Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Development ; 145(12)2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29777010

RESUMEN

Normal development requires tight regulation of cell proliferation and cell death. Here, we have investigated these control mechanisms in the hyaloid vessels, a temporary vascular network in the mammalian eye that requires a Wnt/ß-catenin response for scheduled regression. We investigated whether the hyaloid Wnt response was linked to the oncogene Myc, and the cyclin-dependent kinase inhibitor CDKN1A (P21), both established regulators of cell cycle progression and cell death. Our analysis showed that the Wnt pathway co-receptors LRP5 and LRP6 have overlapping activities that mediate the Wnt/ß-catenin signaling in hyaloid vascular endothelial cells (VECs). We also showed that both Myc and Cdkn1a are downstream of the Wnt response and are required for hyaloid regression but for different reasons. Conditional deletion of Myc in VECs suppressed both proliferation and cell death. By contrast, conditional deletion of Cdkn1a resulted in VEC overproliferation that countered the effects of cell death on regression. When combined with analysis of MYC and CDKN1A protein levels, this analysis suggests that a Wnt/ß-catenin and MYC-CDKN1A pathway regulates scheduled hyaloid vessel regression.


Asunto(s)
Apoptosis/fisiología , Proliferación Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Endotelio Vascular/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , beta Catenina/metabolismo , Animales , Línea Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Ojo/irrigación sanguínea , Células HEK293 , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-myc/genética , Vía de Señalización Wnt/fisiología
2.
Proc Natl Acad Sci U S A ; 107(9): 4194-9, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20160075

RESUMEN

Macrophages are required for tissue homeostasis through their role in regulation of the immune response and the resolution of injury. Here we show, using the kidney as a model, that the Wnt pathway ligand Wnt7b is produced by macrophages to stimulate repair and regeneration. When macrophages are inducibly ablated from the injured kidney, the canonical Wnt pathway response in kidney epithelial cells is reduced. Furthermore, when Wnt7b is somatically deleted in macrophages, repair of injury is greatly diminished. Finally, injection of the Wnt pathway regulator Dkk2 enhances the repair process and suggests a therapeutic option. Because Wnt7b is known to stimulate epithelial responses during kidney development, these findings suggest that macrophages are able to rapidly invade an injured tissue and reestablish a developmental program that is beneficial for repair and regeneration.


Asunto(s)
Riñón/fisiología , Macrófagos/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Regeneración , Proteínas Wnt/fisiología , Animales , Secuencia de Bases , Ciclo Celular , Cartilla de ADN , Péptidos y Proteínas de Señalización Intercelular/fisiología , Riñón/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo
3.
Ann Hematol ; 87(1): 11-7, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17712557

RESUMEN

Circulating erythropoietin (EPO) is mainly produced by the kidneys and mediates erythrogenesis in bone marrow and nonhematopoietic cell survival. EPO is also produced in other tissues where it functions as a paracrine. Moreover, the hypoxic induction of EPO is known to be mediated by HIF-1alpha and HIF-2alpha, but it remains obscure as to which of these two mediators mainly contributes to EPO expression. Thus, we designed in vivo experiments to evaluate the contributions made by HIF-1alpha and HIF-2alpha to EPO expression. In mice exposed to mild whole body hypoxia, HIF-1alpha and HIF-2alpha were both induced in all tissues examined. However, EPO mRNA was expressed in kidney and brain, but not in liver and lung. Likewise, chromatin immunoprecipitation (CHIP) analyses demonstrated that HIF-1alpha or HIF-2alpha binding to the EPO gene increased under hypoxic conditions only in kidney and brain. A comparison of CHIP data and EPO mRNA levels suggested that, during mild hypoxia, renal EPO transcription is induced equally by HIF-1alpha and HIF-2alpha, but that brain EPO is mainly induced during hypoxia by HIF-2alpha. Thus, HIF-1alpha and HIF-2alpha appear to contribute to EPO expression tissue specifically.


Asunto(s)
Eritropoyetina/metabolismo , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factores de Transcripción/metabolismo , Animales , Inmunoprecipitación de Cromatina , Eritropoyetina/genética , Hipoxia/metabolismo , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Especificidad de Órganos , ARN Mensajero/genética , Transcripción Genética/genética
4.
Cancer Res ; 66(12): 6345-52, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16778212

RESUMEN

Hypoxia-inducible factor-1alpha (HIF-1alpha) seems central to tumor growth and progression because it up-regulates genes essential for angiogenesis and the hypoxic adaptation of cancer cells, which is why HIF-1alpha inhibition is viewed as a cancer therapy strategy. Paradoxically, HIF-1alpha also leads to cell cycle arrest or the apoptosis of cancer cells. Thus, the possibility cannot be ruled out that HIF-1alpha inhibitors unlock cell cycle arrest under hypoxic conditions and prevent cell death, which would limit the anticancer effect of HIF-1alpha inhibitors. Previously, we reported on the development of YC-1 as an anticancer agent that inhibits HIF-1alpha. In the present study, we evaluated the effects of YC-1 on hypoxia-induced cell cycle arrest and cell death. It was found that YC-1 does not reverse the antiproliferative effect of hypoxia, but rather that it induces S-phase arrest and apoptosis at therapeutic concentrations that inhibit HIF-1alpha and tumor growth; however, YC-1 did not stimulate cyclic guanosine 3',5'-monophosphate production in this concentration range. It was also found that YC-1 activates the checkpoint kinase-mediated intra-S-phase checkpoint, independently of ataxia-telangiectasia mutated kinase or ataxia-telangiectasia mutated and Rad3-related kinase. These results imply that YC-1 does not promote the regrowth of hypoxic tumors because of its cell cycle arrest effect. Furthermore, YC-1 may induce the combined anticancer effects of HIF-1alpha inhibition and cell growth inhibition.


Asunto(s)
Apoptosis/efectos de los fármacos , Indazoles/farmacología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Fase S/efectos de los fármacos , Apoptosis/fisiología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Activación Enzimática/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Indazoles/farmacocinética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Fase S/fisiología
5.
J Comp Neurol ; 503(6): 779-89, 2007 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-17570498

RESUMEN

We have previously described the distribution pattern of inhibitory synapses on rat jaw-closing (JC) alpha- and gamma-motoneurons. In the present study, we investigated developmental changes in inhibitory synapses on JC motoneurons. We performed a quantitative ultrastructural analysis of putative inhibitory synaptic boutons on JC motoneuron somata by using postembedding immunogold labeling for GABA and glycine. In total, 206, 350, and 497 boutons contacting JC motoneuron somata were analyzed at postnatal days 2 (P2), 11 (P11) and 31 (P31), respectively. The size of the somata increased significantly during postnatal development. The size distribution was bimodal at P31. Mean length of the boutons and percentage of synaptic covering also increased during postnatal development, whereas bouton density did not differ significantly among the three age groups. Synaptic boutons on the somata of JC alpha-motoneurons could be classified into four types: boutons immunoreactive for 1) GABA only, 2) glycine only, 3) both GABA and glycine, and 4) neither GABA nor glycine. There was no developmental change in the proportion of putative inhibitory boutons to the total number of studied boutons. However, the glycine-only boutons increased significantly (15.1% to 27.3%), and the GABA-only boutons decreased significantly (17.7% to 2.6%) during the period from P11 to P31. Our ultrastructural data indicate that the inhibitory synaptic input to JC motoneurons is developmentally regulated and that there is a postnatal switch from GABA to glycine. The postnatal changes revealed in the present study could play an important role in the maturation of the oral motor system.


Asunto(s)
Glicina/metabolismo , Maxilares/inervación , Neuronas Motoras/citología , Terminales Presinápticos/ultraestructura , Nervio Trigémino/crecimiento & desarrollo , Ácido gamma-Aminobutírico/metabolismo , Animales , Inmunohistoquímica , Neuronas Motoras/metabolismo , Terminales Presinápticos/metabolismo , Ratas , Nervio Trigémino/metabolismo
6.
Int J Oncol ; 29(1): 255-60, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16773207

RESUMEN

HIF-1alpha is believed to promote tumor growth and metastasis, and many efforts have been made to develop new anticancer agents based on HIF-1alpha inhibition. YC-1 is a widely used HIF-1alpha inhibitor both in vitro and in vivo, and is being developed as a novel class of anticancer drug. However, little is known about the mechanism by which YC-1 degrades HIF-1alpha. As the first step for understanding the mechanism of action of YC-1, we here identified the HIF-1alpha domain responsible for YC-1-induced protein degradation. YC-1 blocked the HIF-1alpha induction by hypoxia, iron chelation, and proteasomal inhibition and also degraded ectopically expressed HIF-1alpha. In deletion analyses, C-terminal HIF-1alpha was found to be sensitively degraded by YC-1. Using a GFP-fusion method, the YC-1-induced degradation domain was identified as the aa. 720-780 region of HIF-1alpha. We next tested the possible involvement of HDAC7 or OS-9 in YC-1-induced HIF-1alpha degradation. However, their binding to HIF-1alpha was not affected by YC-1, suggesting that they are not involved in the YC-1 action. It is also suggested that YC-1 targets a novel pathway regulating HIF-1alpha stability.


Asunto(s)
Antineoplásicos/farmacología , Expresión Génica/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Indazoles/farmacología , Hipoxia de la Célula , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Proteínas Fluorescentes Verdes/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/química , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Mutación , Estructura Terciaria de Proteína/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección
7.
J Natl Cancer Inst ; 95(7): 516-25, 2003 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-12671019

RESUMEN

BACKGROUND: Hypoxia-inducible factor 1 alpha (HIF-1alpha), a component of HIF-1, is expressed in human tumors and renders cells able to survive and grow under hypoxic (low-oxygen) conditions. YC-1, 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole, an agent developed for circulatory disorders that inhibits platelet aggregation and vascular contraction, inhibits HIF-1 activity in vitro. We tested whether YC-1 inhibits HIF-1 and tumor growth in vivo. METHODS: Hep3B hepatoma, NCI-H87 stomach carcinoma, Caki-1 renal carcinoma, SiHa cervical carcinoma, and SK-N-MC neuroblastoma cells were grown as xenografts in immunodeficient mice (69 mice total). After the tumors were 100-150 mm(3), mice received daily intraperitoneal injections of vehicle or YC-1 (30 microg/g) for 2 weeks. HIF-1 alpha protein levels and vascularity in tumors were assessed by immunohistochemistry, and the expression of HIF-1-inducible genes (vascular endothelial growth factor, aldolase, and enolase) was assessed by reverse transcription-polymerase chain reaction. All statistical tests were two-sided. RESULTS: Compared with tumors from vehicle-treated mice, tumors from YC-1-treated mice were statistically significantly smaller (P<.01 for all comparisons), expressed lower levels of HIF-1 alpha (P<.01 for all comparisons), were less vascularized (P<.01 for all comparisons), and expressed lower levels of HIF-1-inducible genes, regardless of tumor type. CONCLUSIONS: The inhibition of HIF-1 alpha activity in tumors from YC-1-treated mice is associated with blocked angiogenesis and an inhibition of tumor growth. YC-1 has the potential to become the first antiangiogenic anticancer agent to target HIF-1 alpha.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Indazoles/farmacología , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Animales , Carcinoma/tratamiento farmacológico , Carcinoma Hepatocelular/tratamiento farmacológico , Hipoxia de la Célula/efectos de los fármacos , Medios de Cultivo Condicionados , Factores de Crecimiento Endotelial/análisis , Ensayo de Inmunoadsorción Enzimática , Femenino , Gangliósido G(M1)/análisis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Immunoblotting , Péptidos y Proteínas de Señalización Intercelular/análisis , Neoplasias Renales/tratamiento farmacológico , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Linfocinas/análisis , Masculino , Ratones , Ratones SCID , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Neuroblastoma/tratamiento farmacológico , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Pruebas de Precipitina , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/tratamiento farmacológico , Trasplante Heterólogo , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
8.
Cancer Res ; 63(24): 8700-7, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14695184

RESUMEN

Hypoxia-inducible factor-1 (HIF-1), which is present at higher levels in human tumors, plays important roles in tumor promotion. It is composed of HIF-1alpha and HIF-1beta subunits and its activity depends on the amount of HIF-1alpha, which is tightly controlled by cellular oxygen tension. In addition to hypoxia, various nonhypoxic stimuli can stabilize HIF-1alpha in tumor cells, implying that both hypoxic and nonhypoxic stimuli contribute to the overexpression of HIF-1alpha in tumors. On the other hand, phorbol esters such as phorbol-12-myristate-13-acetate (PMA) are known to be potent tumor promoters. Here, we identified a novel HIF-1alpha isoform, which is regulated primarily by PMA. The variant mRNA lacks exon 11 and produces a 785-amino acid isoform (HIF-1alpha(785)) without altering the reading frame and therefore the COOH-terminal transcriptional activity. HIF-1alpha(785) is induced markedly by PMA and heat shock, the latter of which is also known to induce HIF-1alpha. HIF-1alpha(785) escapes from lysine acetylation because of the loss of Lys(532) and was stabilized under normoxic conditions. Its expression was blocked by reducing agents and by a mitogen-activated protein/extracellular signal-regulated kinase-1 inhibitor and enhanced by hydrogen peroxide. In addition, HIF-1alpha(785) overexpression strikingly enhanced tumor growth in vivo. These results suggest that HIF-1alpha(785) is induced by PMA under normoxic conditions via a redox-dependent mitogen-activated protein/extracellular signal-regulated kinase-1 pathway and that it plays an important role in tumor promotion.


Asunto(s)
Carcinógenos/farmacología , Acetato de Tetradecanoilforbol/farmacología , Factores de Transcripción/biosíntesis , Empalme Alternativo , Animales , División Celular/fisiología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Metilación de ADN , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Ratones Desnudos , Isoformas de Proteínas , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Activación Transcripcional , Transfección , Regulación hacia Arriba/efectos de los fármacos
9.
J Exp Med ; 212(9): 1433-48, 2015 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-26261265

RESUMEN

Although the link between inflammation and cancer initiation is well established, its role in metastatic diseases, the primary cause of cancer deaths, has been poorly explored. Our previous studies identified a population of metastasis-associated macrophages (MAMs) recruited to the lung that promote tumor cell seeding and growth. Here we show that FMS-like tyrosine kinase 1 (Flt1, also known as VEGFR1) labels a subset of macrophages in human breast cancers that are significantly enriched in metastatic sites. In mouse models of breast cancer pulmonary metastasis, MAMs uniquely express FLT1. Using several genetic models, we show that macrophage FLT1 signaling is critical for metastasis. FLT1 inhibition does not affect MAM recruitment to metastatic lesions but regulates a set of inflammatory response genes, including colony-stimulating factor 1 (CSF1), a central regulator of macrophage biology. Using a gain-of-function approach, we show that CSF1-mediated autocrine signaling in MAMs is downstream of FLT1 and can restore the tumor-promoting activity of FLT1-inhibited MAMs. Thus, CSF1 is epistatic to FLT1, establishing a link between FLT1 and inflammatory responses within breast tumor metastases. Importantly, FLT1 inhibition reduces tumor metastatic efficiency even after initial seeding, suggesting that these pathways represent therapeutic targets in metastatic disease.


Asunto(s)
Neoplasias de la Mama/metabolismo , Macrófagos/metabolismo , Neoplasias Mamarias Animales/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Comunicación Autocrina/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Humanos , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/patología , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
10.
Cancer Lett ; 207(1): 1-7, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15127725

RESUMEN

Since the first article on YC-1 was published in 1994, it has been popularly used as a pharmacological tool to activate soluble guanylate cyclase and to increase cyclic GMP levels in cultured cells or isolated tissues. In terms of the pharmacological actions of YC-1, previous studies tend to be limited to it inhibition of platelet aggregation and vascular concentration. However, recent studies have demonstrated that YC-1 has versatile pharmacological effects other than the anti-platelet and vasodilatory effects. In particular, two recent reports suggest that YC-1 could be developed as a new class of anticancer agent for rapidly growing solid tumors, because it inhibits hypoxia-inducible factor 1 (HIF-1) activity, and has been reported to halt tumor growth in vivo. We here review the cyclic GMP-dependent and independent pharmacological actions of YC-1, and its anti-HIF-1, anticancer effect.


Asunto(s)
Antineoplásicos/farmacología , Indazoles/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Animales , GMP Cíclico/metabolismo , Guanilato Ciclasa/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias/patología , Neoplasias/terapia , Factores de Transcripción/biosíntesis
11.
Biochem Pharmacol ; 68(6): 1061-9, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15313402

RESUMEN

Hypoxia-inducible factor-1 (HIF-1), which is present at high levels in human tumors, plays crucial roles in tumor promotion by up-regulating its target genes, which are involved in anaerobic energy metabolism, angiogenesis, cell survival, cell invasion, and drug resistance. Therefore, it is apparent that the inhibition of HIF-1 activity may be a strategy for treating cancer. Recently, many efforts to develop new HIF-1-targeting agents have been made by both academic and pharmaceutical industry laboratories. The future success of these efforts will be a new class of HIF-1-targeting anticancer agents, which would improve the prognoses of many cancer patients. This review focuses on the potential of HIF-1 as a target molecule for anticancer therapy, and on possible strategies to inhibit HIF-1 activity. In addition, we introduce YC-1 as a new anti-HIF-1, anticancer agent. Although YC-1 was originally developed as a potential therapeutic agent for thrombosis and hypertension, recent studies demonstrated that YC-1 suppressed HIF-1 activity and vascular endothelial growth factor expression in cancer cells. Moreover, it halted tumor growth in immunodeficient mice without serious toxicity during the treatment period. Thus, we propose that YC-1 is a good lead compound for the development of new anti-HIF-1, anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Indazoles/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción , Adaptación Fisiológica/fisiología , Animales , Antineoplásicos/uso terapéutico , Transformación Celular Neoplásica , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Marcación de Gen , Humanos , Hipoxia , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología
12.
Ann N Y Acad Sci ; 1030: 43-51, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15659779

RESUMEN

Reactive oxygen species are involved in tumor promotion or apoptosis. In assaying prooxidant or antioxidant activities, cyanide has been commonly used as an inhibitor of mitochondrial oxidases, peroxidases, or Cu,Zn-superoxide dismutase, which have an influence on intracellular levels of reactive oxygen species. It has also been used to chemically mimic hypoxia. On the other hand, glycerol has been widely used as a stabilizer of various enzymes. In particular, glycerol is required to maintain the enzymatic activities of membrane-bound NAD(P)H oxidases extracted from surrounding phospholipids. Since both cyanide and glycerol are relatively inert, they have been used concomitantly regardless of any mutual interference. In this study, we demonstrate that a mixture of glycerol and cyanide reduced cytochrome c and nitroblue tetrazolium, both of which are superoxide anion indicators. The mixture also enhanced the production of superoxide anion in the presence of redox-cycling compounds. Superoxide production by the mixture was confirmed by electron spin resonance spectra. Moreover, the mixture induced lipid peroxidation and hemolysis in human erythrocytes. These results suggest that cyanide and glycerol should be used carefully in reaction systems used to measure superoxide production or antioxidant activity. However, sucrose and sodium azide in combination do not produce such artifacts and thus may be used as an alternative.


Asunto(s)
Cianuros/química , Glicerol/química , Especies Reactivas de Oxígeno , Citocromos c/química , Espectroscopía de Resonancia por Spin del Electrón , Eritrocitos/metabolismo , Humanos , Técnicas In Vitro , Peroxidación de Lípido , Nitroazul de Tetrazolio/química , Paraquat/química , Vitamina K 3/química
13.
Cancer Res ; 74(11): 2962-73, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24638982

RESUMEN

Oncogenic targets acting in both tumor cells and tumor stromal cells may offer special therapeutic appeal. Interrogation of the Oncomine database revealed that 52 of 53 human breast carcinomas showed substantial upregulation of WNT family ligand WNT7B. Immunolabeling of human mammary carcinoma showed that WNT7B immunoreactivity was associated with both tumor cells and with tumor-associated macrophages. In the MMTV-PymT mouse model of mammary carcinoma, we found tumor progression relied upon WNT7B produced by myeloid cells in the microenvironment. Wnt7b deletion in myeloid cells reduced the mass and volume of tumors due to a failure in the angiogenic switch. In the tumor overall, there was no change in expression of Wnt/ß-catenin pathway target genes, but in vascular endothelial cells (VEC), expression of these genes was reduced, suggesting that VECs respond to Wnt/ß-catenin signaling. Mechanistic investigations revealed that failure of the angiogenic switch could be attributed to reduced Vegfa mRNA and protein expression in VECs, a source of VEGFA mRNA in the tumor that was limiting in the absence of myeloid WNT7B. We also noted a dramatic reduction in lung metastasis associated with decreased macrophage-mediated tumor cell invasion. Together, these results illustrated the critical role of myeloid WNT7B in tumor progression, acting at the levels of angiogenesis, invasion, and metastasis. We suggest that therapeutic suppression of WNT7B signaling might be advantageous due to targeting multiple aspects of tumor progression.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Neoplasias Mamarias Experimentales/irrigación sanguínea , Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Wnt/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Macrófagos/metabolismo , Macrófagos/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Células Mieloides/patología , Metástasis de la Neoplasia , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
14.
J Comp Neurol ; 518(20): 4134-46, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20878780

RESUMEN

Trigeminal primary afferents that express the transient receptor potential vanilloid 1 (TRPV1) are important for the transmission of orofacial nociception. However, little is known about how the TRPV1-mediated nociceptive information is processed at the first relay nucleus in the central nervous system (CNS). To address this issue, we studied the synaptic connectivity of TRPV1-positive (+) terminals in the rat trigeminal caudal nucleus (Vc) by using electron microscopic immunohistochemistry and analysis of serial thin sections. Whereas the large majority of TRPV1+ terminals made synaptic contacts of an asymmetric type with one or two postsynaptic dendrites, a considerable fraction also participated in complex glomerular synaptic arrangements. A few TRPV1+ terminals received axoaxonic contacts from synaptic endings that contained pleomorphic synaptic vesicles and were immunolabeled for glutamic acid decarboxylase, the synthesizing enzyme for the inhibitory neurotransmitter γ-aminobutyric acid (GABA). We classified the TRPV1+ terminals into an S-type, containing less than five dense-core vesicles (DCVs), and a DCV-type, containing five or more DCVs. The number of postsynaptic dendrites was similar between the two types of terminals; however, whereas axoaxonic contacts were frequent on the S-type, the DCV-type did not receive axoaxonic contacts. In the sensory root of the trigeminal ganglion, TRPV1+ axons were mostly unmyelinated, and a small fraction was small myelinated. These results suggest that the TRPV1-mediated nociceptive information from the orofacial region is processed in a specific manner by two distinct types of synaptic arrangements in the Vc, and that the central input of a few TRPV1+ afferents is presynaptically modulated via a GABA-mediated mechanism.


Asunto(s)
Neuronas Aferentes/ultraestructura , Terminales Presinápticos/ultraestructura , Sinapsis/ultraestructura , Canales Catiónicos TRPV/metabolismo , Núcleo Caudal del Trigémino/ultraestructura , Animales , Inmunohistoquímica , Masculino , Microscopía Inmunoelectrónica , Neuronas Aferentes/química , Ratas , Ratas Sprague-Dawley , Ácido gamma-Aminobutírico/metabolismo
15.
Immunopharmacol Immunotoxicol ; 28(4): 609-20, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17190738

RESUMEN

During transplantation, donor organs or cells are subjected to hypoxia. Hypoxia-inducible factor-1 (HIF-1) is essential for cellular adaptation to hypoxia. Immunosuppressive agents should be used for preventing graft rejection, but of these, rapamycin and cyclosporine A have been reported to inhibit HIF-1. We examined whether or not another important immunosuppressant, FK506, inhibits HIF-1. In contrast to cyclosporine A, FK506 neither inhibits HIF-1alpha expression in 8 different cell lines, nor represses the transcriptional activity of HIF-1. Compared with cyclosporine A, FK506 significantly reduced the apoptotic cell death by hypoxia. FK506 could preserve HIF-1 activity in donor organs subjected to hypoxia.


Asunto(s)
Factor 1 Inducible por Hipoxia/fisiología , Inmunosupresores/farmacología , Tacrolimus/farmacología , Animales , Hipoxia de la Célula/fisiología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ciclosporina/farmacología , Humanos , Factor 1 Inducible por Hipoxia/biosíntesis , Immunoblotting , Microscopía Fluorescente , Transcripción Genética
16.
Blood ; 107(3): 916-23, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16189267

RESUMEN

Amphotericin B (AmB) is widely used for treating severe systemic fungal infections. However, long-term AmB treatment is invariably associated with adverse effects such as anemia. The erythropoietin (EPO) suppression by AmB has been proposed to contribute to the development of anemia. However, the mechanism whereby EPO is suppressed remains obscure. In this study, we investigated the possibility that AmB inhibits the transcription of the EPO gene by inactivating HIF-1, which is a known key transcription factor and regulator of EPO expression. EPO mRNA levels were markedly attenuated by AmB treatment both in rat kidneys and in Hep3B cells. AmB inactivated the transcriptional activity of HIF-1alpha, but did not affect the expression or localization of HIF-1 subunits. Moreover, AmB was found to specifically repress the C-terminal transactivation domain (CAD) of HIF-1alpha, and this repression by AmB required Asn803, a target site of the factor-inhibiting HIF-1 (FIH); moreover, this repressive effect was reversed by FIH inhibitors. Furthermore, AmB stimulated CAD-FIH interaction and inhibited the p300 recruitment by CAD. We propose that this mechanism underlies the unexplained anemia associated with AmB therapy.


Asunto(s)
Anfotericina B/administración & dosificación , Antifúngicos/administración & dosificación , Eritropoyetina/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Anfotericina B/efectos adversos , Anemia/inducido químicamente , Anemia/metabolismo , Animales , Antifúngicos/efectos adversos , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Proteína p300 Asociada a E1A/metabolismo , Humanos , Riñón/metabolismo , Masculino , Micosis/complicaciones , Micosis/tratamiento farmacológico , Micosis/metabolismo , Estructura Terciaria de Proteína , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/metabolismo , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA