Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Sci Rep ; 10(1): 20965, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33262487

RESUMEN

This study was designed to identify whether the position and size of the region of interest (ROI) influence extracellular volume fraction (ECV) measurements. Patients with localized (n = 203) or infiltrative (n = 215) cardiomyopathies and 36 normal controls were enrolled in this study. ECV measurements at 4 different regions, including the anterior, septal, posterior and lateral wall regions, were measured, and all groups were compared. Regional ECV was correlated with the corresponding regional wall thickness. The diagnostic power to differentiate the myocardial abnormalities was evaluated for each myocardial region. ECVs measured using five different ROI sizes within each myocardial region were compared. Our results showed that ECVs varied among the myocardial regions, and this variation was significantly associated with regional wall thicknesses. For the detection of myocardial abnormalities, regional ECV revealed similar results as ECV derived from the whole region except for the anterior region. No significant difference was found in the ECVs measured using the five different ROI sizes. In conclusion, CMR-derived ECVs vary with myocardial region, and this variation is significantly associated with the regional wall thickness. In contrast, the measured size of the ROI has less of an effect on the ECV.


Asunto(s)
Espacio Extracelular/metabolismo , Imagen por Resonancia Magnética , Miocardio/citología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Humanos , Modelos Lineales , Masculino , Persona de Mediana Edad , Variaciones Dependientes del Observador , Curva ROC , Reproducibilidad de los Resultados , Adulto Joven
3.
Int J Pharm ; 538(1-2): 79-86, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29341909

RESUMEN

Deferoxamine (DFO) to treat iron overload (IO) has been limited by toxicity issues and short circulation times and it would be desirable to prolong circulation to improve non-transferrin bound iron (NTBI) chelation. In addition, DFO is currently unable to efficiently target the large pool of iron in the liver and spleen. Nanogel-Deferoxamine conjugates (NG-DFO) can prove useful as a model to investigate the pharmacokinetic (PK) properties and biodistribution (BD) behavior of iron-chelating macromolecules and their overall effect on serum ferritin levels. NG-DFO reduced the cytotoxicity of DFO and significantly reduced cellular ferritin levels in IO macrophages in vitro. PK/BD studies in normal rats revealed that NG-DFO displayed prolonged circulation and preferential accumulation into the liver and spleen. IO mice treated with NG1-DFO presented significantly lower levels of serum ferritin compared to DFO. Total renal and fecal elimination data point to the need to balance prolonged circulation with controlled degradation to accelerate clearance of iron-chelating macromolecules.


Asunto(s)
Deferoxamina/administración & dosificación , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Modelos Biológicos , Animales , Deferoxamina/farmacocinética , Deferoxamina/farmacología , Modelos Animales de Enfermedad , Femenino , Ferritinas/sangre , Células Endoteliales de la Vena Umbilical Humana , Humanos , Quelantes del Hierro/farmacocinética , Quelantes del Hierro/farmacología , Hígado/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Nanopartículas , Ratas , Bazo/metabolismo , Distribución Tisular
4.
ACS Appl Mater Interfaces ; 8(39): 25788-25797, 2016 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-27623539

RESUMEN

Chelation therapy is frequently used to help reduce excess iron in the body, but current chelators such as deferoxamine (DFO) are plagued by short blood circulation times, which necessitates infusions and can cause undesirable toxic side effects in patients. To address these issues, polyrotaxanes (PR) were synthesized by threading α-cyclodextrin (α-CD) onto poly(ethylene glycol) bis(amine) (PEG-BA, MW 3400 g/mol) capped with enzymatically cleavable bulky Z-L phenylalanine (Z-L Phe) moieties. The resulting PR was conjugated to DFO and hydroxypropylated to generate the final polyrotaxane-DFO (hPR-DFO). The iron chelating capability of hPR-DFO was verified by UV-vis absorption spectroscopy and the ability of materials to degrade into smaller CD-conjugated DFO fragments (hCD-DFO) in the presence of the protease was confirmed via gel permeation chromatography. In vitro studies in iron-overloaded macrophages reveal that hPR-DFO can significantly reduce the cytotoxicity of the drug while maintaining its chelation efficacy, and that it is more rapidly endocytosed and trafficked to lysosomes of iron-overloaded cells in comparison to non-iron-overloaded macrophages. In vivo studies indicate that iron-overloaded mice treated with hPR-DFO displayed lower serum ferritin levels (a measure of iron burden in the body) and could eliminate excess iron by both the renal and fecal routes. Moreover, there was no gross evidence of acute toxicological damage to the liver or spleen.


Asunto(s)
Deferoxamina/química , Rotaxanos/química , Animales , Terapia por Quelación , Hierro , Quelantes del Hierro , Sobrecarga de Hierro , Ratones
5.
Toxicol Appl Pharmacol ; 305: 242-249, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27151233

RESUMEN

It is well established that the prototypical aryl hydrocarbon receptor (AHR) agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can both cause and protect against carcinogenesis in non-transgenic rodents. But because these animals almost never develop prostate cancer with old age or after carcinogen exposure, whether AHR activation can affect cancer of the prostate remained unknown. We used animals designed to develop this disease, Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mice, to investigate the potential role of AHR signaling in prostate cancer development. We previously reported that AHR itself has prostate tumor suppressive functions in TRAMP mice; i.e., TRAMP mice in which Ahr was knocked out developed neuroendocrine prostate carcinomas (NEPC) with much greater frequency than did those with both Ahr alleles. In the present study we investigated effects of AHR activation by three different xenobiotics. In utero and lactational TCDD exposure significantly increased NEPC tumor incidence in TRAMP males, while chronic TCDD treatment in adulthood had the opposite effect, a significant reduction in NEPC incidence. Chronic treatment of adult TRAMP mice with the low-toxicity selective AHR modulators indole-3-carbinol or 3,3'-diindolylmethane did not significantly protect against these tumors. Thus, we demonstrate, for the first time, that ligand-dependent activation of the AHR can alter prostate cancer incidence. The nature of the responses depended on the timing of AHR activation and ligand structures.


Asunto(s)
Anticarcinógenos , Carcinógenos , Dibenzodioxinas Policloradas , Efectos Tardíos de la Exposición Prenatal , Animales , Anticarcinógenos/farmacología , Anticarcinógenos/toxicidad , Carcinógenos/farmacología , Carcinógenos/toxicidad , Carcinoma Neuroendocrino/tratamiento farmacológico , Femenino , Lactancia , Masculino , Ratones , Ratones Transgénicos , Dibenzodioxinas Policloradas/farmacología , Dibenzodioxinas Policloradas/toxicidad , Embarazo , Neoplasias de la Próstata/tratamiento farmacológico , Receptores de Hidrocarburo de Aril/genética
6.
J Neurosci ; 35(13): 5293-306, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25834054

RESUMEN

The demyelinating disease multiple sclerosis (MS) has an early inflammatory phase followed by an incurable progressive phase with subdued inflammation and poorly understood neurodegenerative mechanism. In this study, we identified various parallelisms between progressive MS and the dysmyelinating mouse model Shiverer and then genetically deleted a major neuron-specific mitochondrial anchoring protein Syntaphilin (SNPH) from the mouse. Prevailing evidence suggests that deletion of SNPH is harmful in demyelination. Surprisingly, SNPH deletion produces striking benefits in the Shiverer by prolonging survival, reducing cerebellar damage, suppressing oxidative stress, and improving mitochondrial health. In contrast, SNPH deletion does not benefit clinical symptoms in experimental autoimmune encephalomyelitis (EAE), a model for early-phase MS. We propose that deleting mitochondrial anchoring is a novel, specific treatment for progressive MS.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/metabolismo , Esclerosis Múltiple Crónica Progresiva/genética , Animales , Cerebelo/patología , Cerebelo/ultraestructura , Encefalomielitis Autoinmune Experimental/genética , Sustancia Gris/patología , Proteínas de la Membrana , Ratones , Ratones Endogámicos , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/genética , Esclerosis Múltiple Crónica Progresiva/terapia , Proteínas del Tejido Nervioso , Estrés Oxidativo/genética , Análisis de Supervivencia , Sustancia Blanca/patología
7.
Pharm Res ; 31(11): 3106-19, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24848339

RESUMEN

PURPOSE: Although Cu complexes have been investigated as anticancer agents, there has been no description of Cu itself as a cancer killing agent. A stealth liposomal Cu formulation (LpCu) was studied in vitro and in vivo. METHODS: LpCu was evaluated in prostate cancer origin PC-3 cells by a metabolic cytotoxicity assay, by monitoring ROS, and by flow cytometry. LpCu efficacy was evaluated in vivo using intratumoral and intravenous injections into mice bearing PC-3 xenograft tumors. Toxicology was assessed by performing hematological and blood biochemistry assays, and tissue histology and Cu distribution was investigated by elemental analysis. RESULTS: LpCu and free Cu salts displayed similar levels of cell metabolic toxicity and ROS. Flow cytometry indicated that the mechanisms of cell death were both apoptosis and necrosis. Animals injected i.t. with 3.5 mg/kg or i.v. with 3.5 and 7.0 mg/kg LpCu exhibited significant tumor growth inhibition. Kidney and eye were the main organs affected by Cu-mediated toxicities, but spleen and liver were the major organs of Cu deposition. CONCLUSIONS: LpCu was effective at reducing tumor burden in the xenograft prostate cancer model. There was histological evidence of Cu toxicity in kidneys and eyes of animals treated at the maximum tolerated dose of LpCu 7.0 mg/kg.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/farmacocinética , Cobre/farmacología , Cobre/farmacocinética , Liposomas/farmacología , Liposomas/farmacocinética , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Química Farmacéutica/métodos , Modelos Animales de Enfermedad , Xenoinjertos/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Desnudos , Próstata/efectos de los fármacos , Distribución Tisular/fisiología , Trasplante Heterólogo/métodos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Dev Biol ; 376(2): 125-35, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23396188

RESUMEN

Fetal prostate development is initiated by androgens and patterned by androgen dependent and independent signals. How these signals integrate to control epithelial cell differentiation and prostatic bud patterning is not fully understood. To test the role of beta-catenin (Ctnnb1) in this process, we used a genetic approach to conditionally delete or stabilize Ctnnb1 in urogenital sinus (UGS) epithelium from which the prostate derives. Two opposing mechanisms of action were revealed. By deleting Ctnnb1, we found it is required for separation of UGS from cloaca, emergence or maintenance of differentiated UGS basal epithelium and formation of prostatic buds. By genetically inducing a patchy subset of UGS epithelial cells to express excess CTNNB1, we found its excess abundance increases Bmp expression and leads to a global impairment of prostatic bud formation. Addition of NOGGIN partially restores prostatic budding in UGS explants with excess Ctnnb1. These results indicate a requirement for Ctnnb1 in UGS basal epithelial cell differentiation, prostatic bud initiation and bud spacing and suggest some of these actions are mediated in part through activation of BMP signaling.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Regulación del Desarrollo de la Expresión Génica , beta Catenina/biosíntesis , Animales , Tipificación del Cuerpo , Diferenciación Celular , Cruzamientos Genéticos , Células Epiteliales/citología , Eliminación de Gen , Masculino , Ratones , Microscopía Electrónica de Rastreo/métodos , Modelos Genéticos , Próstata/embriología , Transducción de Señal
9.
Dev Dyn ; 239(2): 373-85, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19941349

RESUMEN

The ventral urogenital sinus (UGS) of control male mice has two rows of 3-4 prostatic buds at birth, but how androgens regulate ventral bud (VB) number and patterning is unclear. VBs in both sexes appeared to be a mixture of prostatic and urethral buds. UGSs from Tfm male and antiandrogen (flutamide)-exposed mice had small VBs, suggesting that initiation of some VBs is androgen independent. Tfm male mice are widely considered completely androgen insensitive yet their UGSs were 5alpha-dihydrotestosterone (DHT)- responsive. VBs (6-8) were generally distributed bimodally on the left-right axis at both minimal and normal male androgen signaling. Yet control females and DHT-exposed Tfm males had 13-14 VBs, whose left-right distribution was fairly uniform. These results suggest that VB number and distribution respond biphasically as androgen signaling increases from minimal, and that androgens regulate bud specification. Complete VB agenesis by the selective budding inhibitor 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) required high androgen signaling.


Asunto(s)
Andrógenos/metabolismo , Tipificación del Cuerpo , Feto/metabolismo , Próstata/embriología , Receptores Androgénicos/metabolismo , Antagonistas de Andrógenos , Síndrome de Resistencia Androgénica/metabolismo , Animales , Animales Recién Nacidos , Dihidrotestosterona , Femenino , Flutamida , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Técnicas de Cultivo de Órganos , Dibenzodioxinas Policloradas , Caracteres Sexuales , Transducción de Señal , Teratógenos , Terminología como Asunto , Factores de Transcripción/metabolismo , Uretra/embriología
10.
Toxicol Appl Pharmacol ; 239(1): 80-6, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19523480

RESUMEN

Estrogens play an important role in prostatic development, health, and disease. While estrogen signaling is essential for normal postnatal prostate development, little is known about its prenatal role in control animals. We tested the hypothesis that estrogen signaling is needed for normal male prostatic bud patterning. Budding patterns were examined by scanning electron microscopy of urogenital sinus epithelium from wild-type mice, mice lacking estrogen receptor (ER)alpha, ERbeta, or both, and wild-type mice exposed to the antiestrogen ICI 182,780. Budding phenotypes did not detectably differ among any of these groups, strongly suggesting that estrogen signaling is not needed to establish the prototypical prostatic budding pattern seen in control males. This finding contributes to our understanding of the effects of low-level estrogen exposure on early prostate development. In utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) can greatly alter the pattern in which prostatic buds form and reduce their number. For several reasons, including a prior observation that inhibitory effects of TCDD on prostatic budding in rats depend heavily on the sex of adjacent fetuses, we tested the hypothesis that estrogen signaling is needed for TCDD to disrupt prostatic budding. However, budding did not detectably differ among wild-type mice, or mice lacking ERalpha, ERbeta, or both, that were exposed prenatally to TCDD (5 microg/kg on embryonic day 13.5). Nor did ICI 182,780 detectably affect the response to TCDD. These results strongly suggest that estrogen signaling is not needed for TCDD to inhibit prostatic epithelial budding.


Asunto(s)
Disruptores Endocrinos/toxicidad , Estrógenos/fisiología , Exposición Materna/efectos adversos , Organogénesis/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Próstata/efectos de los fármacos , Animales , Receptor alfa de Estrógeno/biosíntesis , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/biosíntesis , Receptor beta de Estrógeno/genética , Estrógenos/biosíntesis , Femenino , Edad Gestacional , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Embarazo , Próstata/embriología , Próstata/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Biochem Pharmacol ; 77(7): 1151-60, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19166822

RESUMEN

The aryl hydrocarbon receptor (AhR) is a basic-helix-loop-helix transcription factor that binds halogenated aromatic hydrocarbons, polycyclic aromatic hydrocarbons, and endogenous compounds. We previously reported that AhR null (Ahr(-/-)) transgenic adenocarcinoma of the mouse prostate (TRAMP) mice on a C57BL/6J background develop prostate tumors with much greater frequency than AhR wild-type (Ahr(+/+)) TRAMP mice, suggesting that the AhR has tumor suppressor properties. Because AhR signaling pathway inactivation increased susceptibility to prostate tumorigenesis, we tested the hypothesis that a selective AhR modulator (SAhRM), 6-methyl-1,3,8-trichlorodibenzofuran (6-MCDF), can protect against prostate tumorigenesis. TRAMP mice on the standard C57BL/6JxFVB genetic background were fed 0, 10, or 40mg 6-MCDF/kg diet beginning at 8 weeks of age. Tumor incidence, pelvic lymph node metastasis, and serum vascular endothelial growth factor (VEGF) concentrations were determined at 140 days of age. Prostate tumor incidence and size were not significantly reduced in mice fed 6-MCDF. However, the frequency of pelvic lymph node metastasis was reduced fivefold in mice fed the 40mg 6-MCDF/kg diet. Serum VEGF concentrations were also reduced by 6-MCDF treatment, particularly in mice without prostate tumors, and 6-MCDF was shown to act directly on cultured prostates to inhibit VEGF secretion. Together, these results suggest that 6-MCDF inhibits metastasis, in part, by inhibiting prostatic VEGF production prior to tumor formation. This is the first report that 6-MCDF can confer protection against prostate cancer in vivo.


Asunto(s)
Benzofuranos/uso terapéutico , Neoplasias Pélvicas/prevención & control , Neoplasias Pélvicas/secundario , Neoplasias de la Próstata/tratamiento farmacológico , Receptores de Hidrocarburo de Aril/fisiología , Animales , Benzofuranos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Neoplasias Pélvicas/patología , Neoplasias de la Próstata/patología
12.
Biochem Pharmacol ; 77(4): 566-76, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18977204

RESUMEN

Most evidence of aryl hydrocarbon receptor (AHR) signaling in prostate growth, morphogenesis, and disease stems from research using 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) to pharmacologically activate the AHR at various stages of development. This review discusses effects of TCDD on prostate morphogenesis and highlights interactions between AHR and other signaling pathways during normal and aberrant prostate growth. Although AHR signaling modulates estrogen and androgen signaling in other tissues, crosstalk between these steroid hormone receptors and AHR signaling cannot account for actions of TCDD on prostate morphogenesis. Instead, the AHR appears to act within a cooperative framework of developmental signals to regulate timing and patterning of prostate growth. Inappropriate activation of AHR signaling as a result of early life TCDD exposure disrupts the balance of these signals, impairs prostate morphogenesis, and has an imprinting effect on the developing prostate that predisposes to prostate disease in adulthood. Mechanisms of AHR signaling in prostate growth and disease are only beginning to be unraveled and recent studies have revealed its interactions with WNT5A, retinoic acid, fibroblast growth factor 10, and vascular endothelial growth factor signaling pathways.


Asunto(s)
Organogénesis/fisiología , Próstata/metabolismo , Enfermedades de la Próstata/metabolismo , Receptores de Hidrocarburo de Aril/fisiología , Animales , Contaminantes Ambientales/toxicidad , Humanos , Masculino , Organogénesis/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Próstata/embriología , Próstata/crecimiento & desarrollo , Próstata/patología , Enfermedades de la Próstata/inducido químicamente , Enfermedades de la Próstata/patología , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Esteroides/metabolismo , Transducción de Señal/efectos de los fármacos
13.
Dev Biol ; 324(1): 10-7, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18804104

RESUMEN

The establishment of prostatic budding patterns occurs early in prostate development but mechanisms responsible for this event are poorly understood. We investigated the role of WNT5A in patterning prostatic buds as they emerge from the fetal mouse urogenital sinus (UGS). Wnt5a mRNA was expressed in UGS mesenchyme during budding and was focally up-regulated as buds emerged from the anterior, dorsolateral, and ventral UGS regions. We observed abnormal UGS morphology and prostatic bud patterns in Wnt5a null male fetuses, demonstrated that prostatic bud number was decreased by recombinant mouse WNT5A protein during wild type UGS morphogenesis in vitro, and showed that ventral prostate development was selectively impaired when these WNT5A-treated UGSs were grafted under under kidney capsules of immunodeficient mice and grown for 28 d. Moreover, a WNT5A inhibitory antibody, added to UGS organ culture media, rescued prostatic budding from inhibition by a ventral prostatic bud inhibitor, 2,3,8,7-tetrachlorodibenzo-p-dioxin, and restored ventral prostate morphogenesis when these tissues were grafted under immunodeficient mouse kidney capsules and grown for 28 d. These results suggest that WNT5A participates in prostatic bud patterning by restricting mouse ventral prostate development.


Asunto(s)
Próstata/embriología , Proteínas Wnt/fisiología , Animales , Tipificación del Cuerpo , Masculino , Ratones , Ratones Noqueados , Organogénesis , Próstata/metabolismo , Técnicas de Cultivo de Tejidos , Proteína Wnt-5a
14.
Toxicol Sci ; 106(2): 488-96, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18779384

RESUMEN

Prostate ductal development is initiated by androgen-dependent signals in fetal urogenital sinus (UGS) mesenchyme that stimulate prostatic bud formation in UGS epithelium. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD, 5 microg/kg maternal dose) inhibited ventral and dorsolateral but not anterior prostatic budding. We sought to determine which stage of budding, specification or initiation, was inhibited. Ventral prostatic bud formation was maximally inhibited when TCDD exposure spanned E15.5-16.5 and dorsolateral prostatic bud formation when it spanned E14.5-15.5. Because ventral and dorsolateral buds are specified at these times, TCDD impaired bud specification. We hypothesized that TCDD inhibited ventral bud specification by forming a continuous smooth muscle barrier between UGS mesenchyme and epithelium in the ventral prostatic UGS region, blocking mesenchymal-epithelial signaling, but no such barrier was found. We hypothesized that increased aryl hydrocarbon receptor (AHR) signaling in ventral and dorsolateral UGS increased their sensitivity to TCDD, but levels of AHR nuclear translocator (ARNT) protein, Ahr mRNA, and AHR-dependent gene expression were not higher than in anterior UGS where budding was unaffected. However, we identified overlapping expression of Ahr, ARNT, and AHR-induced transcripts in the periprostatic mesenchyme which intimately contacts UGS epithelium where buds are specified. This was considered the putative TCDD site of action in the UGS for inhibition of ventral and dorsolateral prostatic bud specification. Thus, hyperactivation of AHR signaling appears to disrupt dorsoventral patterning of the UGS, reprogramming where prostatic buds are specified, and prostate lobes are formed. Disrupted axial patterning provides a new paradigm for understanding how in utero TCDD exposure causes ventral prostate agenesis and may shed light on how TCDD impairs development of other organs.


Asunto(s)
Tipificación del Cuerpo/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Próstata/efectos de los fármacos , Animales , Secuencia de Bases , Cartilla de ADN , Relación Dosis-Respuesta a Droga , Femenino , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Embarazo , Próstata/embriología
15.
J Biol Rhythms ; 23(3): 200-10, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18487412

RESUMEN

Transcription factors belonging to the Per/Arnt/Sim (PAS) domain family are highly conserved and many are involved in circadian rhythm regulation. One member of this family, aryl hydrocarbon receptor (AhR), is an orphan receptor whose physiological role is unknown. Recent findings have led to the hypothesis that AhR has a role in circadian rhythm, which is the focus of the present investigation. First, time-of-day-dependent mRNA expression of AhR and its signaling target, cytochrome p4501A1 (Cyp1a1), was determined in C57BL/6J mice by quantitative RT-PCR. Circadian expression of AhR and Cyp1a1 was observed both in the suprachiasmatic nucleus (SCN) and liver. Next, the circadian phenotype of mice lacking AhR (AhRKO) was investigated using behavioral monitoring. Intact AhRKO mice had robust circadian rhythmicity with a similar tau under constant conditions compared to wild-type mice, but a significant difference in tau was observed between genotypes in ovariectomized female mice. Time to reentrainment following 6-h advances or delays of the light/dark cycle was not significantly different between genotypes. However, mice exposed to the AhR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 1 microg/kg of body weight) displayed decreased phase shifts in response to light and had altered expression of Per1 and Bmal1. These results suggest that chronic activation of AhR may affect the ability of the circadian timekeeping system to adjust to alterations in environmental lighting by affecting canonical clock genes. Further studies are necessary to decipher the mechanism of how AhR agonists could disrupt light-induced phase shifts. If AhR does have a role in circadian rhythm, it may share redundant roles with other PAS domain proteins and/or the role of AhR may not be exhibited in the behavioral activity rhythm, but could be important elsewhere in the peripheral circadian system.


Asunto(s)
Conducta Animal/efectos de los fármacos , Luz , Dibenzodioxinas Policloradas/farmacología , Receptores de Hidrocarburo de Aril/fisiología , Animales , Secuencia de Bases , Ritmo Circadiano , Citocromo P-450 CYP1A1/metabolismo , Cartilla de ADN , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Receptores de Hidrocarburo de Aril/genética , Núcleo Supraquiasmático/enzimología , Núcleo Supraquiasmático/metabolismo
16.
Carcinogenesis ; 29(5): 1077-82, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18359762

RESUMEN

Hypoxia-inducible factor-1 alpha (HIF-1alpha) and aryl hydrocarbon receptor nuclear translocator (ARNT) are basic helix-loop-helix/per-arnt-sim (PAS) family transcription factors. During angiogenesis and tumor growth, HIF-1alpha dimerizes with ARNT, inducing expression of many genes, including vascular endothelial growth factor (VEGF). ARNT also dimerizes with the aryl hydrocarbon receptor (AhR). AhR-null (Ahr(-/-)) transgenic adenocarcinoma of the mouse prostate (TRAMP) mice develop prostate tumors with greater frequency than AhR wild-type (Ahr(+/+)) TRAMP mice, even though prevalence of prostate epithelial hyperplasia is not inhibited. This suggests that Ahr inhibits prostate carcinogenesis. In TRAMP mice, prostatic epithelial hyperplasia results in stabilized HIF-1alpha, inducing expression of VEGF, a prerequisite for tumor growth and angiogenesis. Since ARNT is a common dimerization partner of AhR and HIF-1alpha, we hypothesized that the AhR inhibits prostate tumor formation by competing with HIF-1alpha for ARNT, thereby limiting VEGF production. Prostates from Ahr(+/+), Ahr(+/-) and Ahr(-/-) C57BL/6J TRAMP mice were cultured in the presence of graded concentrations of vanadate, an inducer of VEGF through the HIF-1alpha-ARNT pathway. Vanadate induced VEGF protein in a dose-dependent fashion in Ahr(+/-) and Ahr(-/-) TRAMP cultures, but not in Ahr(+/+) cultures. However, vanadate induced upstream proteins in the phosphatidylinositol 3-kinase-signaling cascade to a similar extent in TRAMPs of each Ahr genotype, evidenced by v-akt murine thymoma viral oncogene homolog (Akt) phosphorylation. These findings suggest that AhR sequesters ARNT, decreasing interaction with HIF-1alpha reducing VEGF production. Since VEGF is required for tumor vascularization and growth, these studies further suggest that reduction in VEGF correlates with inhibited prostate carcinogenesis in Ahr(+/+) TRAMP mice.


Asunto(s)
Neoplasias de la Próstata/prevención & control , Receptores de Hidrocarburo de Aril/fisiología , Vanadatos/antagonistas & inhibidores , Vanadatos/toxicidad , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/fisiología , Dimerización , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neovascularización Patológica/genética , Neoplasias de la Próstata/fisiopatología , Receptores de Hidrocarburo de Aril/deficiencia , Receptores de Hidrocarburo de Aril/genética , Trasplante Heterólogo
17.
J Appl Toxicol ; 28(6): 724-33, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18172886

RESUMEN

To search for genes whose products modify aryl hydrocarbon receptor (AhR)-dependent toxicity caused by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), gene expression profiles in the liver were surveyed using microarrays 24 h after the administration of TCDD to three strains of female mice, BALB/cAnN (BALB), C3H/HeN (C3H) and CBA/JN (CBA) all of identical AhR genotype. The BALB/cAnN strain had a more marked induction of a number of glutathione S-transferase (GST) sub-families, particularly the GSTmicro gene family, compared with the other two strains. To assess the effects of GSTs induction to metabolize carcinogens, TCDD (40 microg kg(-1)) was administered to BALB and CBA strains, followed 24 h later by an i.p. injection of low or high dose of benzo[a]pyrene (B[a]P, 50 or 200 mg kg(-1)). The 32P-postlabelling analysis showed that administration of TCDD alone failed to induce DNA adduct formation in both BALB and CBA strain mouse livers. The low dose of B[a]P alone produced DNA adduct in the liver of both strains to a similar extent. Treatment with TCDD 24 h before the low dose of B[a]P suppressed the formation of B[a]P-induced DNA-adduct more markedly in the BALB strain compared with the CBA strain. Taken together, these findings show that TCDD treatment causes strain-specific alterations in gene expression and B[a]P-induced DNA adduct formation in the liver of female mice of the same AhRb2 genotype. Furthermore, it suggests that TCDD-treated female mice of the BALB strain may have genes whose products modify the toxicity of B[a]P as evidenced by TCDD-induced alterations in B[a]P-DNA adduct formation.


Asunto(s)
Benzo(a)pireno/toxicidad , Aductos de ADN/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Hígado/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/genética , Animales , Cartilla de ADN , Femenino , Expresión Génica/efectos de los fármacos , Genotipo , Hígado/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos CBA , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie
18.
Carcinogenesis ; 28(2): 497-505, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17052998

RESUMEN

The aryl hydrocarbon receptor (AhR) is a transcription factor that mediates the inhibitory effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on prostate growth and also modulates normal prostate development. This is evidenced by AhR null mice (Ahr-/-) having smaller dorsolateral and anterior prostates, even though all prostate lobes remain histologically normal. To test the hypothesis that loss of the AhR increases the rate of prostate carcinogenesis, the incidence of macroscopic prostate tumors was determined in Ahr+/+, Ahr+/- and Ahr-/- C57BL/6J transgenic adenocarcinoma of the mouse prostate (TRAMP) mice at 35, 70, 105, 140, 175 and 210 days of age. From 140 days, prostate tumor incidence was greater in Ahr-/- (60%) and Ahr+/- (43%) mice than in Ahr+/+ mice (16%). Allele quantification did not indicate a loss of the wild-type Ahr allele in heterozygous TRAMP tumors, suggesting that tumor formation in these mice was not due to a loss of Ahr heterozygosity. Prostatic SV40 large T antigen mRNA expression and protein localization were comparable in TRAMP mice of each Ahr genotype. Prostates from all mice of each Ahr genotype were histologically indistinguishable, exhibiting diffuse epithelial hyperplasia by 105 days of age. mRNA expression and protein localization for molecular markers of neuroendocrine differentiation, including chromogranin A and neuropilin-1, were elevated in prostate tumors compared to tumor-free ventral prostates, regardless of Ahr genotype or age. Taken together, these results demonstrate that the Ahr inhibits prostate carcinogenesis in C57BL/6J TRAMP mice by interfering with neuroendocrine differentiation.


Asunto(s)
Transformación Celular Neoplásica , Neoplasias de la Próstata/prevención & control , Receptores de Hidrocarburo de Aril/fisiología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/fisiología , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Secuencia de Bases , Peso Corporal , Diferenciación Celular , Cromogranina A/genética , Cartilla de ADN , Dosificación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropilina-1/genética , Neoplasias de la Próstata/patología , ARN Mensajero/genética , Receptores de Hidrocarburo de Aril/genética , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética
19.
J Immunotoxicol ; 3(1): 21-30, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18958682

RESUMEN

Diesel exhaust particles (DEP) were reported to have adverse effects on the immune system of laboratory animals and to induce thymic involution, particularly when exposure occurred during the fetal or lactational period. DEP consist of a carbon core to which many organic compounds are adsorbed, including polyaromatic hydrocarbons (PAHs) and their derivatives (e.g., dioxins and quinones). Although it has been suggested that these organic compounds were responsible for mediating the effects of DEP through their regulation of gene expression, the molecular mechanism of action of DEP has not been fully elucidated. In this study, we examined the direct effect of DEP extracts and their constituents on gene expression and phenotype in the fetal thymus. Fetal thymuses from C57BL/6 mice were exposed to DEP extracts for 24 hrs, after which their gene expression was analyzed using an Affymetrix GeneChip system. DEP extracts up-regulated several genes known as arylhydrocarbon receptor (AhR)-target genes, including cytochrome P450 1a1 (Cyp1a1), 1b1 (Cyp1b1), TCDD-inducible poly(ADP-ribose) polymerase (Tiparp), and scinderin (Scin). Similarly, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and benzo[a]pyrene (B[a]P), which are AhR ligands, induced remarkably similar changes in gene expression compared to DEP extracts. In addition, our data showed little contribution of quinones to DEP extracts-induced changes in gene expression in fetal thymus through oxidative stress responses. These changes in gene expression were also confirmed by semi-quantitative RT-PCR. Furthermore, DEP extracts skewed thymic T-cell differentiation in favor of the production of CD8 T-cells, which was also observed when exposed to AhR ligands. Our results suggest that organic compounds adsorbed onto DEP alter thymic gene expression and directly affect thymocyte development by activating the AhR.

20.
Toxicol Sci ; 86(2): 387-95, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15888670

RESUMEN

In utero and lactational 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure inhibits ventral, dorsolateral, and anterior prostate development in C57BL/6 mice. To determine if prostatic abnormalities persist into senescence, mice born to dams given TCDD (5 mug/kg, po) or vehicle on gestation day 13 were examined at 100 and 510 days of age. Half the mice were castrated ten days prior to necropsy in order to assess androgen dependence, while the remaining mice were sham castrated. Effects of TCDD on the dorsolateral and anterior prostate of senescent sham-castrated mice were relatively subtle, whereas the ventral prostate was rudimentary or absent. Castration of vehicle-exposed mice caused far greater reductions in prostate lobe weights, epithelial cell height, and androgen-dependent gene expression (MP25 and probasin) in young mice than in senescent ones, while cell proliferation was decreased by castration in young mice and increased in senescence. Responses to castration were similar at 100 days of age in vehicle- and TCDD-exposed mice. At 510 days, however, TCDD-exposed mice were substantially more responsive to castration by most indices than vehicle-exposed mice. These results demonstrate that prostatic androgen dependence in mice declines substantially with age in several key ways, and that in utero and lactational TCDD exposure protects against this decline. Surprisingly, TCDD increased the incidence of cribriform structures in dorsolateral prostate ducts, from 2-3% in vehicle-exposed senescent mice to 16% in sham-castrated and to 7% in castrated senescent mice. Collectively, these results demonstrate that effects of in utero and lactational TCDD exposure on the prostate persist into senescence, and suggest that in utero and lactational TCDD exposure retards the aging process in the prostate. However, because cribriform structures are often considered to be associated with prostate carcinogenesis, these results also suggest that TCDD exposure early in development may increase susceptibility to prostate cancer.


Asunto(s)
Envejecimiento/fisiología , Lactancia , Dibenzodioxinas Policloradas/toxicidad , Efectos Tardíos de la Exposición Prenatal , Próstata/efectos de los fármacos , Proteína de Unión a Andrógenos/genética , Proteína de Unión a Andrógenos/metabolismo , Animales , Castración , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Embarazo , Próstata/crecimiento & desarrollo , Próstata/patología , ARN Mensajero/metabolismo , Vesículas Seminales/efectos de los fármacos , Vesículas Seminales/crecimiento & desarrollo , Vesículas Seminales/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA