Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Nutr Biochem ; 128: 109625, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38521130

ABSTRACT

Maternal obesity might induce obesity and metabolic alterations in the progeny. The study aimed to determine the effect of supplementing obese mothers with Mel (Mel) on thermogenesis and inflammation. C57BL/6 female mice (mothers) were fed from weaning to 12 weeks control diet (C, 17% kJ as fat) or a high-fat diet (HF, 49% kJ as fat) and then matted with male mice fed the control diet. Melatonin (10 mg/kg daily) was supplemented to mothers during gestation and lactation, forming the groups C, CMel, HF, and HFMel (n = 10/group). Twelve-week male offspring were studied (plasma biochemistry, immunohistochemistry, protein, and gene expressions at the hypothalamus - Hyp, subcutaneous white adipose tissue - sWAT, and interscapular brown adipose tissue - iBAT). Comparing HFMel vs. HF offspring, fat deposits and plasmatic proinflammatory markers decreased. Also, HFMel showed decreased Hyp proinflammatory markers and neuropeptide Y (anabolic) expression but improved proopiomelanocortin (catabolic) expression. Besides, HFMel sWAT adipocytes changed to a beige phenotype with-beta-3 adrenergic receptor and uncoupling protein-1 activation, concomitant with browning genes activation, triggering the iBAT thermogenic activity. In conclusion, compelling evidence indicated the beneficial effects of supplementing obese mothers with Mel on the health of their mature male offspring. Mel led to sWAT browning-related gene enhancement, increased iBAT thermogenis, and mitigated hypothalamic inflammation. Also, principal component analysis of the data significantly separated the untreated obese mother progeny from the progeny of treated obese mothers. If confirmed in humans, the findings encourage a future guideline recommending Mel supplementation during pregnancy and breastfeeding.


Subject(s)
Diet, High-Fat , Dietary Supplements , Hypothalamus , Inflammation , Melatonin , Mice, Inbred C57BL , Obesity, Maternal , Thermogenesis , Animals , Thermogenesis/drug effects , Female , Melatonin/pharmacology , Hypothalamus/metabolism , Hypothalamus/drug effects , Male , Pregnancy , Obesity, Maternal/metabolism , Inflammation/metabolism , Diet, High-Fat/adverse effects , Mice , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Obesity/metabolism , Obesity/drug therapy , Maternal Nutritional Physiological Phenomena , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics
2.
J Dev Orig Health Dis ; 14(4): 490-500, 2023 08.
Article in English | MEDLINE | ID: mdl-37366144

ABSTRACT

Melatonin supplementation to obese mothers during gestation and lactation might benefit the pancreatic islet cellular composition and beta-cell function in male offspring adulthood. C57BL/6 females (mothers) were assigned to two groups (n = 20/each) based on their consumption in control (C 17% kJ as fat) or high-fat diet (HF 49% kJ as fat). Mothers were supplemented with melatonin (Mel) (10 mg/kg daily) during gestation and lactation, or vehicle, forming the groups (n = 10/each): C, CMel, HF, and HFMel. The male offspring were studied, considering they only received the C diet after weaning until three months old. The HF mothers and their offspring showed higher body weight, glucose intolerance, insulin resistance, and low insulin sensitivity than the C ones. However, HFMel mothers and their offspring showed improved glucose metabolism and weight loss than the HF ones. Also, the offspring's higher expressions of pro-inflammatory markers and endoplasmic reticulum (ER) stress were observed in HF but reduced in HFMel. Contrarily, antioxidant enzymes were less expressed in HF but improved in HFMel. In addition, HF showed increased beta-cell mass and hyperinsulinemia but diminished in HFMel. Besides, the beta-cell maturity and identity gene expressions diminished in HF but enhanced in HFMel. In conclusion, obese mothers supplemented with melatonin benefit their offspring's islet cell remodeling and function. In addition, improving pro-inflammatory markers, oxidative stress, and ER stress resulted in better glucose and insulin levels control. Consequently, pancreatic islets and functioning beta cells were preserved in the offspring of obese mothers supplemented with melatonin.


Subject(s)
Insulin Resistance , Islets of Langerhans , Melatonin , Prenatal Exposure Delayed Effects , Female , Male , Pregnancy , Humans , Melatonin/pharmacology , Melatonin/metabolism , Obesity/metabolism , Islets of Langerhans/metabolism , Lactation/metabolism , Diet, High-Fat/adverse effects , Dietary Supplements , Maternal Nutritional Physiological Phenomena/physiology , Prenatal Exposure Delayed Effects/metabolism
3.
Nutrition ; 86: 111168, 2021 06.
Article in English | MEDLINE | ID: mdl-33601122

ABSTRACT

OBJECTIVES: Parents' lifestyle and nutrition can program offspring obesity in adulthood. We hypothesized that maternal swimming has beneficial effects on the adversity caused by paternal obesity on offspring. METHODS: Twelve-week-old male C57 BL/6 J mice (fed a high-fat diet, obese father [ObFa], or control diet, lean father [LFa]) were mated with female mice fed only the control diet. Mothers were trained (TMo) or untrained (UMo): swimming for 6 wk before and the first 2 wk of gestation. Pups were fed only the control diet. RESULTS: Fathers showed different body mass (BM) at copulation, but not the mothers. The ObFa had 20% higher BM than the LFa. Twelve-week-old ObFa/UMo offspring showed a higher BM gain than the LFa/UMo and ObFa/TMo. There was BM sexual dimorphism in the LFa/UMo (female mice +24% than male mice). There was hyperglycemia and hyperinsulinemia in the ObFa/UMo, but low glycemia and insulin levels were seen in the ObFa/TMo. There was augmented liver steatosis in the ObFa/UMo compared with the LFa/UMo, and the ObFa/TMo compared with the LFa/TMo, but reduced steatosis in the ObFa/TMo compared with the ObFa/UMo. In addition, lipogenic markers were more expressed and beta-oxidation markers less expressed in the ObFa/UMo compared with the LFa/UMo, but the opposite was observed in the ObFa/TMo compared with the ObFa/UMo. Proinflammatory markers were higher in the liver of the ObFa/UMo compared with the LFa/UMo and lower in the ObFa/TMo compared with the ObFa/UMo. CONCLUSIONS: Obese fathers produced offspring that were overweight and had altered fasting glycemia and insulin sensitivity, leading to higher liver lipogenesis and inflammation, as well as lower beta-oxidation. The swimming mother mitigated these adverse effects in mice offspring.


Subject(s)
Fathers , Prenatal Exposure Delayed Effects , Adult , Animals , Diet, High-Fat/adverse effects , Female , Humans , Liver , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Pregnancy , Swimming
4.
Int. j. morphol ; 39(1): 327-334, feb. 2021. tab
Article in English | LILACS | ID: biblio-1385298

ABSTRACT

SUMMARY: Rodents are animals extensively used in biomedical and nutrition research, a necessary step before the research in humans. The composition and type of administration of the experimental diets are relevant and should be thought, considering each type of animal used in the research. It is particularly important to consider, among others, the metabolic differences between species and food needs in macro- and micronutrients to avoid possible bias. The American Institute of Nutrition (AIN) made recommendations for rodents, adapted to the period of growth (AIN-93G), which are pivotal in fetal programming studies. The experiments can be compared among different studies and better translated into humans, considering these limitations in the nutrition of parents and offspring. The review addresses different compositions of experimental food for rodents during development with the ability to induce fetal programming in the offspring and chronic diseases in adulthood due to the nutrition of the mother and father. The 'developmental origins of health and disease' (DOHaD) concept due to maternal nutrition is commented considering the protein restriction, vitamin D restriction, obesity, and intake of fructose or fish-oil. The 'paternal origins of health and disease transmission' (POHaD), because of the nutritional state of the father, were also analyzed in the review, primarily considering the obesity of the father. The review proposes some diet compositions to experimental research considering varied nutritional situations, hoping to assist young researchers or researches not familiar with experimental diet manipulations in the elaboration of the projects.


RESUMEN: Los roedores son animales utilizados frecuentemente en la investigación biomédica y nutricional, un paso necesario antes de la investigación en humanos. La composición y el tipo de administración de las dietas experimentales son relevantes y se debe considerar cada tipo de animal utilizado en los estudios. Es particularmente importante considerar las diferencias metabólicas entre las especies y las necesidades alimentarias de macro y micronutrientes para evitar posibles sesgos. El Instituto Americano de Nutrición (AIN) estableció recomendaciones para los roedores, adaptadas al período de crecimiento (AIN-93G), que son fundamentales en los estudios de programación fetal. Los experimentos se pueden comparar entre diferentes estudios y aplicar en humanos, considerando estas limitaciones en la nutrición de padres e hijos. La revisión aborda diferentes composiciones de alimentos para estudios experimentales en roedores durante su desarrollo, con la capacidad de inducir programación fetal en la descendencia y enfermedades crónicas en la adultez, considerando la nutrición de los padres. El concepto de 'orígenes del desarrollo de la salud y la enfermedad' (DOHaD) debido a la nutrición materna se comenta considerando la restricción de proteínas, la restricción de vitamina D, la obesidad y la ingesta de fructosa o aceite de pescado. Los 'orígenes paternos de la salud y transmisión de enfermedades' (POHaD), debido al estado nutricional del padre, también fueron analizados considerando principalmente la obesidad del padre. La revisión propone algunas composiciones dietéticas a la investigación experimental considerando situaciones nutricionales variadas, con la esperanza de ayudar a jóvenes investigadores o investigadores no familiarizados con las manipulaciones experimentales de la dieta en la elaboración de los proyectos.


Subject(s)
Humans , Animals , Parenteral Nutrition , Fetal Development
5.
Mol Cell Endocrinol ; 513: 110872, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32454192

ABSTRACT

PURPOSE: We studied subcutaneous white adipose tissue (sWAT) of obese mice submitted to intermittent fasting (IF). METHODS: Twelve-week-old C57BL/6 male mice received the diets Control (C) or high-fat (HF) for eight weeks (n = 20/each). Then, part of each group performed IF (24 h feeding/24 h fasting) for four weeks: C, C-IF, HF, and HF-IF (n = 10/each). RESULTS: Food intake did not show a difference in feeding and fasting days, but HF groups had a high energy intake. IF led to multilocular adipocytes in sWAT (browning), and improved respiratory quotient on the fed day. IF decreased gene expression of Leptin, but increased Adiponectin, ß3ar (beta3 adrenoreceptor), and Ucp1 (uncoupling protein). IF enhanced immunostaining of Caspase 3, Pcna (proliferating cell nuclear antigen), and UCP1 in sWAT. IF attenuated pro-inflammatory markers and pro-apoptotic markers in sWAT. CONCLUSIONS: IF in obese mice led to browning in sWAT adipocytes, enhanced thermogenesis, an improved adipose tissue pro-inflammatory profile.


Subject(s)
Adipocytes, Brown/physiology , Adipocytes, White/physiology , Fasting/physiology , Obesity/physiopathology , Subcutaneous Fat/cytology , Animals , Cell Transdifferentiation , Diet, High-Fat , Energy Intake/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Subcutaneous Fat/physiology , Thermogenesis/physiology
6.
Nutrition ; 71: 110612, 2020 03.
Article in English | MEDLINE | ID: mdl-31785517

ABSTRACT

OBJECTIVES: The aim of this study was to observe the developmental origins of health and disease affecting offspring owing to the consumption of a diet containing high fructose by the father or mother or both, considering that progeny only received a control diet during postnatal life. METHODS: Male (future father) and female (future mother) C57 BL/6 mice were fed a high-fructose diet (HFru; 45% energy) or a control diet (C) for 8 wk before mating until lactation. The offspring was termed according to sex, maternal diet (first acrostic), and paternal diet (second acrostic); and received a balanced control diet until 3-mo of age when they were sacrificed. Body mass (BM), plasmatic leptin, adiponectin, uric acid, and systolic blood pressure (BP) were measured in mature offspring. RESULTS: Fasting glycemia and insulin were elevated in HFru fathers and mothers. Although there was no change in BM, fasting glycemia, or insulin of the offspring, those of HFru fathers, HFru mothers, and HFru fathers and mothers presented higher genital fat pad, leptin, uric acid, and BP, and lower adiponectin. The values of leptin and BP were maximized when both parents consumed a HFru diet. Also, there was sexual dimorphism in most of the variables, with the male offspring being affected to a greater extent than the females. CONCLUSIONS: Consumption of a fructose-rich diet by the father, the mother, or both negatively affected the adipokines, BP, and uric acid concentrations of mature offspring, with males being more affected than females. It is significant to consider that high BP and plasmatic uric acid correspond to markers of elevated cardiovascular risk in the progeny.


Subject(s)
Animal Nutritional Physiological Phenomena , Dietary Sugars/adverse effects , Fructose/adverse effects , Prenatal Exposure Delayed Effects/physiopathology , Sex Factors , Adiponectin/blood , Adipose Tissue/physiopathology , Animals , Blood Glucose/analysis , Blood Pressure , Fathers , Feeding Behavior , Female , Heart Disease Risk Factors , Insulin/blood , Leptin/blood , Male , Maternal Exposure , Maternal Nutritional Physiological Phenomena , Mice , Mice, Inbred C57BL , Mothers , Paternal Exposure , Pregnancy , Uric Acid/blood
7.
Nutrition ; 65: 103-112, 2019 09.
Article in English | MEDLINE | ID: mdl-31079017

ABSTRACT

OBJECTIVE: Intermittent fasting (IF) is a nutritional intervention with significant metabolic effects on the liver that are not yet fully understood. The aim of this study was to investigate the effects of IF on body mass, lipid profile, glucose metabolism, liver lipogenesis, ß-oxidation, and inflammation. METHODS: We used cellular and molecular techniques to investigate the effects of IF on 3-mo-old male C57 BL/6 mice that were fed control (10% kcal fat), high-fat (HF; 50% kcal fat), or high-fructose (HFr; 50% kcal fructose) diets for 8 wk. Half of the animals were submitted to IF (1 d fed, 1 d fast) for an additional 4 wk. RESULTS: Although food intake on the fed day did not differ between the groups, mice in the HF and HFr groups showed diminished body mass, total cholesterol, and triacylglycerol levels. Also, plasma adiponectin increased in the HFr group and leptin decreased in the HF mice. Oral glucose tolerance test and insulin were ameliorated by IF, regardless of the diet consumed (HF or HFr), and decreased hepatic lipogenesis and increased ß-oxidation markers, resulting in a reduction of the hepatic steatosis and inflammation. CONCLUSIONS: There were beneficial effects of IF even with the continuity of the obesogenic diet and proinflammatory diet in mice. It is recommended that based on the beneficial effects of IF on glucose and liver metabolism and inflammation that IF be a coadjutant factor in the treatment of hepatic metabolic issues and steatosis.


Subject(s)
Diet, Carbohydrate Loading/methods , Diet, High-Fat/methods , Fasting/metabolism , Fatty Liver/diet therapy , Fructose/administration & dosage , Adiponectin/blood , Animals , Fatty Liver/blood , Fatty Liver/physiopathology , Glucose Tolerance Test , Inflammation , Insulin Resistance , Lipids/blood , Lipogenesis , Liver/metabolism , Mice
8.
Eur J Nutr ; 57(5): 1901-1912, 2018 Aug.
Article in English | MEDLINE | ID: mdl-28534168

ABSTRACT

PURPOSE: Studies demonstrated the influence of mother's obesity on offspring. However, the father is also related to programming the future generation. The study aimed to evaluate the effects of father's obesity upon white adipose tissue (WAT) remodeling, resulting in activation of signaling pathways and inflammation in male and female offspring. METHODS: Male C57BL/6 mice received control diet (lean father group; 17% energy from lipids) or high-fat diet (obese father group; 49% energy from lipids) for 8 weeks before mating. The mothers received control diet throughout the experiment. Mice were mated: lean mother and lean father, and lean mother and obese father. Offspring received control diet from weaning until 3 months of age when they were studied. RESULTS: In the offspring, father's obesity led to decreased QUICKI with impairment of the insulin signaling pathway in both sexes. In line with these findings, in white adipose tissue, male offspring demonstrated hypertrophied adipocytes, enhanced proinflammatory cytokines, overactivation of components of the local renin-angiotensin system (RAS) and extracellular signal-regulated kinase 1/2 (ERK1/2), and inhibition of peroxisome proliferator-activated receptors (alpha and gamma). CONCLUSIONS: We observed that father's obesity influences the offspring in adult life, with an impairment in insulin homeostasis, adipocyte remodeling, and adipose tissue overexpression of IL-6 and TNF-alpha in male offspring. The activation of local RAS and ERK1/2, a concomitant PPAR diminishing, and impairment in phosphorylation of AKT and IRS-1 could explain at least in part the findings regardless of the increase in body mass in the offspring.


Subject(s)
Diet, High-Fat , Obesity/etiology , Renin-Angiotensin System/physiology , Adipose Tissue/metabolism , Animals , Brazil , Diet, High-Fat/adverse effects , Female , Male , Mice , Mice, Inbred C57BL , Obesity/epidemiology
9.
J. pediatr. (Rio J.) ; 93(6): 551-559, Nov.-Dec. 2017. graf
Article in English | LILACS | ID: biblio-894067

ABSTRACT

Abstract Objective: To discuss the recent literature on paternal obesity, focusing on the possible mechanisms of transmission of the phenotypes from the father to the children. Sources: A non-systematic review in the PubMed database found few publications in which paternal obesity was implicated in the adverse transmission of characteristics to offspring. Specific articles on epigenetics were also evaluated. As the subject is recent and still controversial, all articles were considered regardless of year of publication. Summary of findings: Studies in humans and animals have established that paternal obesity impairs their hormones, metabolism, and sperm function, which can be transmitted to their offspring. In humans, paternal obesity results in insulin resistance/type 2 diabetes and increased levels of cortisol in umbilical cord blood, which increases the risk factors for cardiovascular disease. Notably, there is an association between body fat in parents and the prevalence of obesity in their daughters. In animals, paternal obesity led to offspring alterations on glucose-insulin homeostasis, hepatic lipogenesis, hypothalamus/feeding behavior, kidney of the offspring; it also impairs the reproductive potential of male offspring with sperm oxidative stress and mitochondrial dysfunction. An explanation for these observations (human and animal) is epigenetics, considered the primary tool for the transmission of phenotypes from the father to offspring, such as DNA methylation, histone modifications, and non-coding RNA. Conclusions: Paternal obesity can induce programmed phenotypes in offspring through epigenetics. Therefore, it can be considered a public health problem, affecting the children's future life.


Resumo Objetivo: Discutir a literatura recente sobre obesidade paterna, focalizando os possíveis mecanismos de transmissão dos fenótipos do pai para os filhos. Fontes: Uma revisão não-sistemática no banco de dados PubMed encontrou poucas publicações com obesidade paterna implicada com a transmissão adversa das características à prole. Artigos específicos sobre epigenética também foram avaliados. Como o assunto é recente e ainda controverso, todos os trabalhos foram considerados independentemente do ano de publicação. Resumo dos achados: Estudos em seres humanos e animais estabeleceram que a obesidade do pai prejudica seus hormônios, metabolismo e função espermática, que pode ser transmitida à prole. Em humanos, a obesidade paterna resulta em resistência à insulina / diabetes tipo 2 e aumento do nível de cortisol no sangue do cordão umbilical, que aumenta os fatores de risco para doença cardiovascular. Notavelmente, existe associação entre a gordura corporal nos pais e a prevalência de obesidade em suas filhas. Em animais, pais obesos condicionam, na prole, a homeostase glicose-insulina, lipogênese hepática, hipotálamo / comportamento alimentar, rim, prejudicam o potencial reprodutivo da prole masculina com estresse oxidativo espermático e disfunção mitocondrial. Uma explicação para estas observações (humanos e animais) é a epigenética, considerada a ferramenta básica para a transmissão de fenótipos do pai à prole, como a metilação do DNA, modificações nas histonas, e RNA não codificante. Conclusões: A obesidade paterna pode induzir fenótipos programados na prole através da epigenética. Portanto, a obesidade paterna pode ser considerada um problema de saúde pública, afetando a vida futura das crianças.


Subject(s)
Humans , Animals , Male , Female , Epigenesis, Genetic/genetics , Fathers , Obesity/genetics , Obesity/metabolism , Models, Animal
10.
J Pediatr (Rio J) ; 93(6): 551-559, 2017.
Article in English | MEDLINE | ID: mdl-28822233

ABSTRACT

OBJECTIVE: To discuss the recent literature on paternal obesity, focusing on the possible mechanisms of transmission of the phenotypes from the father to the children. SOURCES: A non-systematic review in the PubMed database found few publications in which paternal obesity was implicated in the adverse transmission of characteristics to offspring. Specific articles on epigenetics were also evaluated. As the subject is recent and still controversial, all articles were considered regardless of year of publication. SUMMARY OF FINDINGS: Studies in humans and animals have established that paternal obesity impairs their hormones, metabolism, and sperm function, which can be transmitted to their offspring. In humans, paternal obesity results in insulin resistance/type 2 diabetes and increased levels of cortisol in umbilical cord blood, which increases the risk factors for cardiovascular disease. Notably, there is an association between body fat in parents and the prevalence of obesity in their daughters. In animals, paternal obesity led to offspring alterations on glucose-insulin homeostasis, hepatic lipogenesis, hypothalamus/feeding behavior, kidney of the offspring; it also impairs the reproductive potential of male offspring with sperm oxidative stress and mitochondrial dysfunction. An explanation for these observations (human and animal) is epigenetics, considered the primary tool for the transmission of phenotypes from the father to offspring, such as DNA methylation, histone modifications, and non-coding RNA. CONCLUSIONS: Paternal obesity can induce programmed phenotypes in offspring through epigenetics. Therefore, it can be considered a public health problem, affecting the children's future life.


Subject(s)
Epigenesis, Genetic/genetics , Fathers , Obesity/genetics , Obesity/metabolism , Animals , Female , Humans , Male , Models, Animal
11.
Biochim Open ; 4: 19-30, 2017 Jun.
Article in English | MEDLINE | ID: mdl-29450137

ABSTRACT

Studies show that the continuous consumption of fructose can lead to nonalcoholic fatty liver disease (NAFLD) and steatohepatitis. We aimed to investigate the role of Metformin in an animal model of liver injury caused by fructose intake, focusing on the molecular markers of lipogenesis, beta-oxidation, and antioxidant defenses. Male three months old C57BL/6 mice were divided into control group (C) and fructose group (F, 47% fructose), maintained for ten weeks. After, the groups received Metformin or vehicle for a further eight weeks: control (C), control + Metformin (CM), fructose (F), and fructose + Metformin (FM). Fructose resulted in hepatic steatosis, insulin resistance and lower insulin sensitivity in association with higher mRNA levels of proteins linked with de novo lipogenesis and increased lipid peroxidation. Fructose diminished mRNA expression of antioxidant enzymes, and of proteins responsible for mitochondrial biogenesis. Metformin reduced de novo lipogenesis and increased the expression of proteins related to mitochondrial biogenesis, thereby increasing beta-oxidation and decreasing lipid peroxidation. Also, Metformin upregulated the expression and activity of antioxidant enzymes, providing a defense against increased reactive oxygen species generation. Therefore, a significant reduction in triglyceride accumulation in the liver, steatosis and lipid peroxidation was observed in the FM group. In conclusion, fructose increases de novo lipogenesis, reduces the antioxidant defenses, and diminishes mitochondrial biogenesis. After an extended period of fructose intake, Metformin treatment, even in continuing the fructose intake, can reverse, at least partially, the liver injury and prevents NAFLD progression to more severe states.

12.
Nutrition ; 32(10): 1138-43, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27155954

ABSTRACT

OBJECTIVE: Mothers fed a high-fat (HF) diet can cause different adverse alterations in their offspring. The study aimed to verify the pancreatic islet structure and insulin-signaling pathway in adulthood of offspring of mothers fed a HF diet during the pregnancy. METHODS: Female mice (mothers) were randomly assigned to receive either standard chow (Mo-SC) or a HF diet (Mo-HF) ad libitum. After 2 mo on the experimental diets, 3-mo-old female mice were mated with male C57 BL/6 mice that were fed a SC diet. The male offspring was evaluated at 6 mo old. RESULTS: At 6 mo of age, Mo-HF offspring had an increment in body mass and adiposity, hypercholesterolemia, and hypertriacylglycerolemia, higher levels of insulin, and leptin with a concomitant decrease in adiponectin levels. In the islet, we observed an alteration in the structure characterized by the migration of some alpha cells from the edge to the core of the islet in association with an increase in the masses of the islet, beta cell, and alpha cell, featuring a pancreatic islet remodeling. Additionally, the Mo-HF offspring demonstrated a decrease in IRS1, PI3 k p-Akt, Pd-1, and Glut2 protein expressions compared to Mo-SC offspring. However, an increase was observed in FOXO1 and insulin protein expressions in Mo-HF offspring compared to Mo-SC offspring. CONCLUSION: The present study demonstrated that a maternal HF diet is responsible for remodeling the islet structure coupled with an adverse carbohydrate metabolism and impairment of the insulin-signaling pathway in adult male mice offspring.


Subject(s)
Diet, High-Fat/adverse effects , Insulin/metabolism , Islets of Langerhans/metabolism , Prenatal Exposure Delayed Effects/metabolism , Animals , Carbohydrate Metabolism , Female , Insulin Resistance , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/pathology , Signal Transduction
13.
Physiol Behav ; 153: 47-55, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26485293

ABSTRACT

We aimed to evaluate the effects of maternal and/or paternal obesity on offspring body mass, leptin signaling, appetite-regulating neurotransmitters and local inflammatory markers. C57BL/6 mice received standard chow (SC, lean groups) or high-fat diet (HF, obese groups) starting from one month of age. At three months, HF mice became obese relative to SC mice. They were then mated as follows: lean mother and lean father, lean mother and obese father, obese mother and lean father, and obese mother and obese father. The offspring received the SC diet from weaning until three months of age, when they were sacrificed. In the offspring, paternal obesity did not lead to changes in the Janus kinase (JAK)/signal transducer and activation of the transcription (STAT) pathway or feeding behavior but did induce hypothalamic inflammation. On the other hand, maternal obesity resulted in increased weight gain, hyperleptinemia, decreased leptin OBRb receptor expression, JAK/STAT pathway impairment, and increased SOCS3 signaling in the offspring. In addition, maternal obesity elevated inflammatory markers and altered NPY and POMC expression in the hypothalamus. Interestingly, combined parental obesity exacerbated the deleterious outcomes compared to single-parent obesity. In conclusion, while maternal obesity is known to program metabolic changes and obesity in offspring, the current study demonstrated that obese fathers induce hypothalamus inflammation in offspring, which may contribute to the development of metabolic syndromes in adulthood.


Subject(s)
Hyperphagia/metabolism , Hypothalamus/metabolism , Inflammation Mediators/metabolism , Leptin/metabolism , Obesity/metabolism , Parents , Signal Transduction , Animals , Body Weight , Diet, High-Fat , Energy Intake , Fathers , Female , Janus Kinase 1/metabolism , Leptin/blood , Male , Mice , Mothers , Neuropeptide Y/biosynthesis , Obesity/chemically induced , Obesity/physiopathology , Pro-Opiomelanocortin/biosynthesis , Receptors, Leptin/biosynthesis , STAT1 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism
14.
PLoS One ; 10(4): e0124737, 2015.
Article in English | MEDLINE | ID: mdl-25880318

ABSTRACT

AIM: To determine the impact of paternal obesity, maternal obesity or the combination of two obese parents on markers of adult offspring metabolism, with a focus on body mass (BM), lipid and carbohydrate, components of lipogenesis and beta-oxidation in the liver, sex dimorphism in the offspring that received a SC diet during the postnatal period. MATERIALS AND METHODS: Male and female C57BL/6 mice were fed a high-fat diet (HF; 49% lipids) or standard chow (SC; 17% lipids) for 8 weeks before mating until lactation. The offspring were labeled according to sex, maternal diet (first letters), paternal diet (second letters), and received a SCdiet until 12-weeks of age when they were sacrificed. BM, eating behavior, glucose tolerance, plasma analysis, gene and protein expression of the components of lipogenesis and beta-oxidation in the liver of offspring were evaluated. RESULTS: HF diet-fed mothers and fathers were overweight, hyperglycemic and glucose intolerant and had a deteriorating lipid profile. The adult male and female offspring of HF-mothers were overweight, with an increased adiposity index, hyperphagic, had an impaired glucose metabolism, increased total cholesterol and triacylglycerol levels, increased lipogenesis concomitant with decreased beta-oxidation resulting in liver steatosis. The male and female offspring of HF-father had impaired glucose metabolism, exacerbated lipogenesis without influencing beta-oxidation and enhanced hepatic steatosis. These findings are independent of BM. Male and female offspring of a mother and father that received a HF diet demonstrated these effects most prominently in adult life. CONCLUSION: Paternal obesity leads to alterations in glucose metabolism, increase in components of lipogenesis and liver steatosis. In contrast, maternal obesity leads to overweight and changes in the metabolic profile and liver resulting from activation of hepatic lipogenesis with impaired beta-oxidation. When both parents are obese, the effects observed in the male and female offspring are exacerbated.


Subject(s)
Adiposity , Biomarkers/analysis , Diet/adverse effects , Fatty Liver/etiology , Lipogenesis , Obesity/complications , Prenatal Exposure Delayed Effects/physiopathology , Animals , Blood Glucose/analysis , Blotting, Western , Body Weight , Comorbidity , Fatty Liver/metabolism , Female , Male , Maternal Nutritional Physiological Phenomena , Mice , Mice, Inbred C57BL , Obesity/physiopathology , Parents , Pregnancy , Prenatal Nutritional Physiological Phenomena , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
15.
Nutr Res ; 35(2): 136-45, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25582085

ABSTRACT

We hypothesized that the maternal obesity initiates metabolic disorders associated with oxidative stress in the liver of offspring since early life. Mouse's mothers were assigned into 2 groups according to the diet offered (n = 10 per group): standard chow (SC) or high-fat diet (HF). The results revealed that HF offspring had an increase in body mass at day 10 (+25%, P < .05) and in glucose levels (+25%, P < .0001). Hepatic triacylglycerol was increased in HF offspring at day 1 and day 10 compared with SC offspring (+30%, P < .01 and +40%, P < .01) as was hepatic steatosis (+110%, P < .001; +145%, P < .0001). Fatty acid synthase was increased in HF offspring at day 1 (+450%, P < .01) and peroxisome proliferator activator receptor-γ was elevated at day 1 and day 10 (+140%, P < .01; +2741%, P < .01). Peroxisome proliferator activator receptor-α was diminished in HF offspring at day 10 compared with SC offspring (-100%, P < .01). Moreover, carnitine palmitoyl-CoA transferase-1 was decreased in HF offspring at day 1 and day 10 (-80%, P < .01; -60%, P < .05). In the HF offspring (compared with the SC offspring), the catalase and the superoxide dismutase were significantly lower in both days 1 and 10 (P < .05). In 10-day-old offspring, glutathione peroxidase 1 and glutathione reductase were lower in HF offspring than in SC offspring (P < .0001). Our findings suggest that the maternal obesity in mice induces an early oxidative dysfunction coupled with hepatic steatosis and might contribute to progressive liver injury later in life.


Subject(s)
Animals, Newborn/growth & development , Body Weight , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Liver/metabolism , Prenatal Exposure Delayed Effects/metabolism , Animals , Animals, Newborn/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Catalase/metabolism , Fatty Liver/metabolism , Female , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Male , Mice , Mice, Obese , PPAR alpha/metabolism , PPAR gamma/metabolism , Pregnancy , Superoxide Dismutase/metabolism , Triglycerides/metabolism , Glutathione Peroxidase GPX1
16.
Life Sci ; 93(12-14): 454-63, 2013 Oct 06.
Article in English | MEDLINE | ID: mdl-23973955

ABSTRACT

AIMS: To investigate whether the effects of diet-induced obesity in mothers are passed on to their offspring fed a control diet in a gender-specific manner. MAIN METHODS: Mother mice received either standard chow (SC; 17% energy from fat) or high-fat (HF; 49% energy from fat) diet for eight weeks pre-pregnancy until lactation. After weaning (at 21 days of age), offspring received SC diet and were divided into four groups according to the mother's diet (Mo): male Mo-SC, female Mo-SC, male Mo-HF, and female Mo-HF. Stereology, Elisa and western blotting were performed. KEY FINDINGS: HF diet-fed mothers were overweight, and had metabolic abnormalities, all of which were found in their adult offspring. Male Mo-HF offspring had higher cholesterol, triglycerides, leptin and insulin levels and lower circulating adiponectin than female Mo-HF offspring. Mo-HF offspring of both genders had higher expression of tumor necrosis factor-alpha, interleukin-6 and leptin and lower expression of adiponectin than Mo-SC offspring; however, male Mo-HF were more affected than female Mo-HF offspring for these variables, demonstrating sexual dimorphism. SIGNIFICANCE: Exposure to HF diet is effective in inducing obesity and metabolic alterations in mothers, and this phenotype can be passed on to their offspring. An adverse pattern in the body fat distribution in males probably has favored the intensification of a pro-inflammatory profile compared with females. In adulthood, the male offspring responds to the maternal obesity more than the female offspring, indicating a relevant sexual dimorphism that is a novel finding in this animal study.


Subject(s)
Body Fat Distribution , Obesity/complications , Prenatal Exposure Delayed Effects/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Blood Glucose , Blood Pressure , Diet, High-Fat/adverse effects , Energy Intake , Female , Leptin/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Organ Size , Pregnancy , Prenatal Exposure Delayed Effects/blood , Prenatal Exposure Delayed Effects/physiopathology , Sex Characteristics , Weight Gain
17.
PLoS One ; 8(1): e55711, 2013.
Article in English | MEDLINE | ID: mdl-23383269

ABSTRACT

Maternal obesity induced by a high fat (HF) diet may program susceptibility in offspring, altering pancreatic development and causing later development of chronic degenerative diseases, such as obesity and diabetes. Female mice were fed standard chow (SC) or an HF diet for 8 weeks prior to mating and during the gestational and lactational periods. The male offspring were assessed at birth, at 10 days, and at 3 months of age. The body mass (BM) gain was 50% greater before pregnancy and 80% greater during pregnancy in HF dams than SC dams. Dams fed an HF diet showed higher oral glucose tolerance test (OGTT), blood pressure, serum corticosterone, and insulin levels than dams fed SC. At 10 days of age and at 3 mo old the HF offspring showed greater BM and higher blood glucose levels than the SC offspring. The mean diameter of the islets had increased by 37% in the SC offspring and by 155% in the HF offspring at 10 days of age. The islet mass ratio (IM/PM) was 88% greater in the HF offspring at 10 days of age, and 107% greater at 3 mo of age, compared to the values obtained at birth. The HF offspring had a beta cell mass (BCM)/PM ratio 54% lower than SC offspring at birth. However, HF offspring displayed a 146% increase in the BCM/PM ratio at 10 days of age, and 112% increase at 3 months of age than values at birth. A 3 mo of age, the HF offspring showed a greater OGTT and higher levels of than SC offspring. In conclusion, a maternal HF diet consumed during the preconceptional period and throughout the gestational and lactational periods in mice results in dramatic alterations in the pancreata of the offspring.


Subject(s)
Maternal Nutritional Physiological Phenomena , Obesity/etiology , Pancreas/growth & development , Prenatal Exposure Delayed Effects , Adiposity , Animals , Blood Glucose , Blood Pressure , Body Weight , Corticosterone/blood , Diet, High-Fat , Disease Susceptibility , Female , Insulin/blood , Islets of Langerhans/cytology , Mice , Organ Size , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...