Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Biol Rhythms ; 33(3): 289-301, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29761750

RESUMO

Obesity during pregnancy causes numerous maternal and fetal health complications, but the underlying mechanisms remain unclear. Adipose tissue dysfunction in obesity has previously been linked to disruption of the intrinsic adipose clock gene network that is crucial for normal metabolic function. This adipose clock also undergoes major change as part of the maternal metabolic adaptation to pregnancy, but whether this is affected by maternal obesity is unknown. Consequently, in this study we tested the hypothesis that obesity disturbs rhythmic gene expression in maternal adipose tissue across pregnancy. A rat model of maternal obesity was established by cafeteria (CAF) feeding, and adipose expression of clock genes and associated nuclear receptors ( Ppars and Pgc1α) was measured across days 15-16 and 21-22 of gestation (term = 23 days). CAF feeding suppressed the mesor and/or amplitude of adipose tissue clock genes (most notably Bmal1, Per2, and Rev-erbα) relative to chow-fed controls (CON) across both days of gestation. On day 15, the CAF diet also induced adipose Pparα, Pparδ, and Pgc1α rhythmicity but repressed that of Pparγ, while expression of Pparα, Pparδ, and Pgc1α was reduced at select time points. CAF mothers were hyperleptinemic at both stages of gestation, and at day 21 this effect was time-of-day dependent. Fetal plasma leptin exhibited clear rhythmicity, albeit with low amplitude, but interestingly these levels were unaffected by CAF feeding. Our data show that maternal obesity disrupts rhythmic expression of clock and metabolic genes in maternal adipose tissue and leads to maternal but not fetal hyperleptinemia.


Assuntos
Tecido Adiposo/metabolismo , Proteínas CLOCK/genética , Expressão Gênica , Obesidade/complicações , Tecido Adiposo/patologia , Animais , Ritmo Circadiano , Feminino , Feto/fisiologia , Leptina/sangue , Gravidez , Prenhez , Ratos
2.
Chronobiol Int ; 35(8): 1077-1087, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29659304

RESUMO

Obesity during pregnancy causes adverse maternal and fetal health outcomes and programs offspring for adult-onset diseases, including cardiovascular disease. Obesity also disrupts core body temperature (Tc) regulation in nonpregnant rodents; however, it is unknown whether obesity alters normal maternal Tc adaptations to pregnancy. Since Tc is influenced by the circadian system, and both obesity and pregnancy alter circadian biology, it was hypothesized that obesity disrupts the normal rhythmic patterns of Tc before and during gestation. Obesity was induced by cafeteria (CAF) feeding in female Wistar rats for 8 weeks prior to and during gestation, whereas control (CON) animals had free access to chow. Intraperitoneal temperature loggers measured daily Tc profiles throughout the study, while maternal body composition and leptin levels were assessed near term. Daily temperature profiles were examined for rhythmic features (mesor, amplitude and acrophase) by cosine regression analysis. CAF animals exhibited increased fat mass (93%) and associated hyperleptinemia (3.2-fold increase) compared to CON animals. CAF consumption reduced the average Tc (by up to 0.29°C) across the estrous cycle and most of pregnancy; however, Tc for CAF and CON animals converged toward the end of gestation. Obesity reduced the amplitude of Tc rhythms at estrus and proestrus and on day 8 of pregnancy, but increased the amplitude at day 20 of pregnancy. Photoperiod analysis revealed that obesity reduced Tc exclusively in the light period during pre-pregnancy but only during the dark period in late gestation. In conclusion, obesity alters rhythmic Tc profiles and reduces the magnitude of the Tc decline late in rat gestation, which may have implications for maternal health and fetal development.


Assuntos
Regulação da Temperatura Corporal , Ritmo Circadiano , Dieta Hiperlipídica , Ciclo Estral , Obesidade/fisiopatologia , Adiposidade , Animais , Modelos Animais de Doenças , Ciclo Estral/sangue , Feminino , Idade Gestacional , Leptina/sangue , Obesidade/sangue , Fotoperíodo , Gravidez , Ratos Wistar , Fatores de Tempo
3.
Biol Reprod ; 98(1): 75-88, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29186286

RESUMO

Maternal obesity induces pregnancy complications and disturbs fetal development, but the specific mechanisms underlying these outcomes are unclear. Circadian rhythms are implicated in metabolic complications associated with obesity, and maternal metabolic adaptations to pregnancy. Accordingly, obesity-induced circadian dysfunction may drive adverse outcomes in obese pregnancy. This study investigated whether maternal obesity alters the rhythmic expression of clock genes and associated nuclear receptors across maternal, fetal, and placental tissues. Wistar rats were maintained on a cafeteria (CAF) diet prior to and throughout gestation to induce maternal obesity. Maternal and fetal liver and placental labyrinth zone (LZ) were collected at four-hourly time points across days 15-16 and 21-22 of gestation (term = 23 days). Gene expression was analyzed by RT-qPCR. Expression of the accessory clock gene Nr1d1 was rhythmic in the maternal and fetal liver and LZ of chow-fed controls, but in each case CAF feeding reduced peak Nr1d1 expression. Obesity resulted in a phase advance (approx. 1.5 h) in the rhythms of several clock genes and Ppar-delta in maternal liver. Aside from Nr1d1, expression of clock genes was mostly arrhythmic in LZ and fetal liver, and was unaffected by the CAF diet. In conclusion, maternal obesity suppressed Nr1d1 expression across maternal, fetal, and placental compartments and phase-advanced the rhythms of maternal hepatic clock genes. Given the key role of Nr1d1 in regulating metabolic, vascular, and inflammatory processes, our data suggest that disruptions to rhythmic Nr1d1 expression in utero may contribute to programmed health complications in offspring of obese pregnancies.


Assuntos
Proteínas CLOCK/metabolismo , Regulação da Expressão Gênica/fisiologia , Redes Reguladoras de Genes/fisiologia , Fígado/metabolismo , Obesidade/metabolismo , Placenta/metabolismo , Animais , Proteínas CLOCK/genética , Feminino , Gravidez , Ratos , Ratos Wistar
4.
J Biol Rhythms ; 32(6): 534-549, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28920512

RESUMO

From the perspective of circadian biology, mammalian pregnancy presents an unusual biological scenario in which an entire circadian system (i.e., that of the fetus) is embodied within another (i.e., that of the mother). Moreover, both systems are likely to be influenced at their interface by a third player, the placenta. Successful pregnancy requires major adaptations in maternal physiology, many of which involve circadian changes that support the high metabolic demands of the growing fetus. A functional role for maternal circadian adaptations is implied by the effects of circadian disruption, which result in pregnancy complications including higher risks for miscarriage, preterm labor, and low birth weight. Various aspects of fetal physiology lead to circadian variation, at least in late gestation, but it remains unclear what drives this rhythmicity. It likely involves contributions from the maternal environment and possibly from the placenta and the developing intrinsic molecular clocks within fetal tissues. The role of the placenta is of particular significance because it serves not only to relay signals about the external environment (via the mother) but may also exhibit its own circadian rhythmicity. This review considers how the fetus may be influenced by dynamic circadian signals from the mother and the placenta during gestation, and how, in the face of these changing influences, a new fetal circadian system emerges. Particular emphasis is placed on the role of endocrine signals, most notably melatonin and glucocorticoids, as mediators of maternal-fetal circadian interactions, and on the expression of the clock gene in the 3 compartments. Further study is required to understand how the mother, placenta, and fetus interact across pregnancy to optimize circadian adaptations that support adequate growth and development of the fetus and its transition to postnatal life in a circadian environment.


Assuntos
Ritmo Circadiano , Placenta/fisiologia , Prenhez/fisiologia , Animais , Feminino , Desenvolvimento Fetal , Humanos , Gravidez
5.
Sci Rep ; 7(1): 2866, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28588270

RESUMO

Multimodal polymeric nanoparticles have many exciting diagnostic and therapeutic applications, yet their uptake and passage by the placenta, and applications in the treatment of pregnancy complications have not been thoroughly investigated. In this work, the maternal-fetal-placental biodistribution of anionic and cationic multimodal poly(glycidyl methacrylate) (PGMA) nanoparticles in pregnant rats at mid (ED10) and late (ED20) gestation was examined. Fluorescently-labelled and superparamagnetic PGMA nanoparticles functionalized with/without poly(ethyleneimine) (PEI) were administered to pregnant rats at a clinically-relevant dose and biodistribution and tissue uptake assessed. Quantitative measurement of fluorescence intensity or magnetic resonance relaxometry in tissue homogenates lacked the sensitivity to quantify tissue uptake. Confocal microscopy, however, identified uptake by maternal organs and the decidua (ectoplacental cone) and trophoblast giant cells of conceptuses at ED10. At ED20, preferential accumulation of cationic vs. anionic nanoparticles was observed in the placenta, with PGMA-PEI nanoparticles localised mainly within the chorionic plate. These findings highlight the significant impact of surface charge and gestational age in the biodistribution of nanoparticles in pregnancy, and demonstrate the importance of using highly sensitive measurement techniques to evaluate nanomaterial biodistribution and maternal-fetal exposure.


Assuntos
Troca Materno-Fetal , Nanopartículas , Polímeros/metabolismo , Animais , Feminino , Idade Gestacional , Metilmetacrilatos/química , Metilmetacrilatos/metabolismo , Microscopia Confocal , Sistema Fagocitário Mononuclear/metabolismo , Imagem Óptica/métodos , Polímeros/química , Gravidez , Ratos , Distribuição Tecidual
6.
Reprod Fertil Dev ; 29(10): 1971-1981, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27997334

RESUMO

Kisspeptin is crucial for the generation of the circadian-gated preovulatory gonadotrophin-releasing hormone (GnRH)-LH surge in female rodents, with expression in the anteroventral periventricular nucleus (AVPV) peaking in the late afternoon of pro-oestrus. Given kisspeptin expression is established before puberty, the aim of the present study was to investigate kisspeptin and clock gene rhythms during the neonatal period. Anterior and posterior hypothalami were collected from C57BL/6J mice on Postnatal Days (P) 5, 15 and 25, at six time points across 24h, for analysis of gene expression by reverse transcription-quantitative polymerase chain reaction. Expression of aryl hydrocarbon receptor nuclear translocator-like gene (Bmal1) and nuclear receptor subfamily 1, group D, member 2 (Rev-erbα) in the anterior hypothalamus (containing the suprachiasmatic nucleus) was not rhythmic at P5 or P15, but Bmal1 expression exhibited rhythmicity in P25 females, whereas Rev-erbα expression was rhythmic in P25 males. KiSS-1 metastasis-suppressor (Kiss1) expression did not exhibit time-of-day variation in the anterior (containing the AVPV) or posterior (containing the arcuate nucleus) hypothalami in female and male mice at P5, P15 or P25. The data indicate that the kisspeptin circadian peak in expression observed in the AVPV of pro-oestrous females does not manifest at P5, P15 or P25, likely due to inadequate oestrogenic stimuli, as well as incomplete development of clock gene rhythmicity before puberty.


Assuntos
Proteínas CLOCK/metabolismo , Ritmo Circadiano/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Animais , Proteínas CLOCK/genética , Feminino , Kisspeptinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Proestro/genética , Proestro/metabolismo , Receptores de Kisspeptina-1/genética , Receptores de Kisspeptina-1/metabolismo , Fatores Sexuais
7.
Endocrinology ; 157(9): 3320-31, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27409644

RESUMO

Maternal adaptations in lipid metabolism are crucial for pregnancy success due to the role of white adipose tissue as an energy store and the dynamic nature of energy needs across gestation. Because lipid metabolism is regulated by the rhythmic expression of clock genes, it was hypothesized that maternal metabolic adaptations involve changes in both adipose clock gene expression and the rhythmic expression of downstream metabolic genes. Maternal core body temperature (Tc) was investigated as a possible mechanism driving pregnancy-induced changes in clock gene expression. Gonadal adipose tissue and plasma were collected from C57BL/6J mice before and on days 6, 10, 14, and 18 of pregnancy (term 19 d) at 4-hour intervals across a 24-hour period. Adipose expression of clock genes and downstream metabolic genes were determined by quantitative RT-PCR, and Tc was measured by intraperitoneal temperature loggers. Adipose clock gene expression showed robust rhythmicity throughout pregnancy, but absolute levels varied substantially across gestation. Rhythmic expression of the metabolic genes Lipe, Pnpla2, and Lpl was clearly evident before pregnancy; however, this rhythmicity was lost with the onset of pregnancy. Tc rhythm was significantly altered by pregnancy, with a 65% decrease in amplitude by term and a 0.61°C decrease in mesor between days 6 and 18. These changes in Tc, however, did not appear to be linked to adipose clock gene expression across pregnancy. Overall, our data show marked adaptations in the adipose clock in pregnancy, with an apparent decoupling of adipose clock and lipolytic/lipogenic gene rhythms from early in gestation.


Assuntos
Tecido Adiposo/metabolismo , Temperatura Corporal , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Ritmo Circadiano , Prenhez/fisiologia , Animais , Feminino , Expressão Gênica , Lipase Lipoproteica/metabolismo , Camundongos Endogâmicos C57BL , PPAR gama/metabolismo , Gravidez , Progesterona/sangue
8.
Biol Reprod ; 95(3): 55, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27465137

RESUMO

Maternal obesity increases the risk of abnormal fetal growth, but the underlying mechanisms remain unclear. Because steroid hormones regulate fetal growth, and both pregnancy and obesity markedly alter circadian biology, we hypothesized that maternal obesity disrupts the normal rhythmic profiles of steroid hormones in rat pregnancy. Obesity was established by cafeteria (CAF) feeding for 8 wk prior to mating and throughout pregnancy. Control (CON) animals had ad libitum access to chow. Daily profiles of plasma corticosterone, 11-dehydrocorticosterone, progesterone, and testosterone were measured at Days 15 and 21 of gestation (term = 23 days) in maternal (both days) and fetal (Day 21) plasma. CAF mothers exhibited increased adiposity relative to CON and showed fetal and placental growth restriction. There was no change, however, in total fetal or placental mass due to slightly larger litter sizes in CAF. Nocturnal declines in progesterone were observed in maternal (39% lower) and fetal (45% lower) plasma in CON animals, but these were absent in CAF animals. CAF mothers were hyperlipidemic at both days of gestation, but this effect was isolated to the dark period at Day 21. CAF maternal testosterone was slightly lower at Day 15 (8%) but increased above CON by Day 21 (16%). Despite elevated maternal testosterone, male fetal testosterone was suppressed by obesity on Day 21. Neither maternal nor fetal glucocorticoid profiles were affected by obesity. In conclusion, obesity disrupts rhythmic profiles of maternal and fetal progesterone, preventing the normal nocturnal decline. Obesity subtly changed testosterone profiles but did not alter maternal and fetal glucocorticoids.


Assuntos
Ritmo Circadiano/fisiologia , Obesidade/sangue , Prenhez/sangue , Progesterona/sangue , Animais , Corticosterona/análogos & derivados , Corticosterona/sangue , Feminino , Gravidez , Ratos , Testosterona/sangue
9.
Am J Physiol Endocrinol Metab ; 311(3): E575-86, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27406739

RESUMO

Adaptations in maternal carbohydrate metabolism are particularly important in pregnancy because glucose is the principal energy substrate used by the fetus. As metabolic homeostasis is intricately linked to the circadian system via the rhythmic expression of clock genes, it is likely that metabolic adaptations during pregnancy also involve shifts in maternal circadian function. We hypothesized that maternal adaptation in pregnancy involves changes in the hepatic expression of clock genes, which drive downstream shifts in circadian expression of glucoregulatory genes. Maternal liver and plasma (n = 6-8/group) were collected across 24-h periods (0800, 1200, 1600, 2000, 0000, 0400) from C57Bl/6J mice under isoflurane-nitrous oxide anesthesia prior to and on days 6, 10, 14 and 18 of pregnancy (term = day 19). Hepatic expression of clock genes and glucoregulatory genes was determined by RT-qPCR. Hepatic clock gene expression was substantially altered across pregnancy, most notably in late gestation when the circadian rhythmicity of several clock genes was attenuated (≤64% reduced amplitude on day 18). These changes were associated with a similar decline in rhythmicity of the key glucoregulatory genes Pck1, G6Pase, and Gk, and by day 18, Pck1 was no longer rhythmic. Overall, our data show marked adaptations in the liver clock during mouse pregnancy, changes that may contribute to the altered circadian variation in glucoregulatory genes near term. We propose that the observed reduction of daily oscillations in glucose metabolism ensure a sustained supply of glucose to meet the high demands of fetal growth.


Assuntos
Relógios Circadianos/genética , Relógios Circadianos/fisiologia , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/biossíntese , Ritmo Circadiano/fisiologia , Glucose/metabolismo , Fígado/metabolismo , Prenhez/fisiologia , Animais , Glicemia/metabolismo , Feminino , Feto/metabolismo , Gluconeogênese/genética , Homeostase , Insulina/sangue , Glicogênio Hepático/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
10.
J Endocrinol ; 229(3): 307-18, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27068699

RESUMO

Kisspeptin, the neuropeptide product of the Kiss1 gene, is critical in driving the hypothalamic-pituitary-gonadal (HPG) axis. Kisspeptin neurons in the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (Arc) of the hypothalamus mediate differential effects, with the Arc regulating negative feedback of sex steroids and the AVPV regulating positive feedback, vital for the preovulatory surge and gated under circadian control. We aimed to characterize hypothalamic Kiss1 and Kiss1r mRNA expression in nonpregnant and pregnant mice, and investigate potential circadian regulation. Anterior and posterior hypothalami were collected from C57BL/6J mice at diestrus, proestrus, and days 6, 10, 14, and 18 of pregnancy, at six time points across 24h, for real-time PCR analysis of gene expression. Analysis confirmed that Kiss1 mRNA expression in the AVPV increased at ZT13 during proestrus, with a luteinizing hormone surge observed thereafter. No diurnal regulation was seen at diestrus or at any stage of pregnancy. Anterior hypothalamic Avp mRNA expression exhibited no diurnal variation, but Avpr1a peaked at 12:00h during proestrus, possibly reflecting the circadian input from the suprachiasmatic nucleus to AVPV Kiss1 neurons. Rfrp (Npvf) expression in the posterior hypothalamus did not demonstrate diurnal variation at any stage. Clock genes Bmal1 and Rev-erbα were strongly diurnal, but there was little change between diestrus/proestrus and pregnancy. Our data indicate the absence of the circadian input to Kiss1 in pregnancy, despite high gestational estradiol levels and normal clock gene expression, and may suggest a disruption of a kisspeptin-specific diurnal rhythm that operates in the nonpregnant state.


Assuntos
Ritmo Circadiano/fisiologia , Hipotálamo/fisiologia , Kisspeptinas/fisiologia , Prenhez/fisiologia , Animais , Arginina Vasopressina/genética , Ritmo Circadiano/genética , Feminino , Expressão Gênica , Hormônios/sangue , Kisspeptinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Prenhez/genética , Proestro/genética , Proestro/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Vasopressinas/genética
11.
Placenta ; 39: 33-40, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26992672

RESUMO

INTRODUCTION: Obesity during pregnancy can cause serious complications for maternal and infant health. While this has often been attributed to increased inflammation during obese pregnancy, human and animal studies exhibit variable results with respect to the inflammatory status of the mother, placenta and fetus. Cafeteria (CAF) feeding induces more inflammation than standard high-fat feeding in non-pregnant animal models. This study investigated whether maternal obesity induced by a CAF diet increases maternal, fetal or placental inflammation. METHODS: Maternal obesity was established in rats by 8 weeks of pre-pregnancy CAF feeding. Maternal plasma inflammatory markers (IL-1ß, IL-6, IL-10, IL-12p40, MCP1, GRO/KC, MIP-2 and TNFα) and expression of inflammatory genes (Tnfα, Il-6, Il-1ß, Tlr2, Tlr4, Cox2 and Emr1) in maternal, placental and fetal tissues were measured at day 21 of gestation. RESULTS: Despite CAF animals having 63% more central body fat than controls at day 21 of gestation, plasma inflammatory markers were not increased; indeed, levels of IL-6, IL-12p40 and MIP2 were reduced slightly. Similarly, inflammatory gene expression remained largely unaffected by CAF feeding, except for slight reductions to Tlr4 and Emr1 expression in CAF maternal adipose tissue, and reduced Tlr4 expression in male labyrinth zone (LZ). The junctional zone (JZ) displayed increased Il-6 expression in CAF animals when fetal sexes were combined, but no inflammatory genes were affected by the CAF diet in fetal liver. CONCLUSIONS: Maternal obesity induced by a CAF diet before and during pregnancy does not increase the inflammatory status of the mother, placenta or fetus in late gestation.


Assuntos
Dieta Hiperlipídica , Inflamação/etiologia , Fenômenos Fisiológicos da Nutrição Materna , Obesidade/imunologia , Complicações na Gravidez/imunologia , Animais , Dieta Hiperlipídica/efeitos adversos , Feminino , Feto/metabolismo , Feto/patologia , Idade Gestacional , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Masculino , Obesidade/etiologia , Obesidade/metabolismo , Placenta/metabolismo , Placenta/patologia , Gravidez , Complicações na Gravidez/etiologia , Complicações na Gravidez/metabolismo , Ratos
12.
J Endocrinol ; 228(3): 135-47, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26883207

RESUMO

Maternal physiological adaptations, such as changes to the hypothalamic-pituitary-adrenal (HPA) axis, are central to pregnancy success. Circadian variation of the HPA axis is dependent on clock gene rhythms in the hypothalamus, but it is not known whether pregnancy-induced changes in maternal glucocorticoid levels are mediated via this central clock. We hypothesized that hypothalamic expression of clock genes changes across mouse pregnancy and this is linked to altered HPA activity. The anterior hypothalamus and maternal plasma were collected from C57Bl/6J mice prior to pregnancy and on days 6, 10, 14 and 18 of gestation (term=d19), across a 24-h period (0800, 1200, 1600, 2000, 0000, 0400 h). Hypothalamic expression of clock genes and Crh was determined by qPCR, plasma ACTH concentration measured by Milliplex assay and plasma corticosterone concentration by LC-MS/MS. Expression of all clock genes varied markedly across gestation, most notably at mid-gestation when levels of each gene were elevated. The pregnancy-induced increase in maternal corticosterone levels (by up to 14-fold on day 14) was not accompanied by a parallel shift in plasma ACTH (28% lower on day 14 compared with non-pregnant levels). Moreover, while circadian rhythmicity in corticosterone was maintained up to day 14 of gestation, this was effectively lost by day 18. Overall, our data show that the central circadian clock undergoes marked adaptations throughout mouse pregnancy, changes that are likely to contribute to maternal physiological adaptations. Importantly, however, neither hypothalamic clock genes nor plasma ACTH levels appear to drive the marked increase in maternal corticosterone after mid-gestation.


Assuntos
Relógios Circadianos/fisiologia , Glucocorticoides/sangue , Adaptação Fisiológica , Glândulas Suprarrenais/fisiologia , Hormônio Adrenocorticotrópico/sangue , Animais , Relógios Circadianos/genética , Corticosterona/análogos & derivados , Corticosterona/sangue , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/fisiologia , Feminino , Expressão Gênica , Idade Gestacional , Hipotálamo/química , Hipotálamo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Hipófise/fisiologia , Gravidez , RNA Mensageiro/análise
13.
Reproduction ; 149(2): 171-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25504868

RESUMO

The aim of this study was to determine whether supplementation with fish oil-derived n-3 polyunsaturated fatty acids (n-3 PUFA) during pregnancy modifies placental PUFA composition, the accumulation of specialised pro-resolving lipid mediators (SPMs, specifically resolvins (Rv), protectins (PD) and upstream precursors) and inflammatory gene expression. Placentas were collected from women (n=51) enrolled in a randomised, placebo controlled trial of n-3 PUFA supplementation from 20-week gestation. Lipids were extracted for fatty acid analysis and SPMs were quantitated by mass spectrometry. Gene expression was determined by qRT-PCR. Using multiple regression analysis, data were correlated for placental n-3 PUFA and SPM levels with PUFA levels in maternal and cord blood erythrocytes. Supplementation with n-3 PUFAs increased placental docosahexaenoic acid (DHA) levels, but not eicosapentaenoic acid (EPA) levels (P<0.05), and increased the levels of the SPM precursors 18-hydroxyeicosapentaenoic acid and 17-hydroxydocosahexaenoic acid (17-HDHA) by two- to threefold (P<0.0005). RvD1, 17R-RvD1, RvD2 and PD1 were detectable in all placentas, but concentrations were not significantly increased by n-3 PUFA supplementation. Placental DHA levels were positively associated with maternal and cord DHA levels (P<0.005), and with placental 17-HDHA concentrations (P<0.0001). Placental mRNA expression of PTGS2, IL1ß, IL6 and IL10 was unaffected by n-3 PUFA supplementation, but TNFα expression was increased by 14-fold (P<0.05). We conclude that n-3 PUFA supplementation in pregnancy i) enhances placental accumulation of DHA and SPM precursors, ii) does not alter placental EPA levels, and iii) has no stimulatory effects on inflammatory gene expression. Further studies are required to ascertain the biological significance of SPMs in the placenta and the potential immunomodulatory effects of elevating placental SPM levels.


Assuntos
Citocinas/análise , Ácidos Graxos Ômega-3/administração & dosagem , Metabolismo dos Lipídeos/efeitos dos fármacos , Placenta/química , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/análise , Ácido Eicosapentaenoico/análise , Feminino , Expressão Gênica/efeitos dos fármacos , Idade Gestacional , Humanos , Inflamação/genética , Placebos , Placenta/efeitos dos fármacos , Gravidez
14.
Reprod Biol Endocrinol ; 12: 39, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24886466

RESUMO

BACKGROUND: Adverse fetal environments predispose offspring to pathologies associated with the metabolic syndrome. Previously we demonstrated that adult offspring of dexamethasone-treated mothers had elevated plasma insulin and pro-inflammatory cytokines, effects prevented by a postnatal diet enriched with omega (n)-3 fatty acids. Here we tested whether prenatal glucocorticoid excess also programmed the adipose tissue phenotype, and whether this outcome is rescued by dietary n-3 fatty acids. METHODS: Offspring of control and dexamethasone-treated mothers (0.75 µg/ml in drinking water, day 13 to term) were cross-fostered to mothers on a standard (Std) or high n-3 (Hn3) diet at birth. Offspring remained on these diets post-weaning, and serum and retroperitoneal fat were obtained at 6 months of age (n = 5-8 per group). Serum was analysed for blood lipids and fatty acid profiles, adipocyte cross sectional area was measured by unbiased stereological analysis and adipose expression of markers of inflammation, glucocorticoid sensitivity and lipid metabolism were determined by RT-qPCR analysis. RESULTS: Serum total fatty acid levels were elevated (P < 0.01) in male offspring of dexamethasone-treated mothers, an effect prevented by Hn3 consumption. Prenatal dexamethasone also programmed increased adipose expression of Il6, Il1b (both P < 0.05) and Tnfa (P < 0.001) mRNAs regardless of fetal sex, but again this effect was prevented (for Il6 and Il1b) by Hn3 consumption. Offspring of dexamethasone-treated mothers had increased adipose expression of Gr (P = 0.008) and Ppara (P < 0.05) regardless of sex or postnatal diet, while 11bHsd1 was upregulated in males only. The Hn3 diet increased Ppard expression and reduced adipocyte size in all offspring (both P < 0.05) irrespective of prenatal treatment. CONCLUSIONS: Prenatal glucocorticoid exposure programmed increased expression of inflammatory markers and enhanced glucocorticoid sensitivity of adipose tissue. Partial prevention of this phenotype by high n-3 consumption indicates that postnatal dietary manipulations can limit adverse fetal programming effects on adipose tissue.


Assuntos
Suplementos Nutricionais , Modelos Animais de Doenças , Ácidos Graxos Ômega-3/uso terapêutico , Gordura Intra-Abdominal/imunologia , Síndrome Metabólica/prevenção & controle , Complicações na Gravidez/fisiopatologia , Estresse Psicológico/fisiopatologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Animais , Dexametasona/análogos & derivados , Ácidos Graxos Ômega-3/sangue , Ácidos Graxos Ômega-3/metabolismo , Feminino , Desenvolvimento Fetal , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Lactação , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Síndrome Metabólica/imunologia , Síndrome Metabólica/metabolismo , Síndrome Metabólica/patologia , Gravidez , Distribuição Aleatória , Ratos Wistar , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Regulação para Cima , Desmame
15.
Reproduction ; 147(5): R143-52, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24451224

RESUMO

The developing fetus requires substantial amounts of fatty acids to support rapid cellular growth and activity. Although the fatty acid composition delivered to the fetus is largely determined by maternal circulating levels, the placenta preferentially transfers physiologically important long-chain polyunsaturated fatty acids (LC-PUFAs), particularly omega-3 (n-3) PUFAs. Maternal dietary supplementation with n-3 PUFAs during pregnancy has been shown to increase gestation length, enhance fetal growth, and reduce the risk of pregnancy complications, although the precise mechanisms governing these effects remain uncertain. Omega-3 PUFAs are involved in several physiological pathways which could account for these effects, including anti-inflammatory, pro-resolving, and anti-oxidative pathways. Recent studies have shown that maternal dietary n-3 PUFA supplementation during rat pregnancy can reduce placental oxidative damage and increase placental levels of pro-resolving mediators, effects associated with enhanced fetal and placental growth. Because several placental disorders, such as intrauterine growth restriction, preeclampsia, and gestational diabetes mellitus, are associated with heightened placental inflammation and oxidative stress, there is considerable interest in the potential for dietary n-3 PUFAs as a therapeutic intervention for these disorders. In this study, we review the impact of dietary n-3 PUFAs on placental function, with particular focus on placental inflammation, inflammatory resolution, and oxidative stress.


Assuntos
Ácidos Graxos Ômega-3/fisiologia , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Placenta/fisiologia , Animais , Diabetes Gestacional/prevenção & controle , Suplementos Nutricionais , Ácidos Graxos Ômega-3/administração & dosagem , Ácidos Graxos Ômega-3/uso terapêutico , Feminino , Retardo do Crescimento Fetal/prevenção & controle , Humanos , Pré-Eclâmpsia/prevenção & controle , Gravidez
16.
Reproduction ; 146(6): 539-47, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24023246

RESUMO

Placental oxidative stress plays a key role in the pathophysiology of several placenta-related disorders. Oxidative stress occurs when excess reactive oxygen species (ROS) damages cellular components, an outcome limited by antioxidant enzymes; mitochondrial uncoupling protein 2 (UCP2) also limits ROS production. We recently reported that maternal dietary omega-3 polyunsaturated fatty acid (n-3 PUFA) supplementation reduced placental oxidative damage and enhanced fetal and placental growth in the rats. Here, we examined the effect of n-3 PUFAs on placental antioxidant defences and whether n-3 PUFA supplementation could prevent growth restriction induced by placental ischaemia-reperfusion (IR), a known inducer of oxidative stress. Rats were fed either standard or high-n-3 PUFA diets from day 1 of pregnancy. Placentas were collected on days 17 and 22 in untreated pregnancies (term=day 23) and at day 22 following IR treatment on day 17. Expression of several antioxidant enzyme genes (Sod1, Sod2, Sod3, Cat, Txn1 and Gpx3) and Ucp2 was measured by quantitative RT-PCR in the placental labyrinth zone (LZ) and junctional zone (JZ). Cytosolic superoxide dismutase (SOD), mitochondrial SOD and catalase (CAT) activities were also analyzed. Maternal n-3 PUFA supplementation increased LZ mRNA expression of Cat at both gestational days (2- and 1.5-fold respectively; P<0.01) and female Sod2 at day 22 (1.4-fold, P<0.01). Cytosolic SOD activity increased with n-3 PUFA supplementation at day 22 (1.3-fold, P<0.05). Sod1 and Txn1 expression decreased marginally (30 and 22%, P<0.05). JZ antioxidant defences were largely unaffected by diet. Despite increased LZ antioxidant defences, maternal n-3 PUFA supplementation did not protect against placental IR-induced growth restriction of the fetus and placental LZ.


Assuntos
Antioxidantes/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Fenômenos Fisiológicos da Nutrição Materna , Placenta/efeitos dos fármacos , Animais , Ingestão de Alimentos/fisiologia , Feminino , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/prevenção & controle , Expressão Gênica/efeitos dos fármacos , Masculino , Placenta/metabolismo , Circulação Placentária/efeitos dos fármacos , Gravidez , Ratos , Ratos Wistar , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo
17.
Endocrinology ; 154(9): 3110-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23782939

RESUMO

Fetal glucocorticoid excess programs several adverse outcomes in adult offspring, many of which can be prevented by postnatal, dietary omega-3 (n-3) fatty acids. Here we tested 2 separate hypotheses: 1) a postnatal high-fat diet exacerbates the glucocorticoid-programmed phenotype; and 2) postnatal, dietary n-3 fatty acids rescue programmed outcomes, even in the presence of a high-fat diet challenge. Pregnant Wistar rat dams were either untreated or administered dexamethasone acetate (Dex; 0.5 µg/mL drinking water) from day 13 of pregnancy. Offspring were cross-fostered to untreated mothers and males were weaned onto a standard (Std), high-fat, low n-3 (HF), or high-fat, high n-3 (HFHn-3) diet. Prenatal Dex reduced birth weight (26%) and delayed puberty onset by 1.2 days, irrespective of postnatal diet. Prenatal Dex programmed increased blood pressure in adult offspring, an effect worsened by the postnatal HF diet. Supplementation with high n-3 fatty acids, however, prevented both the Dex and HF-induced increases in blood pressure. Prenatal Dex also programmed increased adiposity, plasma cholesterol, and plasma triglyceride levels at 6 months of age, particularly in those offspring raised on the HF diet. But again, each of these adverse outcomes was rescued by supplementation of the HF diet with n-3 fatty acids. In conclusion, the capacity of n-3 fatty acids to overcome adverse programming outcomes remains evident, even in the presence of a HF diet challenge.


Assuntos
Adiposidade , Suplementos Nutricionais , Ácidos Graxos Ômega-3/uso terapêutico , Desenvolvimento Fetal , Glucocorticoides/metabolismo , Hiperlipidemias/prevenção & controle , Hipertensão/prevenção & controle , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/uso terapêutico , Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais/efeitos adversos , Modelos Animais de Doenças , Ácidos Graxos Ômega-3/efeitos adversos , Feminino , Glucocorticoides/sangue , Hiperlipidemias/etiologia , Hiperlipidemias/imunologia , Hipertensão/etiologia , Hipertensão/imunologia , Masculino , Troca Materno-Fetal , Gravidez , Complicações na Gravidez/sangue , Complicações na Gravidez/imunologia , Complicações na Gravidez/fisiopatologia , Distribuição Aleatória , Ratos , Ratos Wistar , Estresse Fisiológico , Estresse Psicológico/sangue , Estresse Psicológico/imunologia , Estresse Psicológico/fisiopatologia
18.
J Lipid Res ; 54(8): 2247-2254, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23723388

RESUMO

Placental inflammation is associated with several pregnancy disorders. Inflammation is limited by anti-inflammatory and proresolving mechanisms, the latter partly mediated by resolvins and protectins derived from omega-3 polyunsaturated fatty acids (n-3PUFA). We examined effects of dietary n-3PUFAs on levels of resolvins, protectins, and lipoxygenase (ALOX) enzymes in the rat placenta. Rats consumed standard (Std) or high n-3PUFA (Hn3) diets from day 1 of pregnancy; tissues were collected on day 17 or 22 (term = day 23). Maternal Hn3 diet increased resolvin and protectin precursors, 18R/S-HEPE (P < 0.001), and 17R/S-HDHA (P < 0.01) at both days. Resolvins (17R-RvD1 and RvD1) increased at day 22 (P < 0.001) after Hn3 consumption, coincident with higher Alox15b and Alox5 mRNA expression, while RvD2 increased at both days (P < 0.05). Protectins, PD1, and 10S,17S-DiHDHA increased over late gestation (P < 0.001), coincident with higher Alox15 mRNA expression (P < 0.001) and further increased with Hn3 diet (P < 0.05). Maternal systemic and placental proinflammatory mediators were not suppressed by Hn3 diet; systemic IL1ß, placental Il1ß, and Il6 mRNA expression increased marginally with Hn3 at day 22 (P < 0.001), while Ptgs1 (Cox1) expression increased both days (P < 0.05). Our data indicate that maternal n-3PUFA supplementation enhances expression of enzymes in the n-3PUFA metabolic pathway and increases placental levels of resolvins and protectins.


Assuntos
Antígenos CD59/análise , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/análise , Ácidos Graxos Ômega-3/administração & dosagem , Lipoxigenase/análise , Placenta/química , Animais , Feminino , Lipoxigenase/genética , Lipoxigenase/metabolismo , Placenta/irrigação sanguínea , Placenta/enzimologia , Gravidez , RNA Mensageiro/análise , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
19.
Biol Reprod ; 88(2): 37, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23269667

RESUMO

Placental oxidative stress plays a key role in the pathophysiology of several placenta-related disorders including intrauterine growth restriction. Oxidative stress occurs when accumulation of reactive oxygen species damages DNA, proteins, and lipids, an outcome normally limited by antioxidant defenses. Dietary supplementation with omega-3 polyunsaturated fatty acids (n-3 PUFAs) may limit oxidative stress by increasing antioxidant capacity, but n-3 PUFAs are also highly susceptible to lipid peroxidation; so n-3 PUFA supplementation is potentially harmful. Here we examined the effect of n-3 PUFAs on placental oxidative stress and on placental and fetal growth in the rat. We also investigated whether diet-induced changes in maternal plasma fatty acid profiles are associated with comparable changes in placental and fetal tissues. Rats were fed either standard or high n-3 PUFA diets from Day 1 of pregnancy, and tissues were collected on Day 17 or 22 (term = Day 23). Dietary supplementation with n-3 PUFAs increased fetal (6%) and placental (12%) weights at Day 22, the latter attributable primarily to growth of the labyrinth zone (LZ). Increased LZ weight was accompanied by reduced LZ F(2)-isoprostanes (by 31% and 11% at Days 17 and 22, respectively), a marker of oxidative damage. Maternal plasma PUFA profiles were altered by dietary fatty acid intake and were strongly predictive of corresponding profiles in placental and fetal tissues. Our data indicate that n-3 PUFA supplementation reduces placental oxidative stress and enhances placental and fetal growth. Moreover, fatty acid profiles in the mother, placenta, and fetus are highly dependent on dietary fatty acid intake.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Desenvolvimento Fetal/efeitos dos fármacos , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Placentação , Prenhez/fisiologia , Animais , Suplementos Nutricionais , Ácidos Graxos/sangue , Ácidos Graxos Ômega-3/administração & dosagem , Feminino , Desenvolvimento Fetal/fisiologia , Peroxidação de Lipídeos/fisiologia , Modelos Animais , Estresse Oxidativo/fisiologia , Placenta/fisiologia , Gravidez , Resultado da Gravidez , Ratos , Ratos Wistar
20.
J Anat ; 220(4): 350-62, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22414226

RESUMO

Glucocorticoids have been implicated in male reproductive function and 11ß-HSD-1 and -2, the glucocorticoid receptor (GR) and mineralocorticoid receptor (MR), all of which are known to modulate glucocorticoid action, have been localised in the adult rat epididymis, but their developmental expression has not been investigated. Na(+)K(+)-ATPase activity, responsible for sodium transport, is induced by both mineralocorticoids and glucocorticoids in the kidney and colon, and has been localised in epididymal epithelium. This study examined the immunolocalisation of 11ß-HSD-1 and -2, GR, MR and Na(+)K(+)-ATPase in rat epididymal epithelium (n = 5) at postnatal days (pnd) 1, 7, 15, 28, 40, 60, 75 and 104, and relative mRNA expression of 11ß-HSD-1 and -2, and GR at pre-puberty (pnd 28) and post-puberty (pnd 75). 11ß-HSD-1, GR and MR were localised in the epididymal epithelium from pnd 1, and 11ß-HSD-2 and Na(+)K(+)-ATPase reactivity from pnd 15. At pnd 28 there was maximal immunoreactivity for both the GR and MR and 11ß-HSD-1 and -2. 11ß-HSD-1 mRNA expression in the caput increased from pre- to post-puberty, whereas 11ß-HSD-2 mRNA expression fell over the same period (P < 0.01). GR mRNA expression was similar at pre- and post-puberty in both caput and cauda. Developmental changes in expression of 11ß-HSD-1 and -2 suggest that overall exposure of the epididymis to glucocorticoids increases post-puberty, but cell-specific expression of the 11ß-HSD enzymes still provides a capacity for intricate local control of glucocorticoid exposure.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Epididimo/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Análise de Variância , Animais , Epididimo/crescimento & desenvolvimento , Imuno-Histoquímica , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA