Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Physiol Gastrointest Liver Physiol ; 319(2): G197-G211, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32597707

RESUMEN

Intrahepatic cholestasis of pregnancy (ICP) is characterized by elevated maternal circulating bile acid levels and associated dyslipidemia. ICP leads to accumulation of bile acids in the fetal compartment, and the elevated bile acid concentrations are associated with an increased risk of adverse fetal outcomes. The farnesoid X receptor agonist obeticholic acid (OCA) is efficient in the treatment of cholestatic conditions such as primary biliary cholangitis. We hypothesized that OCA administration during hypercholanemic pregnancy will improve maternal and fetal bile acid and lipid profiles. Female C57BL/6J mice were fed either a normal chow diet, a 0.5% cholic acid (CA)-supplemented diet, a 0.03% OCA-supplemented diet, or a 0.5% CA + 0.03% OCA-supplemented diet for 1 wk before mating and throughout pregnancy until euthanization on day 18. The effects of CA and OCA feeding on maternal and fetal morphometry, bile acid and lipid levels, and cecal microbiota were investigated. OCA administration during gestation did not alter the maternal or fetal body weight or organ morphometry. OCA treatment during hypercholanemic pregnancy reduced bile acid levels in the fetal compartment. However, fetal dyslipidemia was not reversed, and OCA did not impact maternal bile acid levels or dyslipidemia. In conclusion, OCA administration during gestation had no apparent detrimental impact on maternal or fetal morphometry and improved fetal hypercholanemia. Because high serum bile acid concentrations in ICP are associated with increased rates of adverse fetal outcomes, further investigations into the potential use of OCA during cholestatic gestation are warranted.NEW & NOTEWORTHY We used a mouse model of gestational hypercholanemia to investigate the use of obeticholic acid (OCA), a potent FXR agonist, as a treatment for the hypercholanemia of intrahepatic cholestasis of pregnancy (ICP). The results demonstrate that OCA can improve the fetal bile acid profile. This is relevant not only to women with ICP but also for women who become pregnant while receiving OCA treatment for other conditions such as primary biliary cholangitis and nonalcoholic steatohepatitis.


Asunto(s)
Ácidos y Sales Biliares/sangre , Ácido Quenodesoxicólico/análogos & derivados , Colestasis Intrahepática/tratamiento farmacológico , Complicaciones del Embarazo/tratamiento farmacológico , Animales , Ácidos y Sales Biliares/metabolismo , Ciego , Ácido Quenodesoxicólico/farmacología , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/metabolismo , Dislipidemias/tratamiento farmacológico , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Embarazo , ARN Ribosómico 16S , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
2.
Int J Obes (Lond) ; 44(12): 2382-2393, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33033395

RESUMEN

OBJECTIVES: We hypothesised that maternal diet-induced-obesity has adverse consequences for offspring energy expenditure and susceptibility to obesity in adulthood, and that the prebiotic polydextrose (PDX) would prevent the consequences of programming by maternal obesity. METHODS: Female mice were fed a control (Con) or obesogenic diet (Ob) for 6 weeks prior to mating and throughout pregnancy and lactation. Half the obese dams were supplemented with 5% PDX (ObPDX) in drinking water throughout pregnancy and lactation. Offspring were weaned onto standard chow. At 3 and 6 months, offspring energy intake (EI) and energy expenditure (EE by indirect calorimetry) were measured, and a glucose-tolerance test performed. Offspring of control (OffCon), obese (OffOb) and PDX supplemented (OffObP) dams were subsequently challenged for 3 weeks with Ob, and energy balanced reassessed. Potential modifiers of offspring energy balance including gut microbiota and biomarkers of mitochondrial activity were also evaluated. RESULTS: Six-month-old male OffOb demonstrated increased bodyweight (BW, P < 0.001) and white adipose tissue mass (P < 0.05), decreased brown adipose tissue mass (BAT, P < 0.01), lower night-time EE (P < 0.001) versus OffCon, which were prevented in OffObP. Both male and female OffOb showed abnormal glucose-tolerance test (peak [Glucose] P < 0.001; AUC, P < 0.05) which was prevented by PDX. The Ob challenge resulted in greater BW gain in both male and female OffOb versus OffCon (P < 0.05), also associated with increased EI (P < 0.05) and reduced EE in females (P < 0.01). OffObP were protected from accelerated BW gain on the OB diet compared with controls, associated with increased night-time EE in both male (P < 0.05) and female OffObP (P < 0.001). PDX also prevented an increase in skeletal muscle mtDNA copy number in OffOb versus OffCon (P < 0.01) and increased the percentage of Bacteroides cells in faecal samples from male OffObP relative to controls. CONCLUSIONS: Maternal obesity adversely influences adult offspring energy balance and propensity for obesity, which is ameliorated by maternal PDX treatment with associated changes in gut microbiota composition and skeletal muscle mitochondrial function.


Asunto(s)
Glucanos/administración & dosificación , Obesidad Materna/complicaciones , Prebióticos/administración & dosificación , Efectos Tardíos de la Exposición Prenatal , Animales , Composición Corporal , Peso Corporal , Dieta , Ingestión de Energía , Metabolismo Energético , Femenino , Microbioma Gastrointestinal , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Embarazo
3.
Pancreatology ; 20(3): 454-461, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32014435

RESUMEN

BACKGROUND: The aim was to compare population-based survival for exocrine pancreatic cancer in England in the 23 regions covered by specialist centres. The centres were initiated in 2001, covering populations of 2-4 million. METHODS: We examined incidence for adults diagnosed with a pancreatic exocrine cancer during 1995-2014 and age-standardised net survival up to five years after diagnosis for patients diagnosed during 2000-2013. We examined variation in regional resection rates and survival for patients diagnosed during 2010-2013. The data were extracted from the National Cancer Registration and Analysis Service. RESULTS: Age-standardised annual incidence rates of exocrine pancreatic cancer increased from 17.1 per 100,000 during 1995-1999 to 18.7 during 2010-2014. Age-standardised one-year and five-year net survival increased from 17.9% and 3.6%, respectively, for 2000-2009, to 21.6% and 4.2% during 2010-2013. There were 2086 (8.9%) resections among 23,415 patients diagnosed with an exocrine tumour in 2010-2013. The proportion ranged from 5.1% to 19.6% between centres. Among resected patients, survival was 73.0% at one year and 20.2% at five years. Of the total 2118 resected patients, 18 (0.9%) were at stage 1; 34 (1.6%) at stage 2; 791 (37.3%) at stage 3 and 140 (6.6%) at stage 4, although 53.6% of stage information was missing. Five-year survival was 2.1% for those who were not resected. The number of resections performed in each centre was not correlated with one-year survival. CONCLUSIONS: Despite improvements in the management of pancreatic cancer in England with the introduction of specialist centres, resection rates remain relatively low, and survival remains lower than in comparably wealthy countries.


Asunto(s)
Neoplasias Pancreáticas/epidemiología , Neoplasias Pancreáticas/cirugía , Análisis de Supervivencia , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Terapia Combinada , Inglaterra/epidemiología , Femenino , Hospitales , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Pancreáticas/tratamiento farmacológico , Población , Sistema de Registros , Tasa de Supervivencia , Adulto Joven
4.
Int J Obes (Lond) ; 43(2): 319-330, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29795465

RESUMEN

BACKGROUND: Obesity is a heterogeneous phenotype and risk associations to non-communicable diseases such as cardiovascular disease and type 2 diabetes are influenced by several factors. The paternal metabolic status at the time of conception influences offspring susceptibility to developing obesity and adiposity-associated cardiometabolic disease. Cholestatic liver diseases are characterized by raised circulating serum bile acid levels and dyslipidemia, and are commonly treated with ursodeoxycholic acid (UDCA). We hypothesized that paternal cholestasis alters offspring susceptibility to developing obesity and adiposity-associated cardiometabolic disease and that this may be modified by paternal UDCA treatment. METHODS: Cholestasis was induced in male C57BL/6 mice with a 0.5% cholic acid (CA)-supplemented diet for 10 weeks prior to mating with normal chow (NC)-fed females. Offspring of cholestatic and NC-fed fathers were fed either a NC diet or challenged with an obesogenic 'western diet' (WD) from 12 weeks of age. Offspring body weight and cardiometabolic function were assessed, and the impact of treatment of paternal cholestasis with UDCA was evaluated. RESULTS: Male offspring (18 weeks old) of cholestatic fathers challenged with WD had raised fasting insulin, hepatic triglyceride content and serum cholesterol levels compared to diet-matched controls. At 25-29 weeks of age, WD-fed male offspring of cholestatic fathers had higher systolic and diastolic blood pressure than controls and this was prevented by paternal UDCA treatment. In contrast, WD-challenged female offspring of cholestatic fathers showed improved glucose tolerance compared to controls. CONCLUSIONS: We demonstrated in our model of paternal cholestasis that offspring susceptibility to adiposity-associated cardiometabolic disease is affected in a sex-specific manner and paternal UDCA treatment had a protective effect against hypertension in the obese male offspring. The most prevalent human cholestatic conditions are primary sclerosing cholangitis and primary biliary cholangitis. These findings are of clinical relevance to children of men with these conditions.


Asunto(s)
Colestasis , Hipertensión , Obesidad , Ácido Ursodesoxicólico/uso terapéutico , Animales , Peso Corporal , Colestasis/complicaciones , Colestasis/tratamiento farmacológico , Colestasis/metabolismo , Dieta Occidental/efectos adversos , Padre , Femenino , Hipertensión/complicaciones , Hipertensión/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/complicaciones , Obesidad/metabolismo
5.
FASEB J ; 31(4): 1698-1708, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28082353

RESUMEN

Maternal metabolic adaptations are essential for successful pregnancy outcomes. We investigated how metabolic gestational processes are coordinated, whether there is a functional link with internal clocks, and whether disruptions are related to metabolic abnormalities in pregnancy, by studying day/night metabolic pathways in murine models and samples from pregnant women with normally grown and large-for-gestational age infants. In early mouse pregnancy, expression of hepatic lipogenic genes was up-regulated and uncoupled from the hepatic clock. In late mouse pregnancy, rhythmicity of energy metabolism-related genes in the muscle followed the patterns of internal clock genes in this tissue, and coincided with enhanced lipid transporter expression in the fetoplacental unit. Diurnal triglyceride patterns were disrupted in human placentas from pregnancies with large-for-gestational age infants and this overlapped with an increase in BMAL1 expression. Metabolic adaptations in early pregnancy are uncoupled from the circadian clock, whereas in late pregnancy, energy availability is mediated by coordinated muscle-placenta metabolic adjustments linked to internal clocks. Placental triglyceride oscillations in the third trimester of human pregnancy are lost in large-for-gestational age infants and may be regulated by BMAL1. In summary, disruptions in metabolic and circadian rhythmicity are associated with increased fetal size, with implications for the pathogenesis of macrosomia.-Papacleovoulou, G., Nikolova, V., Oduwole, O., Chambers, J., Vazquez-Lopez, M., Jansen, E., Nicolaides, K., Parker, M., Williamson, C. Gestational disruptions in metabolic rhythmicity of the liver, muscle, and placenta affect fetal size.


Asunto(s)
Ritmo Circadiano , Macrosomía Fetal/metabolismo , Hígado/metabolismo , Músculo Esquelético/metabolismo , Placenta/metabolismo , Factores de Transcripción ARNTL/metabolismo , Adaptación Fisiológica , Animales , Femenino , Macrosomía Fetal/etiología , Humanos , Lipogénesis , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Triglicéridos/metabolismo
6.
Am J Physiol Endocrinol Metab ; 313(4): E463-E472, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28420650

RESUMEN

Human pregnancy is associated with enhanced de novo lipogenesis in the early stages followed by hyperlipidemia during advanced gestation. Liver X receptors (LXRs) are oxysterol-activated nuclear receptors that stimulate de novo lipogenesis and also promote the efflux of cholesterol from extrahepatic tissues followed by its transport back to the liver for biliary excretion. Although LXR is recognized as a master regulator of triglyceride and cholesterol homeostasis, it is unknown whether it facilitates the gestational adaptations in lipid metabolism. To address this question, biochemical profiling, protein quantification, and gene expression studies were used, and gestational metabolic changes in T0901317-treated wild-type mice and Lxrab-/- mutants were investigated. Here, we show that altered LXR signaling contributes to the enhanced lipogenesis in early pregnancy by increasing the expression of hepatic Fas and stearoyl-CoA desaturase 1 (Scd1). Both the pharmacological activation of LXR with T0901317 and the genetic ablation of its two isoforms disrupted the increase in hepatic fatty acid biosynthesis and the development of hypertriglyceridemia during early gestation. We also demonstrate that absence of LXR enhances maternal white adipose tissue lipolysis, causing abnormal accumulation of triglycerides, cholesterol, and free fatty acids in the fetal liver. Together, these data identify LXR as an important factor in early-pregnancy lipogenesis that is also necessary to protect against abnormalities in fetoplacental lipid homeostasis.


Asunto(s)
Metabolismo de los Lípidos , Lipogénesis , Receptores X del Hígado/genética , Embarazo/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G/genética , Animales , Western Blotting , Femenino , Feto/metabolismo , Perfilación de la Expresión Génica , Homeostasis , Hidrocarburos Fluorados/farmacología , Receptores X del Hígado/agonistas , Receptores X del Hígado/metabolismo , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Placenta/metabolismo , Embarazo/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Estearoil-CoA Desaturasa/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Sulfonamidas/farmacología , Receptor fas/genética
7.
J Lipid Res ; 55(12): 2479-90, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25325755

RESUMEN

Arachidonic acid (AA) is a major PUFA that has been implicated in the regulation of adipogenesis. We examined the effect of a short exposure to AA at different stages of 3T3-L1 adipocyte differentiation. AA caused the upregulation of fatty acid binding protein 4 (FABP4/aP2) following 24 h of differentiation. This was mediated by the prostaglandin F(2α) (PGF(2α)), as inhibition of cyclooxygenases or PGF(2α) receptor signaling counteracted the AA-mediated aP2 induction. In addition, calcium, protein kinase C, and ERK are all key elements of the pathway through which AA induces the expression of aP2. We also show that treatment with AA during the first 24 h of differentiation upregulates the expression of the transcription factor Fos-related antigen 1 (Fra-1) via the same pathway. Finally, treatment with AA for 24 h at the beginning of the adipocyte differentiation is sufficient to inhibit the late stages of adipogenesis through a Fra-1-dependent pathway, as Fra-1 knockdown rescued adipogenesis. Our data show that AA is able to program the differentiation potential of preadipocytes by regulating gene expression at the early stages of adipogenesis.


Asunto(s)
Adipocitos Blancos/metabolismo , Adipogénesis , Ácido Araquidónico/metabolismo , Proteínas de Unión a Ácidos Grasos/agonistas , Regulación del Desarrollo de la Expresión Génica , Proteínas Proto-Oncogénicas c-fos/agonistas , Receptores de Prostaglandina/agonistas , Células 3T3-L1 , Adipocitos Blancos/citología , Adipocitos Blancos/enzimología , Animales , Señalización del Calcio , Dinoprost/metabolismo , Regulación hacia Abajo , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Cinética , Sistema de Señalización de MAP Quinasas , Ratones , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Prostaglandina/antagonistas & inhibidores , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Transducción de Señal , Regulación hacia Arriba
8.
Hepatology ; 57(2): 716-26, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22961653

RESUMEN

UNLABELLED: Intrahepatic cholestasis of pregnancy (ICP) is the most prevalent pregnancy-specific liver disease and is associated with an increased risk of adverse fetal outcomes, including preterm labor and intrauterine death. The endocrine signals that cause cholestasis are not known but 3α-sulfated progesterone metabolites have been shown to be elevated in ICP, leading us to study the impact of sulfated progesterone metabolites on farnesoid X receptor (FXR)-mediated bile acid homeostasis pathways. Here we report that the 3ß-sulfated progesterone metabolite epiallopregnanolone sulfate is supraphysiologically raised in the serum of ICP patients. Mice challenged with cholic acid developed hypercholanemia and a hepatic gene expression profile indicative of FXR activation. However, coadministration of epiallopregnanolone sulfate with cholic acid exacerbated the hypercholanemia and resulted in aberrant gene expression profiles for hepatic bile acid-responsive genes consistent with cholestasis. We demonstrate that levels of epiallopregnanolone sulfate found in ICP can function as a partial agonist for FXR, resulting in the aberrant expression of bile acid homeostasis genes in hepatoma cell lines and primary human hepatocytes. Furthermore, epiallopregnanolone sulfate inhibition of FXR results in reduced FXR-mediated bile acid efflux and secreted FGF19. Using cofactor recruitment assays, we show that epiallopregnanolone sulfate competitively inhibits bile acid-mediated recruitment of cofactor motifs to the FXR-ligand binding domain. CONCLUSION: Our results reveal a novel molecular interaction between ICP-associated levels of the 3ß-sulfated progesterone metabolite epiallopregnanolone sulfate and FXR that couples the endocrine component of pregnancy in ICP to abnormal bile acid homeostasis.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colestasis Intrahepática/metabolismo , Complicaciones del Embarazo/metabolismo , Pregnanolona/análogos & derivados , Progesterona/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Ésteres del Ácido Sulfúrico/sangre , Animales , Colestasis/inducido químicamente , Ácido Cólico , Femenino , Homeostasis , Humanos , Ratones , Ratones Endogámicos C57BL , Fenotipo , Embarazo , Pregnanolona/sangre , Receptores Citoplasmáticos y Nucleares/agonistas
9.
Biochim Biophys Acta ; 1812(8): 879-87, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21073948

RESUMEN

Nuclear receptor signalling is essential for physiological processes such as metabolism, development, and reproduction. Alterations in the endocrine state that naturally occur during pregnancy result in maternal adaptations to support the feto-placental unit. A series of studies have shown that nuclear receptor signalling is involved in maternal adaptations of bile acid, cholesterol, and lipid homeostasis pathways to ensure maintenance of the nutritional demands of the fetus. We discuss regulation of hepatic nuclear receptors and their target genes in pregnancy and their impact on the development of disorders such as intrahepatic cholestasis of pregnancy and oestrogen-induced hepatotoxicity. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Metabolismo de los Lípidos , Embarazo/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Ritmo Circadiano , Estrógenos/fisiología , Femenino , Expresión Génica , Humanos , Hígado/metabolismo , Ratones , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal
10.
Dig Dis ; 29(1): 58-61, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21691106

RESUMEN

BACKGROUND/AIMS: Intrahepatic cholestasis of pregnancy (ICP) is complicated by spontaneous preterm labor, fetal anoxia and unexplained fetal death. We aim to evaluate the mechanisms by which raised fetal bile acids cause placental abnormalities and fetal cardiac pathology. METHODS: The study was performed using placental samples taken from ICP pregnancies, placental explant culture, neonatal and adult cardiomyocytes, and murine and human embryonic stem cell-derived cardiomyocytes. RESULTS: Maternal cholestasis causes a placental phenotype with histological abnormalities. This can be evaluated using placental explant cultures. Taurocholate, the principal bile acid raised in the fetal compartment in ICP, causes abnormal cardiomyocyte contraction, rhythm and desynchronization of calcium dynamics. To extend our observations that the muscarinic M2 receptor plays a role in bile acid-induced arrhythmia in cardiomyocytes, we are developing a model containing mixed cell populations to represent the fetal and maternal hearts. This will be used to evaluate the underlying mechanisms to explain fetal arrhythmia in the presence of cholestasis. CONCLUSION: Bile acids signal via a spectrum of pathways in the placenta and the fetal heart.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colestasis Intrahepática/metabolismo , Feto/metabolismo , Complicaciones del Embarazo/metabolismo , Transducción de Señal , Femenino , Corazón Fetal , Humanos , Intercambio Materno-Fetal , Embarazo
11.
Endocr J ; 57(7): 651-6, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20467160

RESUMEN

Estrogen-secreting adrenal cancers are extremely rare, with feminizing symptoms attributed to aromatase expression in the adrenal tumor. We describe a case of hypogonadotropic hypogonadism as a consequence of aberrant aromatase expression in a patient with adrenocortical adenocarcinoma. A 54 year-old man presented with a two month history of gynecomastia and reduced libido. Endocrine biochemistry at presentation showed hypogonadotropic hypogonadism (LH 2.4 U/L, FSH <1.0 IU/L, testosterone 2.8 nmol/L) with increased serum estrone (E(1), 821 pmol/L) and estradiol (E(2), 797 pmol/L) and subclinical ACTH-independent hypercortisolism (serum cortisol post 1mg overnight dexamethasone suppression test, 291 nmol/L). A right adrenal mass was identified on CT scanning and the patient underwent an open adrenalectomy. Post-operative evaluation showed normalization of serum levels of E(1) (95 pmol/L), E(2 )(109 pmol/L), testosterone (11.4 nmol/L), LH (4.1 U/L) and FSH (5.9 IU/L), and of cortisol dynamics. Immunohistochemistry of the adrenal cancer confirmed aberrant expression of aromatase in most, although not all, carcinoma cells. Transcripts associated with utilization of promoters II, I.1 and I.3 were prominently represented in the tumor aromatase mRNA. This case highlights that clinical features of feminizing adrenocortical carcinomas can be secondary to estrogen production by aberrantly transcribed and translated aromatase within the tumor. Even in males, gonadotropin secretion is subject to predominantly estrogen-mediated feedback-inhibition. The diagnosis of adrenocortical adenocarcinoma should be considered in men presenting with low testosterone and gonadotropins, particularly in the presence of feminizing features.


Asunto(s)
Adenocarcinoma/genética , Neoplasias de la Corteza Suprarrenal/genética , Aromatasa/genética , Hipogonadismo/genética , Adenocarcinoma/complicaciones , Adenocarcinoma/metabolismo , Neoplasias de la Corteza Suprarrenal/complicaciones , Neoplasias de la Corteza Suprarrenal/metabolismo , Adulto , Edad de Inicio , Aromatasa/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Predisposición Genética a la Enfermedad , Humanos , Hipogonadismo/epidemiología , Hipogonadismo/etiología , Hipogonadismo/metabolismo , Masculino , Persona de Mediana Edad
12.
Sci Rep ; 10(1): 10361, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32587408

RESUMEN

Perturbations in the intrauterine environment can result in lifelong consequences for metabolic health during postnatal life. Intrahepatic cholestasis of pregnancy (ICP) can predispose offspring to metabolic disease in adulthood, likely due to a combination of the effects of increased bile acids, maternal dyslipidemia and deranged maternal and fetal lipid homeostasis. Whereas ursodeoxycholic acid (UDCA) is a commonly used treatment for ICP, no studies have yet addressed whether it can also prevent the metabolic effects of ICP in the offspring and fetoplacental unit. We therefore analyzed the lipid profile of fetal serum from untreated ICP, UDCA-treated ICP and uncomplicated pregnancies and found that UDCA ameliorates ICP-associated fetal dyslipidemia. We then investigated the effects of UDCA in a mouse model of hypercholanemic pregnancy and showed that it induces hepatoprotective mechanisms in the fetal liver, reduces hepatic fatty acid synthase (Fas) expression and improves glucose tolerance in the adult offspring. Finally, we showed that ICP leads to epigenetic changes in pathways of relevance to the offspring phenotype. We therefore conclude that UDCA can be used as an intervention in pregnancy to reduce features of metabolic disease in the offspring of hypercholanemic mothers.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colestasis Intrahepática/tratamiento farmacológico , Dislipidemias/prevención & control , Epigenoma/efectos de los fármacos , Feto/efectos de los fármacos , Placenta/efectos de los fármacos , Complicaciones del Embarazo/tratamiento farmacológico , Ácido Ursodesoxicólico/farmacología , Adulto , Animales , Colestasis Intrahepática/metabolismo , Colestasis Intrahepática/patología , Dislipidemias/epidemiología , Dislipidemias/metabolismo , Femenino , Feto/metabolismo , Feto/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Placenta/metabolismo , Placenta/patología , Embarazo , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/patología
13.
Sci Rep ; 10(1): 3895, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32127609

RESUMEN

Ursodeoxycholic acid (UDCA) treatment can reduce itch and lower endogenous serum bile acids in intrahepatic cholestasis of pregnancy (ICP). We sought to determine how it could influence the gut environment in ICP to alter enterohepatic signalling. The gut microbiota and bile acid content were determined in faeces from 35 pregnant women (14 with uncomplicated pregnancies and 21 with ICP, 17 receiving UDCA). Faecal bile salt hydrolase activity was measured using a precipitation assay. Serum fibroblast growth factor 19 (FGF19) and 7α-hydroxy-4-cholesten-3-one (C4) concentrations were measured following a standardised diet for 21 hours. Women with a high ratio of Bacteroidetes to Firmicutes were more likely to be treated with UDCA (Fisher's exact test p = 0.0178) than those with a lower ratio. Bile salt hydrolase activity was reduced in women with low Bacteroidetes:Firmicutes. Women taking UDCA had higher faecal lithocholic acid (p < 0.0001), with more unconjugated bile acids than women with untreated ICP or uncomplicated pregnancy. UDCA-treatment increased serum FGF19, and reduced C4 (reflecting lower bile acid synthesis). During ICP, UDCA treatment can be associated with enrichment of the gut microbiota with Bacteroidetes. These demonstrate high bile salt hydrolase activity, which deconjugates bile acids enabling secondary modification to FXR agonists, enhancing enterohepatic feedback via FGF19.


Asunto(s)
Amidohidrolasas/genética , Bacteroidetes/efectos de los fármacos , Bacteroidetes/genética , Colestasis Intrahepática/microbiología , Regulación Bacteriana de la Expresión Génica , Intestinos/microbiología , Complicaciones del Embarazo/microbiología , Ácido Ursodesoxicólico/farmacología , Animales , Estudios de Casos y Controles , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Ratones , Embarazo
14.
Mol Hum Reprod ; 15(6): 379-92, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19414525

RESUMEN

The human ovarian surface epithelium (hOSE) is a squamous-to-cuboidal layer that surrounds the ovary. hOSE undergoes injury and repair cycles as a result of ovulation-induced inflammation, an event relevant to the development of epithelial ovarian cancer (EOC). Locally produced steroids mediate the response to inflammation. 3beta-Hydroxysteroid dehydrogenase (3beta-HSD) drives the intracrine generation of progestogens and androgens that potentially affect cell survival and proliferation. We therefore investigated the regulation of 3beta-HSD along with downstream steroid signalling in hOSE. Double immunofluorescence of cultured primary hOSE cells confirmed the expression of 3beta-HSD protein Interleukin (IL). IL-1alpha treatment of primary cells to mimic ovulation-associated inflammation suppressed 3beta-HSD1 expression and stimulated 3beta-HSD2 mRNA (P < 0.001), without affecting total 3beta-HSD protein and activity or androgen or progesterone receptor (PR) mRNA levels. Conversely, IL-4 as a proxy for a post-ovulatory healing cytokine increased both 3beta-HSD transcripts, total protein and activity (P < 0.01). IL-4 also suppressed androgen receptor expression (P < 0.01) without affecting that of the PR, thereby potentially sustaining both progesterone biosynthesis and its underlying signalling in the ovarian surface. 3beta-HSD protein was immunodetectable in primary ascites of women who were diagnosed with EOC but both mRNA transcripts were diminished relative to normal cells (P < 0.05). Notably, this difference was countered by IL-4 treatment (P < 0.01). We conclude that stimulation by IL-4 could be physiologically relevant to post-ovulatory ovarian healing and suggest a novel therapeutic strategy for the activation of progesterone-associated apoptosis in ovarian cancer. Also, our results suggest an attenuation of 3beta-HSD expression in EOC although further studies are required for confirmation.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/metabolismo , Citocinas/farmacología , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Ovario/efectos de los fármacos , Ovario/metabolismo , 17-Hidroxiesteroide Deshidrogenasas/genética , Adulto , Línea Celular , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Técnicas In Vitro , Interleucina-1alfa/farmacología , Interleucina-4/farmacología , Persona de Mediana Edad , Neoplasias Ováricas/genética , Ovario/citología , Ovario/patología , Adulto Joven
15.
Mol Cell Endocrinol ; 301(1-2): 65-73, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-18778748

RESUMEN

Ovulation-associated inflammation with accompanied cytokines and reproductive hormones impact upon the human ovarian surface epithelium (hOSE) and probably have a role in the aetiology of ovarian cancer. Progesterone and progestin-related events, i.e. pregnancy and oral contraception, protect from the disease. We have investigated the pre-receptor metabolism of progesterone in primary hOSE cells and an immortalised hOSE cell line, OSE-C2, focusing on transcriptional regulation of 3beta-hydroxysteroid dehydrogenase (3beta-HSD) by inflammatory, anti-inflammatory and apoptotic factors. In hOSE cells, we show that anti-inflammatory effects of IL-1alpha and IL-4 on 3beta-HSD2 mRNA involve a p38 MAPK signalling pathway, whereas pro-inflammatory response of IL-1alpha to 3beta-HSD1 mRNA involves a NF-kappaB inflammatory pathway. In OSE-C2 cells, retinoic acid and transforming growth factor-beta1 massively induce 3beta-HSD1 mRNA levels. In conclusion, we elaborate several mechanisms for intracrine formation of progesterone in hOSE that could contribute in the development of novel strategies to prevent, diagnose and/or treat ovarian cancer.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/metabolismo , Epitelio/enzimología , Ovario/enzimología , Receptores de Esteroides/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/genética , Adulto , Línea Celular , Epitelio/efectos de los fármacos , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/enzimología , Interleucina-1alfa/farmacología , Interleucina-4/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/metabolismo , Ovario/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Tretinoina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Mol Cell Endocrinol ; 300(1-2): 115-20, 2009 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-19026713

RESUMEN

Adrenocortical carcinoma is an uncommon malignancy and feminizing symptoms secondary to adrenal estrogen-secretion are extremely rare. The direct secretion of estradiol by adrenocortical tumors requires, in addition to the expression of aromatase (CYP19), the expression of one or more of the reductive 17beta-hydroxysteroid dehydrogenases. The expression of CYP19 transcripts and protein were markedly induced in the H295 adrenocortical carcinoma cell line after treatment with either forskolin or vasoactive intestinal peptide (VIP). Western immunoblotting demonstrated a marked induction of the CYP19 protein of characteristic size after only a short (6h) treatment period with VIP or forskolin. The CYP19 mRNA transcripts were derived from both promoters PII (Ic) and I.3 (Id) after treatment with both agents. The reductive type 5 17beta-hydroxysteroid dehydrogenase (AKR1C3) was also constitutively expressed in the H295 cells but neither its mRNA transcript nor protein levels were altered after forskolin or VIP treatment. Western immunoblotting of an estrogen-secreting adrenal carcinoma revealed notable levels of both aromatase and AKR1C3 expression while an aldosterone-producing adrenal adenoma lacked aromatase expression and showed a reduced level of AKR1C3 expression. Immunohistochemistry of the carcinoma-bearing adrenal revealed localization of AKR1C3 not only in the tumor but also principally in the zona reticularis of the normal adrenal tissue. Adrenal aromatase and AKR1C3 expression therefore appear to be features of adrenocortical malignancies that are associated with biosynthesis of active estrogen.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/metabolismo , Línea Celular Tumoral , Estrógenos/biosíntesis , 17-Hidroxiesteroide Deshidrogenasas/genética , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , 3-Hidroxiesteroide Deshidrogenasas , Neoplasias de la Corteza Suprarrenal/patología , Adulto , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Aromatasa/genética , Aromatasa/metabolismo , Colforsina/metabolismo , Citocromo P-450 CYP11B2/genética , Citocromo P-450 CYP11B2/metabolismo , Femenino , Humanos , Hidroxiprostaglandina Deshidrogenasas , Progesterona Reductasa/genética , Progesterona Reductasa/metabolismo , Regiones Promotoras Genéticas , Esteroide 11-beta-Hidroxilasa/genética , Esteroide 11-beta-Hidroxilasa/metabolismo , Esteroide 17-alfa-Hidroxilasa/genética , Esteroide 17-alfa-Hidroxilasa/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
17.
Sci Rep ; 7(1): 10671, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28878263

RESUMEN

Pregnancy is associated with profound maternal metabolic changes, necessary for the growth and development of the fetus, mediated by reproductive signals acting on metabolic organs. However, the role of brown adipose tissue (BAT) in regulating gestational metabolism is unknown. We show that BAT phenotype is lost in murine pregnancy, while there is a gain of white adipose tissue (WAT)-like features. This is characterised by reduced thermogenic capacity and mitochondrial content, accompanied by increased levels of markers of WAT and lipid accumulation. Surgical ablation of BAT prior to conception caused maternal and fetal hyperlipidemia, and consequently larger fetuses. We show that BAT phenotype is altered from day 5 of gestation, implicating early pregnancy factors, which was confirmed by reduced expression of BAT markers in progesterone challenged oophorectomised mice. Moreover, in vitro data using primary BAT cultures show a direct impact of progesterone on expression of Ucp1. These data demonstrate that progesterone mediates a phenotypic change in BAT, which contributes to the gestational metabolic environment, and thus overall fetal size.


Asunto(s)
Desarrollo Fetal , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Biomarcadores , Diferenciación Celular , Metabolismo Energético , Femenino , Metabolismo de los Lípidos , Lípidos/sangre , Masculino , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Fenotipo , Embarazo , Progesterona/metabolismo , Transducción de Señal
18.
Sci Rep ; 7(1): 2338, 2017 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-28539583

RESUMEN

Transient receptor potential canonical 5 (TRPC5), a calcium-permeable, non-selective cation channel is expressed in the periphery, but there is limited knowledge of its regulatory roles in vivo. Endogenous modulators of TRPC5 include a range of phospholipids that have an established role in liver disease, including lysophosphatidylcholine (LPC). Cholestasis is characterized by impairment of excretion of bile acids, leading to elevation of hepatic bile acids. We investigated the contribution of TRPC5 in a murine model of cholestasis. Wild-type (WT) and TRPC5 knock-out (KO) mice were fed a diet supplemented with 0.5% cholic acid (CA) for 21 days. CA-diet supplementation resulted in enlargement of the liver in WT mice, which was ameliorated in TRPC5 KO mice. Hepatic bile acid and lipid content was elevated in WT mice, with a reduction observed in TRPC5 KO mice. Consistently, liver enzymes were significantly increased in cholestatic WT mice and significantly blunted in TRPC5 KO mice. Localized dyslipidaemia, secondary to cholestasis, was investigated utilizing a selected lipid analysis. This revealed significant perturbations in the lipid profile following CA-diet feeding, with increased cholesterol, triglycerides and phospholipids, in WT, but not TRPC5 KO mice. Our results suggest that activation of TRPC5 contributes to the development of cholestasis and associated dyslipidemia. Modulation of TRPC5 activity may present as a novel therapeutic target for liver disease.


Asunto(s)
Colestasis/metabolismo , Dislipidemias/metabolismo , Hígado/metabolismo , Canales Catiónicos TRPC/fisiología , Animales , Ácidos y Sales Biliares/sangre , Ácidos y Sales Biliares/metabolismo , Colestasis/genética , Colesterol 7-alfa-Hidroxilasa/genética , Colesterol 7-alfa-Hidroxilasa/metabolismo , Dislipidemias/genética , Expresión Génica , Lípidos/análisis , Hígado/patología , Masculino , Ratones Endogámicos ICR , Ratones Noqueados , Canales Catiónicos TRPC/deficiencia , Canales Catiónicos TRPC/genética
19.
Mol Cell Endocrinol ; 368(1-2): 120-8, 2013 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-23159988

RESUMEN

Bile acids have been traditionally thought of as having an important role in fat emulsification. It is now emerging that they act as important signalling molecules that not only autoregulate their own synthesis but also influence lipid and glucose metabolism. Although, the mechanisms that underlie the regulation of bile acid homeostasis have been well characterised in normal physiology, the impact of pregnancy on bile acid regulation is still poorly understood. This review summarises the main regulatory mechanisms underlying bile acid homeostasis and discusses how pregnancy, a unique physiological state, can modify them. The fetoplacental adaptations that protect against fetal bile acid toxicity are reviewed. We highlight the importance of bile acid regulation during gestation by discussing the liver disease of pregnancy, intrahepatic cholestasis of pregnancy (ICP) and how genetic, endocrine and environmental factors contribute to the disease aetiology at a cellular and molecular level.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colestasis Intrahepática/metabolismo , Colesterol/metabolismo , Homeostasis , Complicaciones del Embarazo/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Colagogos y Coleréticos/uso terapéutico , Colestasis Intrahepática/tratamiento farmacológico , Colestasis Intrahepática/etiología , Dieta , Femenino , Humanos , Metabolismo de los Lípidos , Hígado/metabolismo , Intercambio Materno-Fetal , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/etiología , Ácido Ursodesoxicólico/uso terapéutico
20.
J Clin Invest ; 123(7): 3172-81, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23934127

RESUMEN

The intrauterine environment is a major contributor to increased rates of metabolic disease in adults. Intrahepatic cholestasis of pregnancy (ICP) is a liver disease of pregnancy that affects 0.5%-2% of pregnant women and is characterized by increased bile acid levels in the maternal serum. The influence of ICP on the metabolic health of offspring is unknown. We analyzed the Northern Finland birth cohort 1985-1986 database and found that 16-year-old children of mothers with ICP had altered lipid profiles. Males had increased BMI, and females exhibited increased waist and hip girth compared with the offspring of uncomplicated pregnancies. We further investigated the effect of maternal cholestasis on the metabolism of adult offspring in the mouse. Females from cholestatic mothers developed a severe obese, diabetic phenotype with hepatosteatosis following a Western diet, whereas matched mice not exposed to cholestasis in utero did not. Female littermates were susceptible to metabolic disease before dietary challenge. Human and mouse studies showed an accumulation of lipids in the fetoplacental unit and increased transplacental cholesterol transport in cholestatic pregnancy. We believe this is the first report showing that cholestatic pregnancy in the absence of altered maternal BMI or diabetes can program metabolic disease in the offspring.


Asunto(s)
Colestasis Intrahepática/complicaciones , Hígado Graso/etiología , Obesidad/etiología , Efectos Tardíos de la Exposición Prenatal/etiología , Tejido Adiposo Blanco/metabolismo , Adolescente , Animales , Ácidos y Sales Biliares/metabolismo , Glucemia , Estudios de Casos y Controles , Células Cultivadas , Colestasis Intrahepática/metabolismo , Dieta , Susceptibilidad a Enfermedades/etiología , Susceptibilidad a Enfermedades/metabolismo , Epigénesis Genética , Hígado Graso/metabolismo , Femenino , Homeostasis , Humanos , Insulina/sangre , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Placenta/metabolismo , Embarazo , Complicaciones del Embarazo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Fenómenos Fisiologicos de la Nutrición Prenatal , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA