Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
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
2.
Hepatology ; 70(1): 276-293, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30983011

RESUMEN

Pregnancy is associated with progressive hypercholanemia, hypercholesterolemia, and hypertriglyceridemia, which can result in metabolic disease in susceptible women. Gut signals modify hepatic homeostatic pathways, linking intestinal content to metabolic activity. We sought to identify whether enteric endocrine signals contribute to raised serum bile acids observed in human and murine pregnancies, by measuring fibroblast growth factor (FGF) 19/15 protein and mRNA levels, and 7α-hydroxy-4-cholesten-3-one. Terminal ileal farnesoid X receptor (FXR)-mediated gene expression and apical sodium bile acid transporter (ASBT) protein concentration were measured by qPCR and western blotting. Shotgun whole-genome sequencing and ultra-performance liquid chromatography tandem mass spectrometry were used to determine the cecal microbiome and metabonome. Targeted and untargeted pathway analyses were performed to predict the systemic effects of the altered metagenome and metabolite profiles. Dietary CA supplementation was used to determine whether the observed alterations could be overcome by intestinal bile acids functioning as FXR agonists. Human and murine pregnancy were associated with reduced intestinal FXR signaling, with lower FGF19/15 and resultant increased hepatic bile acid synthesis. Terminal ileal ASBT protein was reduced in murine pregnancy. Cecal bile acid conjugation was reduced in pregnancy because of elevated bile salt hydrolase-producing Bacteroidetes. CA supplementation induced intestinal FXR signaling, which was not abrogated by pregnancy, with strikingly similar changes to the microbiota and metabonome as identified in pregnancy. Conclusion: The altered intestinal microbiota of pregnancy enhance bile acid deconjugation, reducing ileal bile acid uptake and lowering FXR induction in enterocytes. This exacerbates the effects mediated by reduced bile acid uptake transporters in pregnancy. Thus, in pregnant women and mice, there is reduced FGF19/15-mediated hepatic repression of hepatic bile acid synthesis, resulting in hypercholanemia.


Asunto(s)
Ácidos Cólicos/sangre , Microbioma Gastrointestinal , Reabsorción Intestinal , Embarazo/sangre , Receptores Citoplasmáticos y Nucleares/metabolismo , Amidohidrolasas/genética , Animales , Bacteroides/aislamiento & purificación , Ciego/efectos de los fármacos , Ciego/microbiología , Ácidos Cólicos/farmacología , Enterocitos/efectos de los fármacos , Femenino , Humanos , Ratones Endogámicos C57BL , Receptores Citoplasmáticos y Nucleares/agonistas
3.
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
4.
Hepatology ; 63(4): 1287-98, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26426865

RESUMEN

UNLABELLED: A challenge in obstetrics is to distinguish pathological symptoms from those associated with normal changes of pregnancy, typified by the need to differentiate whether gestational pruritus of the skin is an early symptom of intrahepatic cholestasis of pregnancy (ICP) or due to benign pruritus gravidarum. ICP is characterized by raised serum bile acids and complicated by spontaneous preterm labor and stillbirth. A biomarker for ICP would be invaluable for early diagnosis and treatment and to enable its differentiation from other maternal diseases. Three progesterone sulfate compounds, whose concentrations have not previously been studied, were newly synthesized and assayed in the serum of three groups of ICP patients and found to be significantly higher in ICP at 9-15 weeks of gestation and prior to symptom onset (group 1 cases/samples: ICP n = 35/80, uncomplicated pregnancy = 29/100), demonstrating that all three progesterone sulfates are prognostic for ICP. Concentrations of progesterone sulfates were associated with itch severity and, in combination with autotaxin, distinguished pregnant women with itch that would subsequently develop ICP from pruritus gravidarum (group 2: ICP n = 41, pruritus gravidarum n = 14). In a third group of first-trimester samples all progesterone sulfates were significantly elevated in serum from low-risk asymptomatic women who subsequently developed ICP (ICP/uncomplicated pregnancy n = 54/51). Finally, we show mechanistically that progesterone sulfates mediate itch by evoking a Tgr5-dependent scratch response in mice. CONCLUSION: Our discovery that sulfated progesterone metabolites are a prognostic indicator for ICP will help predict onset of ICP and distinguish it from benign pruritus gravidarum, enabling targeted obstetric care to a high-risk population. Delineation of a progesterone sulfate-TGR5 pruritus axis identifies a therapeutic target for itch management in ICP.


Asunto(s)
Ácidos y Sales Biliares/sangre , Colestasis Intrahepática/diagnóstico , Complicaciones del Embarazo/diagnóstico , Resultado del Embarazo , Preñez , Progesterona/metabolismo , Prurito/diagnóstico , Adulto , Animales , Conducta Animal , Estudios de Casos y Controles , Colestasis Intrahepática/sangre , Cromatografía Líquida de Alta Presión/métodos , Femenino , Humanos , Oportunidad Relativa , Valor Predictivo de las Pruebas , Embarazo , Complicaciones del Embarazo/sangre , Prurito/metabolismo , Curva ROC , Índice de Severidad de la Enfermedad , Espectrometría de Masas en Tándem/métodos , Reino Unido
5.
Dig Dis ; 33(3): 300-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26045261

RESUMEN

Sulfated progesterone metabolites rise 100-fold in the third trimester of human pregnancy and have been shown to be elevated further in the gestational disorder intrahepatic cholestasis of pregnancy (ICP). Typical concentrations of progesterone sulfates range from 1 to 10 µmol/L in an uncomplicated pregnancy and rise to approximately 40 µmol/L in ICP. At this level they can influence bile acid and lipid metabolism. Studies using human and rodent specimens have shown that sulfated metabolites of progesterone competitively inhibit bile acid homeostasis pathways by functioning as partial agonists of farnesoid X receptor (FXR). This explains the loss of induction of FXR target genes in ICP, and may explain susceptibility to hypercholanaemia and dyslipidaemia in the second half of human pregnancy. Furthermore, progesterone sulfates are competitive inhibitors of biliary influx (NTCP) and efflux (BSEP) transport proteins, actions likely to further exacerbate hypercholanaemia and cholestasis.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colestasis Intrahepática/metabolismo , Ácidos Cólicos/sangre , Complicaciones del Embarazo/metabolismo , Progesterona/metabolismo , Receptores Citoplasmáticos y Nucleares/agonistas , Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Colestasis Intrahepática/etiología , Femenino , Homeostasis , Humanos , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Embarazo , Complicaciones del Embarazo/etiología , Pregnanolona/análogos & derivados , Pregnanolona/metabolismo , Simportadores/metabolismo
6.
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
7.
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
8.
Biochem J ; 434(1): 49-60, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21118154

RESUMEN

LKB1 is a 'master' protein kinase implicated in the regulation of metabolism, cell proliferation, cell polarity and tumorigenesis. However, the long-term role of LKB1 in hepatic function is unknown. In the present study, it is shown that hepatic LKB1 plays a key role in liver cellular architecture and metabolism. We report that liver-specific deletion of LKB1 in mice leads to defective canaliculi and bile duct formation, causing impaired bile acid clearance and subsequent accumulation of bile acids in serum and liver. Concomitant with this, it was found that the majority of BSEP (bile salt export pump) was retained in intracellular pools rather than localized to the canalicular membrane in hepatocytes from LLKB1KO (liver-specific Lkb1-knockout) mice. Together, these changes resulted in toxic accumulation of bile salts, reduced liver function and failure to thrive. Additionally, circulating LDL (low-density lipoprotein)-cholesterol and non-esterified cholesterol levels were increased in LLKB1KO mice with an associated alteration in red blood cell morphology and development of hyperbilirubinaemia. These results indicate that LKB1 plays a critical role in bile acid homoeostasis and that lack of LKB1 in the liver results in cholestasis. These findings indicate a novel key role for LKB1 in the development of hepatic morphology and membrane targeting of canalicular proteins.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Canalículos Biliares/patología , Canalículos Biliares/fisiología , Hígado/anatomía & histología , Hígado/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP , Envejecimiento , Animales , Transporte Biológico/fisiología , Membrana Celular , Colesterol/metabolismo , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética
9.
J Biol Chem ; 285(22): 16504-12, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20177056

RESUMEN

Sulfated progesterone metabolite (P4-S) levels are raised in normal pregnancy and elevated further in intrahepatic cholestasis of pregnancy (ICP), a bile acid-liver disorder of pregnancy. ICP can be complicated by preterm labor and intrauterine death. The impact of P4-S on bile acid uptake was studied using two experimental models of hepatic uptake of bile acids, namely cultured primary human hepatocytes (PHH) and Na(+)-taurocholate co-transporting polypeptide (NTCP)-expressing Xenopus laevis oocytes. Two P4-S compounds, allopregnanolone-sulfate (PM4-S) and epiallopregnanolone-sulfate (PM5-S), reduced [(3)H]taurocholate (TC) uptake in a dose-dependent manner in PHH, with both Na(+)-dependent and -independent bile acid uptake systems significantly inhibited. PM5-S-mediated inhibition of TC uptake could be reversed by increasing the TC concentration against a fixed PM5-S dose indicating competitive inhibition. Experiments using NTCP-expressing Xenopus oocytes confirmed that PM4-S/PM5-S are capable of competitively inhibiting NTCP-mediated uptake of [(3)H]TC. Total serum PM4-S + PM5-S levels were measured in non-pregnant and third trimester pregnant women using liquid chromatography-electrospray tandem mass spectrometry and were increased in pregnant women, at levels capable of inhibiting TC uptake. In conclusion, pregnancy levels of P4-S can inhibit Na(+)-dependent and -independent influx of taurocholate in PHH and cause competitive inhibition of NTCP-mediated uptake of taurocholate in Xenopus oocytes.


Asunto(s)
Cobalto/química , Progesterona/química , Sodio/farmacología , Ácido Taurocólico/química , Animales , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Unión Competitiva , Línea Celular Tumoral , Colestasis , Relación Dosis-Respuesta a Droga , Femenino , Hepatocitos/metabolismo , Humanos , Modelos Biológicos , Oocitos/metabolismo , Péptidos/química , Embarazo , Sodio/química , Esteroides/metabolismo , Xenopus laevis
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
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.
J Cell Mol Med ; 13(9B): 3730-41, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19438812

RESUMEN

Embryonic stem cell-derived cardiomyocytes (ESC-CM) have many of the phenotypic properties of authentic cardiomyocytes, and great interest has been shown in their possibilities for modelling human disease. Obstetric cholestasis affects 1 in 200 pregnant women in the United Kingdom. It is characterized by raised serum bile acids and complicated by premature delivery and unexplained fetal death at late gestation. It has been suggested that the fetal death is caused by the enhanced arrhythmogenic effect of bile acids in the fetal heart, and shown that neonatal susceptibility to bile acid-induced arrhythmia is lost in the adult rat cardiomyocyte. However, the mechanisms of the observed bile acid effects are not fully understood and their in vivo study in human beings is difficult. Here we use ESC-CM from both human and mouse ESCs to test our proposal that immature cardiomyocytes are more susceptible to the effect of raised bile acids than mature ones. We show that early ESC-CM exhibit bile acid-induced disruption of rhythm, depression of contraction and desynchronization of cell coupling. In both species the ESC-CM become resistant to these arrhythmias as the cells mature, and this develops in line with the respective gestational periods of mouse and human. This represents the first demonstration of the use of ESC-CM as a model system for human cardiac pathology, and opens the way for both investigation of mechanisms and a high throughput screen for drug discovery.


Asunto(s)
Arritmias Cardíacas/patología , Ácidos y Sales Biliares/metabolismo , Enfermedades Fetales/patología , Miocitos Cardíacos/citología , Animales , Calcio/metabolismo , Diferenciación Celular , Colestasis/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Embarazo , Complicaciones Cardiovasculares del Embarazo , Factores de Tiempo
15.
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
16.
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
17.
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
18.
PLoS One ; 5(3): e9689, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20300620

RESUMEN

BACKGROUND: Intrahepatic cholestasis of pregnancy (ICP) is a common disease affecting up to 5% of pregnancies and which can cause fetal arrhythmia and sudden intrauterine death. We previously demonstrated that bile acid taurocholate (TC), which is raised in the bloodstream of ICP, can acutely alter the rate and rhythm of contraction and induce abnormal calcium destabilization in cultured neonatal rat cardiomyocytes (NRCM). Apart from their hepatic functions bile acids are ubiquitous signalling molecules with diverse systemic effects mediated by either the nuclear receptor FXR or by a recently discovered G-protein coupled receptor TGR5. We aim to investigate the mechanism of bile-acid induced arrhythmogenic effects in an in-vitro model of the fetal heart. METHODS AND RESULTS: Levels of bile acid transporters and nuclear receptor FXR were studied by quantitative real time PCR, western blot and immunostaining, which showed low levels of expression. We did not observe functional involvement of the canonical receptors FXR and TGR5. Instead, we found that TC binds to the muscarinic M(2) receptor in NRCM and serves as a partial agonist of this receptor in terms of inhibitory effect on intracellular cAMP and negative chronotropic response. Pharmacological inhibition and siRNA-knockdown of the M(2) receptor completely abolished the negative effect of TC on contraction, calcium transient amplitude and synchronisation in NRCM clusters. CONCLUSION: We conclude that in NRCM the TC-induced arrhythmia is mediated by the partial agonism at the M(2) receptor. This mechanism might serve as a promising new therapeutic target for fetal arrhythmia.


Asunto(s)
Arritmias Cardíacas/inducido químicamente , Ácidos y Sales Biliares/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Receptor Muscarínico M2/metabolismo , Animales , Animales Recién Nacidos , Núcleo Celular/metabolismo , Colestasis/inducido químicamente , Silenciador del Gen , Reacción en Cadena de la Polimerasa , Ratas , Ratas Wistar , Receptor Muscarínico M2/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
19.
PLoS One ; 4(12): e8285, 2009 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-20011604

RESUMEN

BACKGROUND: The fibroblast growth factors (FGFs) are key regulators of embryonic development, tissue homeostasis and tumour angiogenesis. Binding of FGFs to their receptor(s) results in activation of several intracellular signalling cascades including phosphoinositide 3-kinase (PI3K) and phospholipase C (PLC)gamma1. Here we investigated the basic FGF (FGF-2)-mediated activation of these enzymes in human umbilical vein endothelial cells (HUVECs) and defined their role in FGF-2-dependent cellular functions. METHODOLOGY/PRINCIPAL FINDINGS: We show that FGF-2 activates PLCgamma1 in HUVECs measured by analysis of total inositol phosphates production upon metabolic labelling of cells and intracellular calcium increase. We further demonstrate that FGF-2 activates PI3K, assessed by analysing accumulation of its lipid product phosphatidylinositol-3,4,5-P(3) using TLC and confocal microscopy analysis. PI3K activity is required for FGF-2-induced PLCgamma1 activation and the PI3K/PLCgamma1 pathway is involved in FGF-2-dependent cell migration, determined using Transwell assay, and in FGF-2-induced capillary tube formation (tubulogenesis assays in vitro). Finally we show that PI3K-dependent PLCgamma1 activation regulates FGF-2-mediated phosphorylation of Akt at its residue Ser473, determined by Western blotting analysis. This occurs through protein kinase C (PKC)alpha activation since dowregulation of PKCalpha expression using specific siRNA or blockade of its activity using chemical inhibition affects the FGF-2-dependent Ser473 Akt phosphorylation. Furthermore inhibition of PKCalpha blocks FGF-2-dependent cell migration. CONCLUSION/SIGNIFICANCE: These data elucidate the role of PLCgamma1 in FGF-2 signalling in HUVECs demonstrating its key role in FGF-2-dependent tubulogenesis. Furthermore these data unveil a novel role for PLCgamma1 as a mediator of PI3K-dependent Akt activation and as a novel key regulator of different Akt-dependent processes.


Asunto(s)
Capilares/efectos de los fármacos , Capilares/enzimología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfolipasa C gamma/metabolismo , Transducción de Señal/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Activación Enzimática/efectos de los fármacos , Humanos , Fosforilación/efectos de los fármacos , Proteína Quinasa C-alfa/metabolismo , Venas Umbilicales/citología
20.
Am J Physiol Heart Circ Physiol ; 287(1): H22-8, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-14962835

RESUMEN

The potassium channel blocker tetraethylammonium blocks the flow-induced increase in endothelial ICAM-1. We have investigated the subtype of potassium channel that modulates flow-induced increased expression of ICAM-1 on saphenous vein endothelium. Cultured human saphenous vein endothelial cells (HSVECs) or intact saphenous veins were perfused at fixed low and high flows in a laminar shear chamber or flow rig, respectively, in the presence or absence of potassium channel blockers. Expression of K(+) channels and endothelial ICAM-1 was measured by real-time polymerase chain reaction and/or immunoassays. In HSVECs, the application of 0.8 N/m(2) (8 dyn/cm(2)) shear stress resulted in a two- to fourfold increase in cellular ICAM-1 within 6 h (P < 0.001). In intact vein a similar shear stress, with pulsatile arterial pressure, resulted in a twofold increase in endothelial ICAM-1/CD31 staining area within 1.5 h (P < 0.001). Both increases in ICAM-1 were blocked by inclusion of 100 nM apamin in the vein perfusate, whereas other K(+) channel blockers were less effective. Two subtypes of small conductance Ca(2+)-activated K(+) channel (selectively blocked by apamin) were expressed in HSVECs and vein endothelium (SK3>SK2). Apamin blocked the upregulation of ICAM-1 on saphenous vein endothelium in response to increased flow to implicate small conductance Ca(2+)-activated K(+) channels in shear stress/flow-mediated signaling pathways.


Asunto(s)
Apamina/farmacología , Ácido Egtácico/análogos & derivados , Endotelio Vascular/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Vena Safena/fisiología , Calcio/metabolismo , Células Cultivadas , Quelantes/farmacología , Ácido Egtácico/farmacología , Endotelio Vascular/citología , Humanos , Membranas Intracelulares/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio Calcio-Activados/metabolismo , Flujo Sanguíneo Regional/fisiología , Vena Safena/citología , Estrés Mecánico
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda