Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
Mais filtros

Medicinas Complementares
Tipo de documento
Intervalo de ano de publicação
1.
Acta Cir Bras ; 39: e391524, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38629649

RESUMO

PURPOSE: Pre-eclampsia (PE) is a pregnancy-related complication. Eucommia is effective in the treatment of hypertensive disorders in pregnancy, but the specific effects and possible mechanisms of Eucommia granules (EG) in PE remain unknown. The aim of this study was to investigate the effects and possible mechanisms of EG in PE rats. METHODS: Pregnant Sprague Dawley rats were divided into five groups (n = 6): the control group, the model group, the low-dose group, the medium-dose group, and the high-dose group of EG. The PE model was established by subcutaneous injection of levonitroarginine methyl ester. Saline was given to the blank and model groups, and the Eucommia granules were given by gavage to the remaining groups. Blood pressure and urinary protein were detected. The body length and weight of the pups and the weight of the placenta were recorded. Superoxide dismutase (SOD) activity and levels of malondialdehyde (MDA), placental growth factor (PIGF), and soluble vascular endothelial growth factor receptor-1 (sFIt-1) were measured in the placenta. Pathological changes were observed by hematoxylin-eosin staining. Wnt/ß-catenin pathway-related protein expression was detected using Western blot. RESULTS: Compared with the model group, the PE rats treated with EG had lower blood pressure and urinary protein. The length and weight of the pups and placental weight were increased. Inflammation and necrosis in the placental tissue was improved. SOD level increased, MDA content and sFIt-1/PIGF ratio decreased, and Wnt/ß-catenin pathway-related protein expression level increased. Moreover, the results of EG on PE rats increased with higher doses of EG. CONCLUSIONS: EG may activate the Wnt/ß-catenin pathway and inhibit oxidative stress, inflammation, and vascular endothelial injury in PE rats, thereby improving the perinatal prognosis of preeclamptic rats. EG may inhibit oxidative stress, inflammation, and vascular endothelial injury through activation of the Wnt/ß-catenin pathway in preeclampsia rats, thereby improving perinatal outcomes in PE rats.


Assuntos
Pré-Eclâmpsia , Complicações na Gravidez , Humanos , Ratos , Feminino , Gravidez , Animais , Pré-Eclâmpsia/tratamento farmacológico , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/patologia , Placenta , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo , Fator de Crescimento Placentário/metabolismo , Fator de Crescimento Placentário/farmacologia , Fator de Crescimento Placentário/uso terapêutico , Estresse Oxidativo , Complicações na Gravidez/metabolismo , Inflamação/patologia , Superóxido Dismutase/metabolismo
2.
Nutrients ; 15(23)2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-38068740

RESUMO

Though firstly identified in cerebral folate deficiency, autoantibodies against folate receptors (FRAbs) have been implicated in pregnancy complications such as miscarriage; however, the underlying mechanism needs to be further elaborated. FRAbs can be produced via sensitization mediated by folate-binding protein as well as gene mutation, aberrant modulation, or degradation of folate receptors (FRs). FRAbs may interfere with folate internalization and metabolism through blocking or binding with FRs. Interestingly, different types of FRs are expressed on trophoblast cells, decidual epithelium or stroma, and macrophages at the maternal-fetal interface, implying FRAbs may be involved in the critical events necessary for a successful pregnancy. Thus, we propose that FRAbs may disturb pregnancy establishment and maintenance by modulating trophoblastic biofunctions, placental development, decidualization, and decidua homeostasis as well as the functions of FOLR2+ macrophages. In light of these findings, FRAbs may be a critical factor in pathological pregnancy, and deserve careful consideration in therapies involving folic acid supplementation for pregnancy complications.


Assuntos
Aborto Espontâneo , Receptor 2 de Folato , Complicações na Gravidez , Gravidez , Feminino , Humanos , Placenta/metabolismo , Autoanticorpos , Ácido Fólico/metabolismo , Complicações na Gravidez/metabolismo , Decídua/metabolismo , Receptor 2 de Folato/metabolismo
3.
Mol Cell Biochem ; 476(11): 4003-4018, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34196872

RESUMO

Pregnancy is a challenging physiological process that involves maternal adaptations to the increasing energetics demands imposed by the growing conceptus. Failure to adapt to these requirements may result in serious health complications for the mother and the baby. The mitochondria are biosynthetic and energy-producing organelles supporting the augmented energetic demands of pregnancy. Evidence suggests that placental mitochondria display a dynamic phenotype through gestation. At early stages of pregnancy placental mitochondria are mainly responsible for the generation of metabolic intermediates and reactive oxygen species (ROS), while at later stages of gestation, the placental mitochondria exhibit high rates of oxygen consumption. This review describes the metabolic fingerprint of the placental mitochondria at different stages of pregnancy and summarises key signs of mitochondrial dysfunction in pathological pregnancy conditions, including preeclampsia, gestational diabetes and intrauterine growth restriction (IUGR). So far, the effects of placental-driven metabolic changes governing the metabolic adaptations occurring in different maternal tissues in both, healthy and pathological pregnancies, remain to be uncovered. Understanding the function and molecular aspects of the adaptations occurring in placental and maternal tissue's mitochondria will unveil potential targets for further therapeutic exploration that could address pregnancy-related disorders. Targeting mitochondrial metabolism is an emerging approach for regulating mitochondrial bioenergetics. This review will also describe the potential therapeutic use of compounds with a recognised effect on mitochondria, for the management of preeclampsia.


Assuntos
Diabetes Gestacional/metabolismo , Mitocôndrias/metabolismo , Pré-Eclâmpsia/metabolismo , Complicações na Gravidez/metabolismo , Animais , Diabetes Gestacional/patologia , Metabolismo Energético , Feminino , Homeostase , Humanos , Oxirredução , Pré-Eclâmpsia/patologia , Gravidez , Complicações na Gravidez/patologia
4.
Mol Reprod Dev ; 88(6): 437-458, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34008284

RESUMO

Genomic imprinting is important for mammalian development and its dysregulation can cause various developmental defects and diseases. The study evaluated the effects of different dietary combinations of folic acid and B12 on epigenetic regulation of IGF2R and KCNQ1OT1 ncRNA in C57BL/6 mice model. Female mice were fed diets with nine combinations of folic acid and B12 for 4 weeks. They were mated and off-springs born (F1) were continued on the same diet for 6 weeks postweaning and were allowed to mate. The placenta and fetal (F2) tissues were collected at day 20 of gestation. Dietary deficiency of folate (BNFD and BOFD) and B12 (BDFN) with either state of other vitamin or combined deficiency of both vitamins (BDFD) in comparison to BNFN, were overall responsible for reduced expression of IGF2R in the placenta (F1) and the fetal liver (F2) whereas a combination of folate deficiency with different levels of B12 revealed sex-specific differences in kidney and brain. The alterations in the expression of IGF2R caused by folate-deficient conditions (BNFD and BOFD) and both deficient condition (BDFD) was found to be associated with an increase in suppressive histone modifications. Over-supplementation of either folate or B12 or both vitamins in comparison to BNFN, led to increase in expression of IGF2R and KCNQ1OT1 in the placenta and fetal tissues. The increase in the expression of IGF2R caused by folate over-supplementation (BNFO) was associated with decreased DNA methylation in fetal tissues. KCNQ1OT1 noncoding RNA (ncRNA), however, showed upregulation under deficient conditions of folate and B12 only in female fetal tissues which correlated well with hypomethylation observed under these conditions. An epigenetic reprograming of IGF2R and KCNQ1OT1 ncRNA in the offspring was evident upon different dietary combinations of folic acid and B12 in the mice.


Assuntos
Dieta , Epigênese Genética/efeitos dos fármacos , Feto/efeitos dos fármacos , Ácido Fólico/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Placenta/efeitos dos fármacos , RNA Longo não Codificante/genética , Receptor IGF Tipo 2/genética , Vitamina B 12/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Encéfalo/embriologia , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Feminino , Feto/metabolismo , Ácido Fólico/administração & dosagem , Ácido Fólico/sangue , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/metabolismo , Impressão Genômica , Homocisteína/sangue , Rim/embriologia , Rim/metabolismo , Fígado/embriologia , Fígado/metabolismo , Masculino , Camundongos , Placenta/metabolismo , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/metabolismo , RNA Longo não Codificante/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor IGF Tipo 2/metabolismo , Vitamina B 12/administração & dosagem , Vitamina B 12/sangue , Deficiência de Vitamina B 12/genética , Deficiência de Vitamina B 12/metabolismo
5.
FASEB J ; 35(5): e21477, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33891326

RESUMO

Chronic fetal hypoxia is one of the most common outcomes in complicated pregnancy in humans. Despite this, its effects on the long-term health of the brain in offspring are largely unknown. Here, we investigated in rats whether hypoxic pregnancy affects brain structure and function in the adult offspring and explored underlying mechanisms with maternal antioxidant intervention. Pregnant rats were randomly chosen for normoxic or hypoxic (13% oxygen) pregnancy with or without maternal supplementation with vitamin C in their drinking water. In one cohort, the placenta and fetal tissues were collected at the end of gestation. In another, dams were allowed to deliver naturally, and offspring were reared under normoxic conditions until 4 months of age (young adult). Between 3.5 and 4 months, the behavior, cognition and brains of the adult offspring were studied. We demonstrated that prenatal hypoxia reduced neuronal number, as well as vascular and synaptic density, in the hippocampus, significantly impairing memory function in the adult offspring. These adverse effects of prenatal hypoxia were independent of the hypoxic pregnancy inducing fetal growth restriction or elevations in maternal or fetal plasma glucocorticoid levels. Maternal vitamin C supplementation during hypoxic pregnancy protected against oxidative stress in the placenta and prevented the adverse effects of prenatal hypoxia on hippocampal atrophy and memory loss in the adult offspring. Therefore, these data provide a link between prenatal hypoxia, placental oxidative stress, and offspring brain health in later life, providing insight into mechanism and identifying a therapeutic strategy.


Assuntos
Ácido Ascórbico/uso terapêutico , Atrofia/tratamento farmacológico , Hipóxia Fetal/complicações , Hipocampo/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Efeitos Tardios da Exposição Pré-Natal/tratamento farmacológico , Animais , Animais Recém-Nascidos , Antioxidantes/uso terapêutico , Atrofia/etiologia , Atrofia/metabolismo , Atrofia/patologia , Suplementos Nutricionais , Modelos Animais de Doenças , Feminino , Retardo do Crescimento Fetal/tratamento farmacológico , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Gravidez , Complicações na Gravidez/tratamento farmacológico , Complicações na Gravidez/etiologia , Complicações na Gravidez/metabolismo , Complicações na Gravidez/patologia , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Ratos , Ratos Wistar
6.
J Endocrinol Invest ; 44(10): 2307-2314, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33704696

RESUMO

BACKGROUND: Although Iran has been considered iodine replete since 2000, the first national survey of iodine intake among Iranian pregnant women in 2014 indicated that despite the adequate intake of iodine by the general population, this vulnerable group has moderate iodine deficiency. Therefore, in this national cross-sectional interventional study, we aimed to assess the iodine intake and thyroid function of Iranian pregnant women 2 years after implementing national iodine supplementation for this vulnerable group. MATERIALS AND METHODS: In this cross-sectional study, we conducted a national interventional survey of pregnant women. A total of 1200 pregnant women (400 women from each trimester) from 12 provinces of Iran were recruited from the antenatal care clinics from October 2018 to March 2019. The median urinary iodine concentration (MUIC), as an indicator of iodine status in three spot urine samples, was measured, along with the serum total T4 (TT4), thyrotropin (TSH), thyroglobulin (Tg), thyroid peroxidase antibody (TPO-Ab), and iodine content of household salt. RESULTS: The mean age of the cohort was 28 ± 6.2 years, with the mean gestational age of 22.7 ± 13.0 weeks. The overall MUIC (IQR) of pregnant women was 188 µg/L (124.2-263 µg/L). Also, the MUICs in the three trimesters of pregnancy were 174 µg/L (110-254), 175 µg/L (116-251), and 165 µg/L (114-235), respectively. The MUICs ≥ 150, 100-149, and < 100 µg/L were found in 63, 19.8, and 16.2% of the subjects, respectively. The mean TT4 level was 12 ± 4.5 µg/dL, and the median (IQR) level of TSH was 2.37 mIU/L (1.66-3.18 mIU/L). According to our local reference range, 118 (10.5%) pregnant women had subclinical hypothyroidism, 6 (0.53%) women had isolated hypothyroxinemia, and 65 (5.7%) women were TPO-Ab positive. Also, the median (IQR) level of Tg was 10.08 µg/dL (5.7-20.4 µg/dL), and the median iodine content of household salt was 29.6 µg/g; the iodine content was ≥ 30 µg/g in 85% of household salt. The results showed that more than 95% of households were under iodized salt coverage. CONCLUSION: The results of this study indicated that iodine supplementation with at least 150 µg of iodine per day improved the iodine intake of pregnant women. Except for subclinical hypothyroidism, the prevalence of clinical hypothyroidism, clinical/subclinical thyrotoxicosis, TPO-Ab positivity, and isolated hypothyroxinemia decreased significantly, which emphasizes the importance of iodine supplementation during pregnancy.


Assuntos
Biomarcadores/sangue , Suplementos Nutricionais , Hipotireoidismo/prevenção & controle , Iodo/administração & dosagem , Iodo/urina , Complicações na Gravidez/prevenção & controle , Gestantes , Cloreto de Sódio na Dieta/administração & dosagem , Cloreto de Sódio na Dieta/urina , Adulto , Autoanticorpos/sangue , Estudos Transversais , Feminino , Seguimentos , Humanos , Hipotireoidismo/epidemiologia , Hipotireoidismo/metabolismo , Irã (Geográfico)/epidemiologia , Gravidez , Complicações na Gravidez/epidemiologia , Complicações na Gravidez/metabolismo , Prevalência , Prognóstico , Tireoglobulina/sangue , Testes de Função Tireóidea , Tireotropina/sangue
7.
Blood Cells Mol Dis ; 88: 102536, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33450539

RESUMO

In a two-part process, we assessed elements of the principal hormonal pathway regulating iron homeostasis in human neonates. Part 1: Quantifying erythropoietin (Epo), erythroferrone (ERFE), hepcidin, and relevant serum and erythrocytic iron-related metrics in umbilical cord blood from term (n = 13) and preterm (n = 10) neonates, and from neonates born to mothers with diabetes and obesity (n = 13); Part 2: Quantifying serum Epo, ERFE, and hepcidin before and following darbepoetin administration. Part 1: We measured Epo, ERFE and hepcidin in all cord blood samples. Epo and ERFE levels did not differ between the three groups. Preterm neonates had the lowest hepcidin levels, while neonates born to diabetic women with a very high BMI had the lowest ferritin and RET-He levels. Part 2: Following darbepoetin dosing, ERFE levels generally increased (p < 0.05) and hepcidin levels generally fell (p < 0.05). Our observations suggest that the Epo/ERFE/hepcidin axis is intact in the newborn period.


Assuntos
Eritropoetina/sangue , Hepcidinas/sangue , Hormônios Peptídicos/sangue , Transdução de Sinais , Eritropoetina/metabolismo , Feminino , Sangue Fetal/metabolismo , Hepcidinas/metabolismo , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Obesidade/sangue , Obesidade/metabolismo , Hormônios Peptídicos/metabolismo , Gravidez , Complicações na Gravidez/sangue , Complicações na Gravidez/metabolismo , Gravidez em Diabéticas/sangue , Gravidez em Diabéticas/metabolismo , Nascimento Prematuro/sangue , Nascimento Prematuro/metabolismo
8.
Front Endocrinol (Lausanne) ; 11: 523319, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33362709

RESUMO

Introduction: An Israeli national survey found that 85% of pregnant women had urinary iodine content (UIC) levels below the adequacy range (<150 µg/L). Widespread desalinated water usage and no national fortification plan are possible causes. Studies assessing relationships between iodine status and maternal and neonatal thyroid function provided varying results. Our aims were to determine whether iodine deficiency was associated with altered maternal or neonatal thyroid function and the factors leading to iodine deficiency. Methods: A cross-sectional study including 100 healthy women without prior thyroid disease, in their first trimester of a singleton pregnancy were recruited from an HMO clinic in central Israel. The women were followed from their first trimester. All women completed a 24-h dietary recall and life habits questionnaires. We tested for UIC, maternal and neonatal thyroid function, maternal autoantibodies, and neonatal outcomes. Results: Median UIC in our cohort was 49 µg/L [25%-75% interquartile range (IQR) 16-91.5 µg/L], with 84% below adequacy range. No correlation was found between iodine deficiency and maternal or neonatal thyroid function which remained within normal ranges. Antibody status did not differ, but thyroglobulin levels were significantly higher in iodine insufficient subjects. UIC was higher in women consuming an iodine containing supplement. There was no association between UIC and dietary iodine content or water source. Conclusions: Moderate iodine deficiency is common in our healthy pregnant women population. Our data imply that moderate iodine deficiency in pregnancy seem sufficient to maintain normal maternal and neonatal thyroid function.


Assuntos
Iodo/deficiência , Complicações na Gravidez/metabolismo , Tireoglobulina/sangue , Tireotropina/sangue , Tiroxina/sangue , Estudos Transversais , Feminino , Humanos , Iodo/urina , Estado Nutricional , Gravidez , Complicações na Gravidez/sangue , Complicações na Gravidez/urina , Testes de Função Tireóidea
9.
BMC Pregnancy Childbirth ; 20(1): 619, 2020 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-33054794

RESUMO

BACKGROUND: Vitamin D deficiency during pregnancy is common and is likely to be associated with metabolic complications in the mother. The aim of this study was to assess the efficacy of two doses of vitamin D supplementation during pregnancy on maternal and cord blood vitamin D status and metabolic and oxidative stress biomarkers. METHODS: The eligible pregnant women (n = 84) invited to participate in the study and randomly allocated to one of the two supplementation groups (1000 IU/d vitamin D and 2000 IU/d). Biochemical assessments of mothers including serum concentrations of 25(OH)D, calcium, phosphate, iPTH, fasting serum sugar (FBS), insulin, triglyceride, total cholesterol, LDL-C, HDL-C, malondialdehyde (MDA) and total antioxidant capacity (TAC) were done at the beginning and 34 weeks of gestation. Cord blood serum concentrations of 25(OH)D, iPTH, MDA and TAC were assessed at delivery as well. To determine the effects of vitamin D supplementation on metabolic markers 1-factor repeated-measures analysis of variance (ANOVA) was used. Between groups comparisons was done by using Independent-samples Student's t-test or Mann-Whitney test. P < 0.05 was considered as significant. RESULTS: Supplementation with 1000 IU/d and 2000 IU/d vitamin D resulted in significant changes in vitamin D status over pregnancy (24.01 ± 21.7, P < 0.001 in 1000 IU/d group and 46.7 ± 30.6 nmol/L, P < 0.001 in 2000 IU/d group). Daily intake of 2000 compared with 1000 IU/d tended to increase the serum concentration of HDL-C (10 ± 8.37, P < 0.001 in 1000 IU/d group and 9.52 ± 11.39 mg/dL, P < 0.001 in 2000 IU/d group). A significant decrement in serum concentration of iPTH observed in both groups (- 4.18 ± 7.5, P = 0.002 in 1000 IU/d group and - 8.36 ± 14.17, P = 0.002 in 2000 IU/d group). CONCLUSIONS: Supplementation with 2000 IU/d vitamin D as compared with 1000 IU/d, is more effective in promoting vitamin D status and HDL-C serum concentration and in decreasing iPTH over pregnancy. TRIAL REGISTRATION: This trial is registered at clinicaltrials.gov ( NCT03308487 ). Registered 12 October 2017 'retrospectively registered'.


Assuntos
Complicações na Gravidez/tratamento farmacológico , Deficiência de Vitamina D/tratamento farmacológico , Vitamina D/administração & dosagem , Vitaminas/administração & dosagem , Adulto , Biomarcadores/sangue , Glicemia/análise , Glicemia/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Sangue Fetal/química , Humanos , Lipídeos/sangue , Estresse Oxidativo/efeitos dos fármacos , Gravidez , Complicações na Gravidez/sangue , Complicações na Gravidez/metabolismo , Resultado do Tratamento , Vitamina D/sangue , Deficiência de Vitamina D/sangue , Deficiência de Vitamina D/metabolismo , Vitaminas/sangue , Adulto Jovem
11.
Eur J Pharmacol ; 882: 173270, 2020 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-32534074

RESUMO

Intrahepatic cholestasis of pregnancy (ICP) is a pregnancy specific liver disease characterized by pruritus, elevated serum bile acids and abnormal liver function that may be associated with severe adverse pregnancy outcomes. We previously reported that plasma coenzyme Q10 (CoQ10) is decreased in women with ICP as it is its analogue coenzyme Q9 (CoQ9) in rats with ethinyl estradiol (EE)-induced cholestasis. The aim of the present study was to evaluate the possible therapeutic role of CoQ10 in experimental hepatocellular cholestasis and to compare it with ursodeoxycholic acid (UDCA) supplementation. Bile acids, CoQ9, CoQ10, transaminases, alkaline phosphatase, retinol, α-tocopherol, ascorbic acid, thiobarbituric acid reactive substances, carbonyls, glutathione, superoxide dismutase and catalase were assessed in plasma, liver and/or hepatic mitochondria in control and cholestatic rats supplemented with CoQ10 (250 mg/kg) administered alone or combined with UDCA (25 mg/kg). CoQ10 supplementation prevented bile flow decline (P < 0.05) and the increase in serum alkaline phosphatase and bile acids, particularly lithocholic acid (P < 0.05) in cholestatic rats. Furthermore, it also improved oxidative stress parameters in the liver, increased both CoQ10 and CoQ9 plasma levels and partially prevented the fall in α-tocopherol (P < 0.05). UDCA also prevented cholestasis, but it was less efficient than CoQ10 to improve the liver redox environment. Combined administration of CoQ10 and UDCA resulted in additive effects. In conclusion, present findings show that CoQ10 supplementation attenuated EE-induced cholestasis by promoting a favorable redox environment in the liver, and further suggest that it may represent an alternative therapeutic option for ICP.


Assuntos
Colestase Intra-Hepática/tratamento farmacológico , Suplementos Nutricionais , Complicações na Gravidez/tratamento farmacológico , Ubiquinona/análogos & derivados , Animais , Catalase/metabolismo , Colestase Intra-Hepática/metabolismo , Feminino , Glutationa/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Gravidez , Complicações na Gravidez/metabolismo , Ratos Sprague-Dawley , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Ubiquinona/farmacologia , Ubiquinona/uso terapêutico , Ácido Ursodesoxicólico/uso terapêutico
12.
Diabetologia ; 63(9): 1822-1835, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32472193

RESUMO

AIMS/HYPOTHESIS: During pregnancy, maternal metabolic disease and hormonal imbalance may alter fetal beta cell development and/or proliferation, thus leading to an increased risk for developing type 2 diabetes in adulthood. Although thyroid hormones play an important role in fetal endocrine pancreas development, the impact of maternal hypothyroidism on glucose homeostasis in adult offspring remains poorly understood. METHODS: We investigated this using a mouse model of hypothyroidism, induced by administration of an iodine-deficient diet supplemented with propylthiouracil during gestation. RESULTS: Here, we show that, when fed normal chow, adult mice born to hypothyroid mothers were more glucose-tolerant due to beta cell hyperproliferation (two- to threefold increase in Ki67-positive beta cells) and increased insulin sensitivity. However, following 8 weeks of high-fat feeding, these offspring gained 20% more body weight, became profoundly hyperinsulinaemic (with a 50% increase in fasting insulin concentration), insulin-resistant and glucose-intolerant compared with controls from euthyroid mothers. Furthermore, altered glucose metabolism was maintained in a second generation of animals. CONCLUSIONS/INTERPRETATION: Therefore, gestational hypothyroidism induces long-term alterations in endocrine pancreas function, which may have implications for type 2 diabetes prevention in affected individuals.


Assuntos
Glicemia/metabolismo , Intolerância à Glucose/metabolismo , Hipotireoidismo/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/embriologia , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Antitireóideos/toxicidade , Proliferação de Células , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Hiperinsulinismo/metabolismo , Resistência à Insulina , Iodo/deficiência , Ilhotas Pancreáticas/metabolismo , Camundongos , Gravidez , Propiltiouracila/toxicidade , Estresse Fisiológico
13.
Nutrients ; 12(5)2020 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-32443575

RESUMO

The circulating level of vitamin A (VA; retinol) was reported to be lower in obese adults. It is unknown if maternal obesity influences the VA status of offspring. The objective of the study was to determine the VA status and deposition of neonatal and weanling rats reared by mothers consuming a normal or high-fat diet (NFD or HFD) with or without supplemented VA. Pregnant Sprague-Dawley rats were randomized to an NFD or HFD with 2.6 mg/kg VA. Upon delivery, half of the rat mothers in the NFD or HFD cohort were switched to an NFD or HFD with supplemented VA at 129 mg/kg (NFD+VA and HFD+VA group). The other half remained on their original diet (NFD and HFD group). At postnatal day 14 (P14), P25, and P35, pups (n = 4 or 3/group/time) were euthanized. The total retinol concentration in the serum, liver, visceral white adipose tissue (WAT), and brown adipose tissue (BAT) was measured. At P14, the HFD+VA group showed a significantly lower serum VA than the NFD+VA group. At P25, both the VA concentration and total mass in the liver, WAT, and BAT were significantly higher in the HFD+VA than the NFD+VA group. At P35, the HFD group exhibited a significantly higher VA concentration and mass in the liver and BAT compared with the NFD group. In conclusion, maternal HFD consumption resulted in more VA accumulation in storage organs in neonatal and/or weanling rats, which potentially compromised the availability of VA in circulation, especially under the VA-supplemented condition.


Assuntos
Fenômenos Fisiológicos da Nutrição Materna , Obesidade/metabolismo , Complicações na Gravidez/metabolismo , Vitamina A/administração & dosagem , Vitamina A/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Animais Recém-Nascidos , Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais , Feminino , Gordura Intra-Abdominal/metabolismo , Fígado/metabolismo , Masculino , Estado Nutricional , Gravidez , Ratos , Ratos Sprague-Dawley
14.
Subcell Biochem ; 95: 27-55, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32297295

RESUMO

The placenta, a hallmark of mammalian embryogenesis, allows nutrients to be exchanged between the mother and the fetus. Vitamin A (VA), an essential nutrient, cannot be synthesized by the embryo, and must be acquired from the maternal circulation through the placenta. Our understanding of how this transfer is accomplished is still in its infancy. In this chapter, we recapitulate the early studies about the relationship between maternal dietary/supplemental VA intake and fetal VA levels. We then describe how the discovery of retinol-binding protein (RBP or RBP4), the development of labeling and detection techniques, and the advent of knockout mice shifted this field from a macroscopic to a molecular level. The most recent data indicate that VA and its derivatives (retinoids) and the pro-VA carotenoid, ß-carotene, are transferred across the placenta by distinct proteins, some of which overlap with proteins involved in lipoprotein uptake. The VA status and dietary intake of the mother influence the expression of these proteins, creating feedback signals that control the uptake of retinoids and that may also regulate the uptake of lipids, raising the intriguing possibility of crosstalk between micronutrient and macronutrient metabolism. Many questions remain about the temporal and spatial patterns by which these proteins are expressed and transferred throughout gestation. The answers to these questions are highly relevant to human health, considering that those with either limited or excessive intake of retinoids/carotenoids during pregnancy may be at risk of obtaining improper amounts of VA that ultimately impact the development and health of their offspring.


Assuntos
Desenvolvimento Embrionário , Vitamina A/metabolismo , Animais , Feminino , Humanos , Gravidez , Complicações na Gravidez/metabolismo , Proteínas de Ligação ao Retinol/metabolismo , Deficiência de Vitamina A/metabolismo , beta Caroteno/metabolismo
15.
Mol Nutr Food Res ; 64(4): e1900377, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31408914

RESUMO

The maternal obesogenic environment plays a role in programing the susceptibility of the fetus to postnatal non-alcoholic fatty liver disease (NAFLD), a risk factor for cardiovascular disease (CVD). NAFLD is a multisystem disease that is characterized by hepatic fat accumulation due in part to dysregulated energy metabolism network through epigenetic mechanisms such as DNA methylation. DNA methylation affects fetal programing and disease risk via regulation of gene transcription; it is affected by methyl donor nutrients such as vitamin B12 , methionine, folic acid, vitamin B6 , and choline. Although several studies have documented the role of several maternal methyl donor nutrients on obesity-induced NAFLD in offspring, currently, data are lacking on its impact on CVD risk as an endpoint. The aim of this paper is to use current knowledge to construct a postulation for the potential role of a comprehensive gestational methyl donor nutrients supplementary approach on the susceptibility of offspring to developing metabolic-syndrome-related cardiovascular complications.


Assuntos
Carbono/metabolismo , Doenças Cardiovasculares/etiologia , Síndrome Metabólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Complicações na Gravidez/metabolismo , Animais , Metilação de DNA , Epigênese Genética , Feminino , Humanos , Síndrome Metabólica/etiologia , Hepatopatia Gordurosa não Alcoólica/complicações , Obesidade/complicações , Gravidez
16.
Hormones (Athens) ; 19(2): 179-186, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31776808

RESUMO

Iodine is a micronutrient used by the thyroid gland to produce thyroid hormones, which manage different aspects of body metabolism. Humans depend on exogenous sources of iodine to maintain the normal concentration of thyroid hormones. Pregnancy alters iodine turnover and is associated with significant changes in thyroid function. Daily iodine requirement during pregnancy increases to 250 µg, compared with 150 µg for nonpregnant women. According to recent guidelines of scientific organizations, to improve maternal thyroid status and to prevent child neurocognitive defects, all pregnant and breastfeeding women should take 150 µg of iodine supplementation, not only in iodine-deficient regions but also in iodine-sufficient areas. However, some recent studies have confirmed that iodine supplementation of mildly iodine-deficient pregnant women has no clear benefits as concerns maternal thyroid function or child neurodevelopment.


Assuntos
Aleitamento Materno , Iodo , Lactação , Complicações na Gravidez , Oligoelementos , Adulto , Feminino , Humanos , Iodo/administração & dosagem , Iodo/deficiência , Iodo/metabolismo , Lactação/metabolismo , Gravidez , Complicações na Gravidez/tratamento farmacológico , Complicações na Gravidez/metabolismo , Oligoelementos/administração & dosagem , Oligoelementos/deficiência , Oligoelementos/metabolismo
17.
Nutrients ; 11(9)2019 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-31505767

RESUMO

This study analyzed how maternal obesity affected fatty acids (FAs) in breast milk and their association with infant growth and cognition to raise awareness about the programming effect of maternal health and to promote a healthy prenatal weight. Mother-child pairs (n = 78) were grouped per maternal pre-pregnancy body mass index (BMI): normal-weight (BMI = 18.5-24.99), overweight (BMI = 25-29.99) and obese (BMI > 30). Colostrum and mature milk FAs were determined. Infant anthropometry at 6, 18 and 36 months of age and cognition at 18 were analyzed. Mature milk exhibited lower arachidonic acid (AA) and docosahexaenoic acid (DHA), among others, than colostrum. Breast milk of non-normal weight mothers presented increased saturated FAs and n6:n3 ratio and decreased α-linolenic acid (ALA), DHA and monounsaturated FAs. Infant BMI-for-age at 6 months of age was inversely associated with colostrum n6 (e.g., AA) and n3 (e.g., DHA) FAs and positively associated with n6:n3 ratio. Depending on the maternal weight, infant cognition was positively influenced by breast milk linoleic acid, n6 PUFAs, ALA, DHA and n3 LC-PUFAs, and negatively affected by n6:n3 ratio. In conclusion, this study shows that maternal pre-pregnancy BMI can influence breast milk FAs and infant growth and cognition, endorsing the importance of a healthy weight in future generations.


Assuntos
Desenvolvimento Infantil/efeitos dos fármacos , Cognição/efeitos dos fármacos , Ácidos Graxos/análise , Leite Humano/química , Obesidade/metabolismo , Índice de Massa Corporal , Pré-Escolar , Colostro/química , Feminino , Seguimentos , Humanos , Lactente , Recém-Nascido , Fenômenos Fisiológicos da Nutrição Materna , Gravidez , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/etiologia
18.
BMJ Open ; 9(9): e026583, 2019 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-31511279

RESUMO

INTRODUCTION: The gut microbiota participates in multiple human biological processes, including metabolism and immune responses. During pregnancy, the dynamics of gut microbiota is involved in physiological adaptation. The disturbed profile of microbiome is associated with maternal complications, such as gestational diabetes mellitus (GDM), which further transfers to the offspring and influence their metabolic and immunological functions in the long term. Prebiotics targeting the gut microbiota and modulating metabolic and immune functions have been shown to be effective in non-pregnant populations with metabolic syndrome. Hence, we propose the use of a prebiotic supplement, oligosaccharide-sialic acid (OS) from the first trimester until delivery in pregnant women, can benefit maternal/new-born gut microbiome, glucose metabolism and innate immunity. METHODS AND ANALYSIS: In this prospective double-blinded randomised clinical trial, recruited singleton pregnancies will be stratified by body mass index (BMI) and randomly assigned to consume the OS preparation or placebo daily from the first trimester. At seven later time points (before and after recruitment in the first trimester, in the middle and third trimesters, before delivery, at birth and 42 days postpartum), compliance will be evaluated and/or biological samples will be collected. Along with maternal clinical information, questionnaires on lifestyle and infant development will be recorded. The primary outcomes are the effect of OS on the maternal-offspring gut microbiome and GDM incidence. The secondary outcomes are maternal glycolipid biochemical parameters, cytokine profiles, weight gain during pregnancy and infant morbidities, growth and development. The study aims to validate the effects of OS on reducing maternal morbidity within different BMI groups. The multiple dimensional dataset generated from the study includes clinical and lifestyle-related information, various biological markers and associated protective or risk factors for morbidity and prognosis. An extended follow-up through 42 days after birth could further explore the intrauterine influence on the long-term health of offspring. ETHICS AND DISSEMINATION: This protocol has been approved by Peking University First Hospital, National Unit of Clinical Trial Ethics Committee (reference number: 164). The results are expected to be published in scientific manuscripts by 2021. TRIAL REGISTRATION NUMBER: ChiCTR1800017192.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Glucose/metabolismo , Ácido N-Acetilneuramínico/farmacologia , Oligossacarídeos/farmacologia , Prebióticos , Adaptação Fisiológica , Suplementos Nutricionais , Método Duplo-Cego , Feminino , Microbioma Gastrointestinal/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Recém-Nascido , Avaliação de Resultados em Cuidados de Saúde , Gravidez , Complicações na Gravidez/imunologia , Complicações na Gravidez/metabolismo , Complicações na Gravidez/prevenção & controle , Ensaios Clínicos Controlados Aleatórios como Assunto , Projetos de Pesquisa
19.
Trends Mol Med ; 25(10): 866-881, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31383623

RESUMO

Several studies have made strong efforts to understand how age and parity modulate the risk of breast cancer. A holistic understanding of the dynamic regulation of the morphological, cellular, and molecular milieu of the mammary gland offers insights into the drivers of breast cancer development as well as into potential prophylactic interventions, the latter being a longstanding ambition of the research and clinical community aspiring to eradicate the disease. In this review we discuss mechanisms that react to pregnancy signals, and we delineate the nuances of pregnancy-associated dynamism that contribute towards either breast cancer development or prevention. Further definition of the molecular basis of parity and breast cancer risk may allow the elaboration of tools to predict and survey those who are at risk of breast cancer development.


Assuntos
Neoplasias da Mama/prevenção & controle , Complicações na Gravidez/prevenção & controle , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Epigenômica , Feminino , Humanos , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/metabolismo , Fatores de Risco
20.
Nutrients ; 11(7)2019 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-31252534

RESUMO

By serving as a precursor for the synthesis of nitric oxide, polyamines, and other molecules with biological importance, arginine plays a key role in pregnancy and fetal development. Arginine supplementation is a potential therapy for treating many human diseases. An impaired arginine metabolic pathway during gestation might produce long-term morphological or functional changes in the offspring, namely, developmental programming to increase vulnerability to developing a variety of non-communicable diseases (NCDs) in later life. In contrast, reprogramming is a strategy that shifts therapeutic interventions from adulthood to early-life, in order to reverse the programming processes, which might counterbalance the rising epidemic of NCDs. This review presented the role of arginine synthesis and metabolism in pregnancy. We also provided evidence for the links between an impaired arginine metabolic pathway and the pathogenesis of compromised pregnancy and fetal programming. This was followed by reprogramming strategies targeting the arginine metabolic pathway, to prevent the developmental programming of NCDs. Despite emerging evidence from experimental studies showing that targeting the arginine metabolic pathway has promise as a reprogramming strategy in pregnancy to prevent NCDs in the offspring, these results need further clinical application.


Assuntos
Arginina/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Doenças não Transmissíveis/epidemiologia , Estado Nutricional , Complicações na Gravidez/epidemiologia , Efeitos Tardios da Exposição Pré-Natal , Animais , Feminino , Desenvolvimento Fetal , Humanos , Gravidez , Complicações na Gravidez/metabolismo , Prognóstico , Medição de Risco , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA