2 and PHD3 are hypoxia-inducible in vitro and expressed in hypoxic areas of tumors in vivo. Comparison with other PHDs revealed distinct cytoplasmatic and nuclear localization patterns of PHD2 and PHD3. Gain and loss of function experiments defined PHD2 and PHD3 as HIF target genes that remained operative even at low oxygen concentrations. We found that increased PHD levels could compensate for reduced oxygen availability to regulate the HIF response. This negative feedback loop protected tumor cells against hypoxia-induced cell death, functionally implicating this pathway in the control of the tumor-suppressive components of the HIF system in glioblastoma. Moreover, PHD inhibition facilitated cell death induction by staurosporine or tumor necrosis factor-related apoptosis-inducing ligand, hinting at a more general protective role of PHD in the regulation of cell viability. In summary, our findings recognize the PHD/HIF regulatory axis as a novel therapeutic target to disable a tumor's ability to adjust to hypoxic conditions and control cell survival, helping to potentially overcome therapeutic cell death resistance in glioblastomas.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Dioxigenasas/metabolismo , Glioma/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Western Blotting , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Retroalimentación Fisiológica/fisiología , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Inmunohistoquímica , Hibridación Fluorescente in Situ , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
10.
J Biol Chem ; 283(46): 31745-53, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-18776187

RESUMEN

The abundance of the transcription factor hypoxia-inducible factor is regulated through hydroxylation of its alpha-subunits by a family of prolyl-hydroxylases (PHD1-3). Enzymatic activity of these PHDs is O2-dependent, which enables PHDs to act as cellular O2 sensor enzymes. Herein we studied endogenous PHD activity that was induced in cells grown under hypoxia or in the presence of nitric oxide. Under such conditions nuclear extracts contained much higher PHD activity than the respective cytoplasmic extracts. Although PHD1-3 were abundant in both compartments, knockdown experiments for each isoenzyme revealed that nuclear PHD activity was only due to PHD2. Maximal PHD2 activity was found between 120 and 210 microm O2. PHD2 activity was strongly decreased below 100 microm O2 with a half-maximum activity at 53 +/- 13 microm O2 for the cytosolic and 54 +/- 10 microm O2 for nuclear PHD2 matching the physiological O2 concentration within most cells. Our data suggest a role for PHD2 as a decisive oxygen sensor of the hypoxia-inducible factor degradation pathway within the cell nucleus.


Asunto(s)
Núcleo Celular/metabolismo , Óxido Nítrico/metabolismo , Oxígeno/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Hipoxia de la Célula , Línea Celular , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Humanos , Procolágeno-Prolina Dioxigenasa/genética
11.
J Pineal Res ; 45(4): 489-96, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18673420

RESUMEN

N-nitrosomelatonin (NOMela) is well-known for its capabilities of transnitrosating nucleophiles such as thiols and ascorbate, thereby generating nitric oxide (NO)-releasing compounds. It is unknown, however, whether NOMela can be successfully applied as a precursor of NO in a complex biological environment like a cell culture system. NO donors may be useful to induce the transcription factor hypoxia inducible factor 1 (HIF-1), which coordinates the protection of cells and tissues from the lack of oxygen (hypoxia). In this study, the effects of NOMela in an in vitro cell-free assay [NO-release, inhibition of prolylhydroxylase1 (PHD1)] and in living cells (upregulation of HIF-1, reduction of HIF-1 hydroxylation, upregulation of the HIF-1-target gene PHD2) were compared with those of the frequently applied NO donor S-nitrosoglutathione (GSNO) under normoxic and hypoxic conditions. In contrast to GSNO, NOMela released NO in a predictable manner and this release in vitro was found to be independent of the composition of the buffer system. The NOMela-mediated effects in oxygenated cells were in all cases comparable to the hypoxic response, whereas unphysiological strong effects were observed with GSNO. Probably, because of the antioxidative power of the NOMela-dependent formation of melatonin, cells were completely protected against the attack of reactive nitrogen oxygen species, which are generated by autoxidation of NO. In conclusion, NOMela had to be an excellent NO precursor for cells in culture and potentially tissues.


Asunto(s)
Melatonina/análogos & derivados , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/metabolismo , Compuestos Nitrosos/farmacología , Antioxidantes , Western Blotting , Tampones (Química) , Técnicas de Cultivo de Célula , Hipoxia de la Célula , Glutatión/farmacología , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Melatonina/farmacología , Procolágeno-Prolina Dioxigenasa/metabolismo , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , S-Nitrosoglutatión/farmacología , Transfección
12.
Methods Enzymol ; 435: 43-60, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17998048

RESUMEN

The prolyl-4-hydroxylase domain (PHD) oxygen sensor proteins hydroxylate hypoxia-inducible transcription factor (HIF)-alpha (alpha) subunits, leading to their subsequent ubiquitinylation and degradation. Since oxygen is a necessary cosubstrate, a reduction in oxygen availability (hypoxia) decreases PHD activity and, subsequently, HIF-alpha hydroxylation. Non-hydroxylated HIF-alpha cannot be bound by the ubiquitin ligase von Hippel-Lindau tumor suppressor protein (pVHL), and HIF-alpha proteins thus become stabilized. HIF-alpha then heterodimerizes with HIF-beta (beta) to form the functionally active HIF transcription factor complex, which targets approximately 200 genes involved in adaptation to hypoxia. The three HIF-alpha PHDs are of a different nature compared with the prototype collagen prolyl-4-hydroxylase, which hydroxylates a mass protein rather than a rare transcription factor. Thus, novel assays had to be developed to express and purify functionally active PHDs and to measure PHD activity in vitro. A need also exists for such assays to functionally distinguish the three different PHDs in terms of substrate specificity and drug function. We provide a detailed description of the expression and purification of the PHDs as well as of an HIF-alpha-dependent and a HIF-alpha-independent PHD assay.


Asunto(s)
Procolágeno-Prolina Dioxigenasa/biosíntesis , Procolágeno-Prolina Dioxigenasa/química , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/química , Cromatografía en Capa Delgada , Descarboxilación , Glutaratos/química , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Oxidación-Reducción , Oxígeno/química , Oxígeno/metabolismo , Péptidos/química , Péptidos/genética , Procolágeno-Prolina Dioxigenasa/genética , Estructura Terciaria de Proteína , Proteínas Recombinantes/aislamiento & purificación , Extractos de Tejidos/química
13.
Blood ; 107(2): 584-90, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16189264

RESUMEN

Hypoxia-inducible factors (HIFs) are transcriptional regulators that mediate the cellular response to low oxygen levels. By stimulating the expression of angiogenic growth factors such as vascular endothelial growth factor (VEGF), they trigger the neovascularization of tissues under physiologic and pathologic conditions. Here, we have investigated the endothelial cell-autonomous HIF function in blood vessel growth and development by expressing a dominant-negative HIF mutant (HIFdn) that inhibits the transcriptional responses mediated by both HIF-1 and HIF-2, specifically in endothelial cells of transgenic mice. HIFdn transgenic embryos were growth retarded and died around E11.5. Primitive vascular networks were established, but vascular remodeling in the yolk sac and in the embryo proper was defective, and vascular sprouts failed to invade the neuroepithelium. In addition, heart looping was incomplete, and the ventricles of the heart were thin-walled and lacked trabeculation. Similar cardiovascular defects have been observed in Tie2-deficient mouse embryos. Consistently, HIFdn transgenic embryos expressed reduced levels of the endothelial angiopoietin receptor, Tie-2, whereas other endothelial markers, such as PECAM-1, Tie-1, and VE-cadherin were not affected. These results show that HIFs in endothelial cells are essential for embryonic heart and blood vessel development and control angiogenesis and vascular remodeling.


Asunto(s)
Vasos Sanguíneos , Sistema Cardiovascular/embriología , Sistema Cardiovascular/metabolismo , Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Animales , Antígenos CD , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Cadherinas/metabolismo , Desarrollo Embrionario , Endotelio Vascular/citología , Genes Dominantes , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones Transgénicos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor TIE-2/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Cancer Cell ; 8(2): 131-41, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16098466

RESUMEN

The hypoxia-inducible transcription factors HIF-1alpha and HIF-2alpha are activated in hypoxic tumor regions. However, their role in tumorigenesis remains controversial, as tumor growth promoter and suppressor activities have been ascribed to HIF-1alpha, while the role of HIF-2alpha remains largely unknown. Here, we show that overexpression of HIF-2alpha in rat glioma tumors enhances angiogenesis but reduces growth of these tumors, in part by increasing tumor cell apoptosis. Moreover, siRNA knockdown of HIF-2alpha reduced apoptosis in hypoxic human malignant glioblastoma cells. Furthermore, inhibition of HIF by overexpression of a dominant-negative HIF transgene in glioma cells or HIF-2alpha deficiency in teratomas reduced vascularization but accelerated growth of these tumor types. These findings urge careful consideration of using HIF inhibitors as cancer therapeutic strategies.


Asunto(s)
Glioma/irrigación sanguínea , Neovascularización Patológica , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Glioma/genética , Glioma/metabolismo , Humanos , Ratones , Neoplasias Neuroepiteliales/irrigación sanguínea , Neoplasias Neuroepiteliales/genética , Neoplasias Neuroepiteliales/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , ARN Interferente Pequeño/genética , Ratas , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Activación Transcripcional , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
15.
Blood ; 105(12): 4613-9, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15741220

RESUMEN

Cellular oxygen partial pressure is sensed by a family of prolyl-4-hydroxylase domain (PHD) enzymes that modify hypoxia-inducible factor (HIF)alpha subunits. Upon hydroxylation under normoxic conditions, HIFalpha is bound by the von Hippel-Lindau tumor suppressor protein and targeted for proteasomal destruction. Since PHD activity is dependent on oxygen and ferrous iron, HIF-1 mediates not only oxygen- but also iron-regulated transcriptional gene expression. Here we show that copper (CuCl(2)) stabilizes nuclear HIF-1alpha under normoxic conditions, resulting in hypoxia-response element (HRE)-dependent reporter gene expression. In in vitro hydroxylation assays CuCl(2) inhibited prolyl-4-hydroxylation independently of the iron concentration. Ceruloplasmin, the main copper transport protein in the plasma and a known HIF-1 target in vitro, was also induced in vivo in the liver of hypoxic mice. Both hypoxia and CuCl(2) increased ceruloplasmin (as well as vascular endothelial growth factor [VEGF] and glucose transporter 1 [Glut-1]) mRNA levels in hepatoma cells, which was due to transcriptional induction of the ceruloplasmin gene (CP) promoter. In conclusion, our data suggest that PHD/HIF/HRE-dependent gene regulation can serve as a sensory system not only for oxygen and iron but also for copper metabolism, regulating the oxygen-, iron- and copper-binding transport proteins hemoglobin, transferrin, and ceruloplasmin, respectively.


Asunto(s)
Ceruloplasmina/metabolismo , Cobre/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Hierro/metabolismo , Proteínas Nucleares/metabolismo , Oxígeno/metabolismo , Factores de Transcripción/metabolismo , Animales , Células CHO , Carcinoma Hepatocelular/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Proliferación Celular , Colorantes/farmacología , Cricetinae , Relación Dosis-Respuesta a Droga , Genes Reporteros , Transportador de Glucosa de Tipo 1 , Células HeLa , Humanos , Hipoxia , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Immunoblotting , Hígado/metabolismo , Luciferasas/metabolismo , Ratones , Microscopía Fluorescente , Proteínas de Transporte de Monosacáridos/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Sales de Tetrazolio/farmacología , Tiazoles/farmacología , Transcripción Genética , Transfección , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Anal Biochem ; 330(1): 74-80, 2004 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15183764

RESUMEN

The transcriptional activation of hypoxia-inducible genes is essential for the adaptation of mammalian tissues to oxygen deficiency. The hypoxia-inducible transcription factor (HIF) is a cellular switch for the up-regulation of these genes during hypoxia. Under normoxia, HIFs are hydroxylated on conserved prolyl residues by a recently discovered family of HIF prolyl hydroxylases (HIF-PHD1-3). Hydroxylated HIF specifically interacts with the von Hippel-Lindau protein-elongin B-elongin C complex (VBC) which leads to ubiquitination and subsequent proteasomal degradation of HIF. We developed a nonradioactive microtiter plate assay based on the interaction of hydroxylated HIF with VBC which enabled us to detect hydroxylated HIF in the nanomolar concentration range. A biotinylated HIF peptide substrate was bound to a streptavidin-coated microtiter plate and hydroxylated with the HIF-PHD3 isoenzyme. Recombinant VBC complex with a thioredoxin (Trx) tag was purified from Escherichia coli and bound to the hydroxylated HIF peptide. The interaction between VBC and hydroxylated HIF was detected by using an anti-thioredoxin antibody.


Asunto(s)
Proteínas de Unión al ADN/análisis , Complejos Multiproteicos/química , Proteínas Nucleares/análisis , Péptidos/análisis , Procolágeno-Prolina Dioxigenasa/química , Factores de Transcripción/análisis , Factores de Transcripción/química , Anticuerpos Monoclonales/química , Bioensayo/métodos , Dioxigenasas , Elonguina , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Complejos Multiproteicos/genética , Péptidos/química , Procolágeno-Prolina Dioxigenasa/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Sensibilidad y Especificidad , Estreptavidina/química , Tiorredoxinas/química , Tiorredoxinas/genética , Factores de Transcripción/genética
17.
J Biol Chem ; 278(9): 7520-30, 2003 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-12464608

RESUMEN

Interactions between Ets family members and a variety of other transcription factors serve important functions during development and differentiation processes, e.g. in the hematopoietic system. Here we show that the endothelial basic helix-loop-helix PAS domain transcription factor, hypoxia-inducible factor-2alpha (HIF-2alpha) (but not its close relative HIF-1alpha), cooperates with Ets-1 in activating transcription of the vascular endothelial growth factor receptor-2 (VEGF-2) gene (Flk-1). The receptor tyrosine kinase Flk-1 is indispensable for angiogenesis, and its expression is closely regulated during development. Consistent with the hypothesis that HIF-2alpha controls the expression of Flk-1 in vivo, we show here that HIF-2alpha and Flk-1 are co-regulated in postnatal mouse brain capillaries. A tandem HIF-2alpha/Ets binding site was identified within the Flk-1 promoter that acted as a strong enhancer element. Based on the analysis of transgenic mouse embryos, these motifs are essential for endothelial cell-specific reporter gene expression. A single HIF-2alpha/Ets element conferred strong cooperative induction by HIF-2alpha and Ets-1 when fused to a heterologous promoter and was most active in endothelial cells. The physical interaction of HIF-2alpha with Ets-1 was demonstrated and localized to the HIF-2alpha carboxyl terminus and the autoinhibitory exon VII domain of Ets-1, respectively. The deletion of the DNA binding and carboxyl-terminal transactivation domains of HIF-2alpha, respectively, created dominant negative mutants that suppressed transactivation by the wild type protein and failed to synergize with Ets-1. These results suggest that the interaction between HIF-2alpha and endothelial Ets factors is required for the full transcriptional activation of Flk-1 in endothelial cells and may therefore represent a future target for the manipulation of angiogenesis.


Asunto(s)
Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Edad , Secuencias de Aminoácidos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Sitios de Unión , Western Blotting , Diferenciación Celular , División Celular , Línea Celular , Núcleo Celular/metabolismo , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/metabolismo , Endotelio/citología , Endotelio/metabolismo , Exones , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Vectores Genéticos , Glutatión Transferasa/metabolismo , Humanos , Inmunohistoquímica , Hibridación in Situ , Luciferasas/metabolismo , Ratones , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Neovascularización Patológica , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Proteína Proto-Oncogénica c-ets-1 , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas c-ets , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Transactivadores/química , Factores de Transcripción/química , Transfección , Transgenes
18.
Biochem Biophys Res Commun ; 296(2): 343-9, 2002 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-12163023

RESUMEN

Hypoxia-inducible transcription factors (HIFs) are important for transcriptional adaptation to hypoxia. Availability of HIFs is regulated via posttranslational modification of their alpha subunits (HIF-1alpha and HIF-2alpha). Under normoxia, two highly conserved proline residues within the oxygen-dependent degradation domain (ODDD) are hydroxylated by oxoglutarate-dependent proline 4-hydroxylases EGLN1-3. Hydroxylated HIF-alpha interacts with the pVHL-E3 ubiquitin ligase complex and, subsequently, is degraded via the proteasomal pathway. We identified a novel putative proline 4-hydroxylase, PH-4, with an aminoterminal EF-hand motif and a carboxyterminal catalytic domain, which was highly expressed in most organs, and-unlike EGLNs which localize to the cytoplasm and nucleus-was associated with the endoplasmic reticulum. Like EGLNs, PH-4 overexpressed in cellular reporter assays suppressed the HIF transactivation activity, dependent on the consensus ODDD proline residues. Suppression of transactivation was correlated with decrease of cellular contents of HIF. Thus, PH-4 might be related to cellular oxygen sensing.


Asunto(s)
Oxígeno/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Genes Reporteros , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Datos de Secuencia Molecular , Procolágeno-Prolina Dioxigenasa/química , Procolágeno-Prolina Dioxigenasa/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Distribución Tisular , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...