Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Inherit Metab Dis ; 46(4): 618-633, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37114839

RESUMEN

BACKGROUND: Glycogen storage disease type Ia (GSDIa) is caused by biallelic pathogenic variants in the glucose-6-phosphatase gene (G6PC) and mainly characterized by hypoglycemia, hepatomegaly, and renal insufficiency. Although its symptoms are reportedly mild in patients carrying the G6PC c.648G>T variant, the predominant variant in Japanese patients, details remain unclear. Therefore, we examined continuous glucose monitoring (CGM) data and daily nutritional intake to clarify their associations in Japanese patients with GSDIa with G6PC c.648G>T. METHODS: This cross-sectional study enrolled 32 patients across 10 hospitals. CGM was performed for 14 days, and nutritional intake was recorded using electronic diaries. Patients were divided according to genotype (homozygous/compound heterozygous) and age. The durations of biochemical hypoglycemia and corresponding nutritional intake were analyzed. Multiple regression analysis was performed to identify factors associated with the duration of biochemical hypoglycemia. RESULTS: Data were analyzed for 30 patients. The mean daily duration of hypoglycemia (<4.0 mmol/L) in the homozygous group increased with age (2-11 years [N = 8]: 79.8 min; 12-18 years [5]: 84.8 min; ≥19 years [10]: 131.5 min). No severe hypoglycemic symptoms were recorded in the patients' diaries. The mean frequency of snack intake was approximately three times greater in patients aged 2-11 years (7.1 times/day) than in those aged 12-18 years (1.9 times/day) or ≥19 years (2.2 times/day). Total cholesterol and lactate were independently associated with the duration of biochemical hypoglycemia. CONCLUSION: Although nutritional therapy prevents severe hypoglycemia in patients with GSDIa with G6PC c.648G>T, patients often experience asymptomatic hypoglycemia.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo I , Hipoglucemia , Humanos , Glucemia , Estudios Transversales , Automonitorización de la Glucosa Sanguínea , Enfermedad del Almacenamiento de Glucógeno Tipo I/complicaciones , Glucosa-6-Fosfatasa/genética , Hipoglucemia/complicaciones
2.
JAMA Netw Open ; 5(8): e2228500, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35997978

RESUMEN

Importance: The prevalence of atrial fibrillation (AF) increases with age and is more common in frail patients. However, data are lacking on outcomes of oral anticoagulants (OACs) in very elderly patients with AF with frailty, who are ineligible for standard anticoagulant treatment. Objective: To compare very-low-dose edoxaban (15 mg daily) vs placebo across frailty status, including each of 5 frailty assessment parameters, among patients with AF involved in the ELDERCARE-AF (Edoxaban Low-Dose for Elder Care Atrial Fibrillation Patients) trial. Design, Setting, and Participants: This is a cohort study using data from ELDERCARE-AF, a multicenter, randomized, double-blind, placebo-controlled phase 3 study of Japanese patients with AF aged 80 years or older who were ineligible for OACs at doses approved for stroke prevention because of their high bleeding risks. Eligible patients were randomly assigned (1:1) to receive edoxaban or placebo. The study duration was from August 5, 2016, to November 5, 2019, with the last patient followed up on December 27, 2019. Data analysis was performed from February 2021 to February 2022. Exposure: Edoxaban (15 mg) once daily or placebo. Main Outcomes and Measures: The primary efficacy end point was the composite of stroke or systemic embolism, and the primary safety end point was major bleeding. Results: A total of 984 patients were randomly assigned to treatment (492 each to the edoxaban and placebo groups); 944 patients (402 frail patients [42.6%]; 542 nonfrail patients [57.4%]; mean [SD] age, 86.6 [4.3] years; 541 women [57.3%]) were included in this analysis. In the placebo group, the estimated event rates (SE) for stroke or systemic embolism were 7.1% (1.6%) per patient-year in the frail group and 6.1% (1.3%) per patient-year in the nonfrail group. Edoxaban was associated with lower event rates for stroke or systemic embolism with no interaction with frailty status or frailty assessment parameters. Major bleeding and major or clinically relevant nonmajor bleeding events were both numerically higher in the edoxaban group than in the placebo group, and no heterogeneity was observed with frailty status. Although both all-cause death and net clinical composite outcome occurred more frequently in the frail group than in the nonfrail group, there was no association with frailty status between the edoxaban and placebo groups. Conclusions and Relevance: Regardless of frailty status, among Japanese patients with AF aged 80 years or older who were ineligible for standard OACs, once-daily 15-mg edoxaban was associated with reduced incidence of stroke or systemic embolism and may be a suitable treatment option for these patients.


Asunto(s)
Fibrilación Atrial , Embolia , Fragilidad , Accidente Cerebrovascular , Anciano , Anciano de 80 o más Años , Anticoagulantes/uso terapéutico , Fibrilación Atrial/complicaciones , Fibrilación Atrial/tratamiento farmacológico , Estudios de Cohortes , Embolia/epidemiología , Inhibidores del Factor Xa/efectos adversos , Inhibidores del Factor Xa/uso terapéutico , Femenino , Anciano Frágil , Fragilidad/complicaciones , Fragilidad/epidemiología , Hemorragia/inducido químicamente , Hemorragia/epidemiología , Humanos , Piridinas , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/prevención & control , Tiazoles
3.
JAMA Cardiol ; 7(6): 583-590, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35416910

RESUMEN

Importance: Long-term use of oral anticoagulants (OACs) is necessary for stroke prevention in patients with atrial fibrillation (AF). The effectiveness and safety of OACs in extremely older patients (ie, aged 80 years or older) with AF and at high risk of bleeding needs to be elucidated. Objective: To examine the effects of very low-dose edoxaban (15 mg) vs placebo across 3 age strata (80-84 years, 85-89 years, and ≥90 years) among patients with AF who were a part of the Edoxaban Low-Dose for Elder Care Atrial Fibrillation Patients (ELDERCARE-AF) trial. Design, Setting, and Participants: This prespecified subanalysis of a phase 3, randomized, double-blind, placebo-controlled trial was conducted from August 5, 2016, to December 27, 2019. Patients with AF aged 80 years or older who were not considered candidates for standard-dose OACs were included in the study; reasons these patients could not take standard-dose OACs included low creatinine clearance (<30 mL per minute), low body weight (≤45 kg), history of bleeding from critical organs, continuous use of nonsteroidal anti-inflammatory drugs, or concomitant use of antiplatelet drugs. Eligible patients were recruited randomly from 164 hospitals in Japan and were randomly assigned 1:1 to edoxaban or placebo. Interventions: Edoxaban (15 mg once daily) or placebo. Main Outcomes and Measures: The primary efficacy end point was the composite of stroke or systemic embolism. The primary safety end point was International Society on Thrombosis and Hemostasis-defined major bleeding. Results: A total of 984 patients (mean [SD] age: age group 80-84 years, 82.2 [1.4] years; age group 85-89 years, 86.8 [1.4] years; age group ≥90 years, 92.3 [2.1] years; 565 women [57.4%]) were included in this study. In the placebo group, estimated (SE) event rates for stroke or systemic embolism increased with age and were 3.9% (1.2%) per patient-year in the group aged 80 to 84 years (n = 181), 7.3% (1.7%) per patient-year in the group aged 85 to 89 years (n = 184), and 10.1% (2.5%) per patient-year in the group aged 90 years or older (n = 127). A 15-mg dose of edoxaban consistently decreased the event rates for stroke or systemic embolism with no interaction with age (80-84 years, hazard ratio [HR], 0.41; 95% CI, 0.13-1.31; P = .13; 85-89 years, HR, 0.42; 95% CI, 0.17-0.99; P = .05; ≥90 years, HR, 0.23; 95% CI, 0.08-0.68; P = .008; interaction P = .65). Major bleeding and major or clinically relevant nonmajor bleeding events were numerically higher with edoxaban, but the differences did not reach statistical significance, and there was no interaction with age. There was no difference in the event rate for all-cause death between the edoxaban and placebo groups in all age strata. Conclusions and Relevance: Results of this subanalysis of the ELDERCARE-AF randomized clinical trial revealed that among Japanese patients aged 80 years or older with AF who were not considered candidates for standard OACs, a once-daily 15-mg dose of edoxaban was superior to placebo in preventing stroke or systemic embolism consistently across all 3 age strata, including those aged 90 years or older, albeit with a higher but nonstatistically significant incidence of bleeding. Trial Registration: ClinicalTrials.gov Identifier: NCT02801669.


Asunto(s)
Fibrilación Atrial , Embolia , Accidente Cerebrovascular , Anciano , Anciano de 80 o más Años , Anticoagulantes , Fibrilación Atrial/complicaciones , Fibrilación Atrial/tratamiento farmacológico , Embolia/epidemiología , Embolia/etiología , Embolia/prevención & control , Inhibidores del Factor Xa , Femenino , Hemorragia/inducido químicamente , Hemorragia/complicaciones , Hemorragia/epidemiología , Humanos , Piridinas , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/prevención & control , Tiazoles , Warfarina/uso terapéutico
4.
Commun Biol ; 4(1): 994, 2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34417555

RESUMEN

Reduced glucose uptake into the skeletal muscle is an important pathophysiological abnormality in type 2 diabetes, and is caused by impaired translocation of glucose transporter 4 (GLUT4) to the skeletal muscle cell surface. Here, we show a xanthene derivative, DS20060511, induces GLUT4 translocation to the skeletal muscle cell surface, thereby stimulating glucose uptake into the tissue. DS20060511 induced GLUT4 translocation and stimulated glucose uptake into differentiated L6-myotubes and into the skeletal muscles in mice. These effects were completely abolished in GLUT4 knockout mice. Induction of GLUT4 translocation by DS20060511 was independent of the insulin signaling pathways including IRS1-Akt-AS160 phosphorylation and IRS1-Rac1-actin polymerization, eNOS pathway, and AMPK pathway. Acute and chronic DS20060511 treatment attenuated the glucose intolerance in obese diabetic mice. Taken together, DS20060511 acts as a skeletal muscle-specific GLUT4 translocation enhancer to facilitate glucose uptake. Further studies of DS20060511 may pave the way for the development of novel antidiabetic medicines.


Asunto(s)
Intolerancia a la Glucosa/genética , Transportador de Glucosa de Tipo 4/genética , Músculo Esquelético/metabolismo , Translocación Genética , Xantenos/metabolismo , Animales , Intolerancia a la Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Ratones
5.
Biochem Pharmacol ; 186: 114495, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33711284

RESUMEN

Although dioxins and related chemicals have been suspected to disrupt child development, their toxic mechanism remains poorly understood. Our previous studies in rat fetuses revealed that maternal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a highly-toxic dioxin, suppresses fetal synthesis of pituitary growth hormone (GH) that is essential for development. This study examined the hypothesis that attenuating GH expression in fetuses triggers developmental disorders. Treating pregnant rats with 1 µg/kg TCDD reduced the circulating level of GH and its downstream factor, insulin-like growth factor-1 (IGF-1), in the offspring only during the fetal and early neonatal stages. Although maternal TCDD exposure resulted in low body weight and length at babyhood and defects in the learning and memory ability at adulthood, GH supplementation in TCDD-exposed fetuses restored or tended to restore the defects including IGF-1 downregulation. Moreover, maternal TCDD exposure decreased the number of GH-positive cells during the fetal/neonatal stage. A microarray analysis showed that TCDD reduced the expression of death-associated protein-like 1 (DAPL1), a cell cycle-dependent proliferation regulator, in the fetal pituitary gland. In addition, TCDD treatment attenuated proliferating cells and cyclin mRNA expression in the fetal pituitary gland. Aryl hydrocarbon receptor (AHR)-knockout fetuses were insensitive to TCDD treatment, indicating that the TCDD-induced reduction in DAPL1 and GH mRNAs expression was due to AHR activation. Finally, DAPL1 knockdown suppressed GH and cyclin D2 expression in fetal pituitary cells. These results provide a novel evidence that dioxin suppresses GH-producing cell proliferation and GH synthesis due to partly targeting DAPL1, thereby impairing offspring development.


Asunto(s)
Discapacidades del Desarrollo/metabolismo , Dioxinas/toxicidad , Feto/metabolismo , Hormona del Crecimiento/deficiencia , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Discapacidades del Desarrollo/inducido químicamente , Femenino , Feto/efectos de los fármacos , Hormona del Crecimiento/antagonistas & inhibidores , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Ratas , Ratas Transgénicas , Ratas Wistar
6.
EMBO Rep ; 21(9): e49828, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32672883

RESUMEN

While brown adipose tissue (BAT) is well-recognized for its ability to dissipate energy in the form of heat, recent studies suggest multifaced roles of BAT in the regulation of glucose and lipid homeostasis beyond stimulating thermogenesis. One of the functions involves interorgan communication with metabolic organs, such as the liver, through BAT-derived secretory factors, a.k.a., batokine. However, the identity and the roles of such mediators remain insufficiently understood. Here, we employed proteomics and transcriptomics in human thermogenic adipocytes and identified previously unappreciated batokines, including phospholipid transfer protein (PLTP). We found that increased circulating levels of PLTP, via systemic or BAT-specific overexpression, significantly improve glucose tolerance and insulin sensitivity, increased energy expenditure, and decrease the circulating levels of cholesterol, phospholipids, and sphingolipids. Such changes were accompanied by increased bile acids in the circulation, which in turn enhances glucose uptake and thermogenesis in BAT. Our data suggest that PLTP is a batokine that contributes to the regulation of systemic glucose and lipid homeostasis as a mediator of BAT-liver interorgan communication.


Asunto(s)
Tejido Adiposo Pardo , Glucosa , Tejido Adiposo Pardo/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Homeostasis , Humanos , Lípidos , Hígado , Termogénesis
7.
J Toxicol Sci ; 40(6): 909-16, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26558472

RESUMEN

The effects of endocrine disruptors on testicular steroidogenesis in fetal rats were investigated in a study involving in utero exposure. In the major part of this study, pregnant rats at gestational day (GD)15 were given a single oral administration of the test substance, and then the expression of the following mRNAs in GD20 fetuses was determined: testicular steroidogenic acute-regulatory protein (StAR), a cholesterol transporter mediating the rate-limiting step of steroidogenesis, a ß-subunit of pituitary luteinizing hormone (LH), and a regulator of gonadal steroidogenesis. Among the substances tested, only di(2-ethylhexyl)phthalate (DEHP) reduced the expression of fetal testicular StAR. The others listed below exhibited little effect on fetal StAR: 2,2',4,4'-tetrabromodiphenylether, tributyltin chloride, atrazine, permethrin, cadmium chloride (Cd), lead acetate (Pb) and methylmercury (CH3HgOH). None of them, including DEHP, lacked the ability to reduce the expression of pituitary LHß mRNA. The present study also examined the potential of metals as modifiers of fetal steroidogenesis by giving them to pregnant dams in drinking water during GD1 and GD20. Under these conditions, Cd and Pb at a low concentration (0.01 ppm) significantly attenuated the fetal testicular expression of StAR mRNA without a concomitant reduction in LHß. No such effect was detected with CH3HgOH even at 1 ppm. These results suggest that: 1) DEHP, Cd and Pb attenuate the fetal production of sex steroids by directly acting on the testis, and 2) chronic treatment during the entire gestational period is more useful than a single administration for determining the hazardous effect of a suspected endocrine disruptor on fetal steroidogenesis.


Asunto(s)
Dietilhexil Ftalato/toxicidad , Disruptores Endocrinos/toxicidad , Feto/metabolismo , Gónadas/fisiología , Hormona Luteinizante/metabolismo , Exposición Materna/efectos adversos , Fosfoproteínas/metabolismo , Hipófisis/fisiología , Testículo/embriología , Testículo/metabolismo , Animales , Compuestos de Cadmio/toxicidad , Femenino , Plomo/toxicidad , Hormona Luteinizante/genética , Masculino , Intercambio Materno-Fetal , Embarazo , ARN Mensajero/metabolismo , Ratas Wistar
8.
Fukuoka Igaku Zasshi ; 106(5): 127-34, 2015 May.
Artículo en Japonés | MEDLINE | ID: mdl-26226673

RESUMEN

Maternal exposure to dioxins causes a number of developmental disorders in the offspring. Previous studies have suggested that lactational exposure to 2,3,7,8-tetrachlorodizenzo-p-dioxin (TCDD) reduces the pup level of thyroid hormone after weaning, leading to the damage to their development including neural maturation. However, the specificity for age and dioxin congeners as well as dose dependency in terms of a reduction in pup thyroid hormone remains to be clarified. To address this issue, we investigated whether TCDD or 2,3,4,7,8-pentachlorodibenzofuran (PenCDF), one of the dioxins which caused 'Yusho' incident, affects the status of thyroid hormone during the fetal and neonatal periods. Treating pregnant rats at gestational day (GD)15 with 1 µg/kg TCDD scarcely affected the serum concentration of thyroxine, although a significant reduction by TCDD was detected at limited endpoints [GD21 and postnatal day (PND)21]. In addition, maternal exposure to TCDD (0.05-30 µg/kg) or PenCDF (1-1,000 µg/kg) did not have any change in the serum level of thyroxine in GD20 fetuses even at the maximum dose. Neither the expression of pituitary thyroid-stimulating hormone ß (TSHß) nor hypothalamic thyrotropin-releasing hormone was sensitive to TCDD treatment. In pregnant dams, TCDD decreased the serum level of thyroxine at GD20 and 21, while the pituitary expression of TSHß was induced. These results suggest that a single administration of dioxins to pregnant rats at GD15 have little effect on the level of thyroxine in the fetuses and infants, while a reduced level of this hormone observed in the offspring at GD21 and PND21 and pregnant dams at GD20 and 21.


Asunto(s)
Dioxinas/toxicidad , Feto/efectos de los fármacos , Animales , Femenino , Masculino , Dibenzodioxinas Policloradas/toxicidad , Embarazo , Ratas , Ratas Wistar , Tiroxina/sangre
9.
Toxicol Appl Pharmacol ; 281(1): 48-57, 2014 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-25220434

RESUMEN

The effect of 2,3,4,7,8-pentachlorodibenzofuran (PnCDF) on the fetal pituitary-gonad axis was compared with that produced by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in Wistar rats. Maternal treatment at gestational day (GD) 15 with PnCDF and TCDD reduced the fetal expression at GD20 of pituitary luteinizing hormone (LH) and the testicular proteins necessary for steroidogenesis. The relative potencies of PnCDF ranged from 1/42nd to 1/63rd of the TCDD effect. While PnCDF, at a dose sufficient to cause a reduction in fetal LH, provoked defects in sexual behavior at adulthood, a dose less than the ED50 failed to produce any abnormality. There was a loss of fetal body weight following in utero exposure to PnCDF, and the effect of PnCDF was also much less than that of TCDD. The disturbance in fetal growth was suggested to be due to a reduction in the level of fetal growth hormone (GH) by dioxins. The disorder caused by PnCDF/TCDD in the fetal pituitary-gonad axis occurred at doses less than those needed to cause wasting syndrome in pubertal rats. The harmful effect of PnCDF relative to TCDD was more pronounced in fetal rats than in pubertal rats. These lines of evidence suggest that: 1) PnCDF as well as TCDD imprints defects in sexual behavior by disrupting the fetal pituitary-gonad axis; 2) these dioxins hinder fetal growth by reducing the expression of fetal GH; and 3) the fetal effects of PnCDF/TCDD are more sensitive than sub-acute toxicity during puberty, and the relative effect of PnCDF varies markedly depending on the indices used.


Asunto(s)
Benzofuranos/toxicidad , Feto/efectos de los fármacos , Hipófisis/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Conducta Sexual Animal/efectos de los fármacos , Testículo/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/toxicidad , Femenino , Desarrollo Fetal/efectos de los fármacos , Desarrollo Fetal/fisiología , Feto/metabolismo , Hormona Luteinizante/sangre , Masculino , Exposición Materna , Hipófisis/metabolismo , Embarazo , Ratas , Ratas Wistar , Conducta Sexual Animal/fisiología , Testículo/fisiología
10.
Toxicol Appl Pharmacol ; 278(3): 220-9, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24793433

RESUMEN

We have previously revealed that treating pregnant rats with 2,3,7,8-tetracholorodibenzo-p-dioxin (TCDD) reduces the expression of gonadotropins and growth hormone (GH) in the fetal and neonatal pituitary. A change in gonadotropin expression impairs the testicular expression of steroidogenic proteins in perinatal pups, and imprint defects in sexual behavior after reaching maturity. In this study, we examined whether TCDD also affects the expression of gonadotropin and GH in mice using C57BL/6J and DBA/2J strains which express the aryl hydrocarbon receptor (Ahr) exhibiting a different affinity for TCDD. When pregnant C57BL/6J mice at gestational day (GD) 12 were given oral TCDD (0.2-20 µg/kg), all doses significantly attenuated the pituitary expression of gonadotropin mRNAs in fetuses at GD18. On the other hand, in DBA/2J mice, a much higher dose of TCDD (20 µg/kg) was needed to produce a significant attenuation. Such reduction in the C57BL/6J strain continued until at least postnatal day (PND) 4. In agreement with this, TCDD reduced the testicular expression of steroidogenic proteins in C57BL/6J neonates at PND2 and 4, although the same did not occur in the fetal testis and ovary. Furthermore, TCDD reduced the perinatal expression of GH, litter size and the body weight of newborn pups only in the C57BL/6J strain. These results suggest that 1) also in mice, maternal exposure to TCDD attenuates gonadotropin-regulated steroidogenesis and GH expression leading to the impairment of pup development and sexual immaturity; and 2) Ahr activation during the late fetal and early postnatal stages is required for these defects.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Contaminantes Ambientales/toxicidad , Exposición Materna/efectos adversos , Hipófisis/efectos de los fármacos , Hormonas Adenohipofisarias/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/agonistas , Teratógenos/toxicidad , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Contaminantes Ambientales/administración & dosificación , Femenino , Desarrollo Fetal/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Hipófisis/embriología , Hipófisis/metabolismo , Hormonas Adenohipofisarias/genética , Dibenzodioxinas Policloradas/administración & dosificación , Embarazo , Receptores de Hidrocarburo de Aril/metabolismo , Desarrollo Sexual/efectos de los fármacos , Organismos Libres de Patógenos Específicos , Testículo/efectos de los fármacos , Testículo/metabolismo
11.
Endocrine ; 47(2): 572-80, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24723259

RESUMEN

Exposure of pregnant rats to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) at a low dose causes developmental disorders such as growth retardation and sexual immaturity in their pups. Our previous studies have demonstrated that TCDD attenuates the expression of pituitary luteinizing hormone in fetuses, resulting in the impairment of sexual behavior after they reach maturity. In this study, we focused on growth disturbance and investigated whether TCDD affects the expression of growth hormone (GH), another pituitary hormone which is essential for normal development in perinatal pups. The result showed that maternal exposure to TCDD (1 µg/kg) at gestational day (GD) 15 reduced the fetal expression of GH from the onset at GD18. In accordance with this, TCDD attenuated the pup weight during the perinatal period. We then examined the effect of TCDD on the serum concentration of corticosterone, which plays a key role in the proliferation of GH-producing cells, and found that TCDD reduces the circulating level of corticosterone in the mothers at GD18 and the male fetuses at GD19. The reduction in fetuses seems to be due to increased inactivation rather than reduced synthesis, because TCDD induces the fetal expression of hepatic enzymes participating in the metabolism of glucocorticoids without changing the expression of steroidogenic proteins in the pituitary-adrenal axis. Supplying corticosterone to TCDD-exposed mothers restored or tended to restore a TCDD-induced reduction in pup weight as well as the levels of pituitary GH mRNA and serum GH. These results suggest that TCDD lowers GH expression and growth retardation owing, at least partially, to a reduction in the circulating level of glucocorticoid in pregnant mothers and their fetuses.


Asunto(s)
Hormona Adrenocorticotrópica/sangre , Corticosterona/sangre , Retardo del Crecimiento Fetal/inducido químicamente , Feto/efectos de los fármacos , Exposición Materna , Dibenzodioxinas Policloradas , Animales , Animales Recién Nacidos , Femenino , Retardo del Crecimiento Fetal/metabolismo , Hormona del Crecimiento/sangre , Hormona del Crecimiento/metabolismo , Masculino , Hipófisis/metabolismo , Embarazo , Ratas , Ratas Wistar
12.
Fukuoka Igaku Zasshi ; 104(4): 143-51, 2013 Apr.
Artículo en Japonés | MEDLINE | ID: mdl-23858793

RESUMEN

Maternal exposure to 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD) causes a number of toxic effects on development such as growth retardation and sexual immaturity in the offspring. However, the toxic mechanism remains unknown. Our previous studies have revealed that single oral administration of TCDD (1 jg/kg) to pregnant rats at gestational day (GD) 15 attenuates the fetal expression of testicular steroidogenic proteins such as steroidogenic acute-regulatory protein (StAR) and cytochrome P450 (CYP) 17 by targeting the fetal production of pituitary gonadotropins. In addition, we provided evidence that TCDD-produced damage on the fetal pituitary-gonad axis leads to imprint defects in sexual behaviors at adulthood. In this study, we investigated whether TCDD also affects fetal steroidogenesis in the adrenal gland. When pregnant Wistar rats were orally treated with TCDD, the fetal expression of CYP21, CYP11B1 and CYP11B2 mRNAs was either induced or tended to be induced in the male adrenal gland during GD17 and GD19, while the expression of mRNAs coding for StAR, CYP11A1 and 313-hydroxysteroid dehydrogenase was insensitive to TCDD treatment. The above alterations did not seem to be caused through a change in the upstream regulator, because TCDD exhibited little ability to attenuate the expression of adrenocorticotropin, a pituitary hormone stimulating adrenal steroidogenesis, in the male and female fetuses. In contrast to the males, TCDD effect on the adrenal gland was not observed in the female fetuses. These results suggest that maternal exposure to TCDD disrupts fetal steroidogenesis in adrenal as well as gonadal glands in a male specific manner, and the mechanism underlying the effect on adrenal gland is independent of the alteration of pituitary regulator.


Asunto(s)
Glándulas Suprarrenales/efectos de los fármacos , Glándulas Suprarrenales/metabolismo , Feto/efectos de los fármacos , Hormonas Esteroides Gonadales/biosíntesis , Exposición Materna , Dibenzodioxinas Policloradas/toxicidad , Animales , Femenino , Feto/metabolismo , Masculino , Embarazo , Ratas , Ratas Wistar , Factores Sexuales
13.
J Biol Chem ; 287(22): 18440-50, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22493514

RESUMEN

Maternal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) causes the impairment of reproduction and development in the pups. Our previous studies have revealed that maternal treatment with TCDD attenuates the fetal production of pituitary gonadotropins (luteinizing hormone (LH) and follicle-stimulating hormone) at gestational day (GD) 20, leading to the impairment of sexual behavior in adulthood. However, the mechanism underlying such a reduction has remained unknown until now. When pregnant rats at GD15 were given an oral dose of TCDD (1 µg/kg), the testicular expression of steroidogenic proteins was reduced between GD20 and postnatal days (PND) 2. In accordance with this, the pituitary expression of gonadotropin ß-subunit and serum gonadotropin were also attenuated from GD20 to PND0 in a pup-specific fashion. To identify the target genes linked to a fetal reduction in gonadotropin ß-subunit, we performed a DNA microarray analysis using the fetal pituitary and its regulatory organ, the hypothalamus. The results obtained showed that TCDD induced histone deacetylases (HDACs) in the fetal pituitary. In support with this, TCDD markedly deacetylated histones H3 and H4 twined around the promoter of the fetal LHß gene. This effect was fetus- and LHß-specific, and this was not observed in the maternal pituitary or for other pituitary hormone genes. Finally, an LHß reduction caused by TCDD was completely restored by maternal co-treatment with valproic acid, an HDAC inhibitor. These results strongly suggest that the increased deacetylation of histone owing to HDAC induction plays a critical role in the TCDD-induced reduction in LHß in the fetal pituitary.


Asunto(s)
Gonadotropinas/metabolismo , Histona Desacetilasas/biosíntesis , Dibenzodioxinas Policloradas/farmacología , Maduración Sexual , Esteroides , Animales , Secuencia de Bases , Inmunoprecipitación de Cromatina , Metilación de ADN , Inducción Enzimática , Epigénesis Genética , Femenino , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Hipófisis/efectos de los fármacos , Hipófisis/embriología , Embarazo , Ratas , Ratas Wistar , Esteroides/biosíntesis
14.
Fukuoka Igaku Zasshi ; 102(4): 159-66, 2011 Apr.
Artículo en Japonés | MEDLINE | ID: mdl-21706896

RESUMEN

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) exposed to pregnant or lactational mother impairs the reproduction and development of the pups. The defect is a serious problem, because it is caused by TCDD at much lower doses than that needed for acute toxicity in the mother. However, the toxic mechanism underlying the defect remains to be obscure. We have previously revealed that maternal exposure to TCDD (1 microg/kg) causes a reduction in luteinizing hormone in the fetal pituitary, leading to the reduced expression of testicular steroidogenic proteins such as steroidogenic acute-regulatory protein (StAR) and cytochrome P450 (CYP) 17. In addition, we have provided evidence that such a reduction imprints defects in sexual behaviors at adulthood. In this study, we investigated TCDD effect on fetal steroidogenesis in the extra-gonadal tissues. Even when pregnant Wistar rats at gestational day (GD) 15 were orally treated with TCDD (0.25, 1 or 3 microg/kg), neither expression of StAR nor CYP17 mRNA was affected in the adrenal gland, placenta and hypothalamus of male fetuses (GD20). However, TCDD induced placental StAR (3 microg/kg) and adrenal CYP17 mRNAs (0.25 microg/kg) in female fetuses. Therefore, our study suggests that while TCDD gives damage to male fetal steroidogenesis in a testis-specific manner, the dioxin enhances the steroidogenesis of the fetal adrenal gland and placenta in females. Thus, the mechanism whereby TCDD exerts its endocrine-disrupting properties is considered to differ, at least partially, between male and female fetuses.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Feto/metabolismo , Exposición Materna , Placenta/metabolismo , Dibenzodioxinas Policloradas/envenenamiento , Esteroides/biosíntesis , Animales , Femenino , Masculino , Fosfoproteínas/metabolismo , Embarazo , Ratas , Ratas Wistar , Factores Sexuales , Esteroide 17-alfa-Hidroxilasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...