Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(3)2023 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-36768137

RESUMO

There is growing evidence that poor paternal diet at the time of conception increase the risk of offspring developing a range of non-communicable metabolic diseases, such as obesity, diabetes and cardiovascular disease, in adulthood. We hypothesise that a paternal low protein-high carbohydrate diet perturbs offspring tissue lipid abundance through both sperm and seminal plasma-mediated mechanisms. To test our hypothesis, we fed male C57BL/6 mice either a control normal protein diet (NPD; 18% protein) or an isocaloric low protein diet (LPD; 9% protein) for a minimum of 8 weeks. We generated offspring through artificial insemination, in combination with vasectomised male mating. Using this approach, we derived offspring from either NPD or LPD sperm but in the presence of NPD or LPD seminal plasma. Using high resolution mass-spectrometry, we found that offspring derived from either LPD sperm or seminal fluid displayed perturbed cardiac and brain lipid abundance from just three weeks of age, typically associated with the altered abundance of tissue triglycerides. We also observed the differential sex-specific patterns of lipids between the control and experimental offspring's hearts and brains. These observations indicate that poor paternal diet at the time of conception affects offspring cardiac and brain lipid profiles in an age-, sex- and generation-specific manner.


Assuntos
Encéfalo , Sêmen , Feminino , Camundongos , Masculino , Animais , Camundongos Endogâmicos C57BL , Homeostase , Lipídeos
2.
Semin Cell Dev Biol ; 97: 131-137, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31254609

RESUMO

The concept that a father's wellbeing at the time of conception influences the development and long-term health of his offspring is now well established. However, the mechanisms underlying the paternal programming of offspring health are not fully defined. While sperm-mediated effects on offspring development have been investigated in detail, the significance of seminal plasma has been over-looked. Typically, the seminal plasma is viewed as a simple medium, with a main role to transport sperm into the female reproductive tract at the time of conception. However, a more sophisticated role for seminal plasma in the modulation of the maternal periconception cell-signalling, inflammatory and immunological physiology is emerging. Seminal plasma comprises a complex mix of nutrients, proteins, signalling molecules and cell-free genetic material which all interact with the endometrium to regulate gene expression, vascular remodelling, leukocyte recruitment and the priming of regulatory T cells (Tregs). These seminal plasma effects on the maternal periconception environment all act to facilitate uterine remodelling, embryo implantation and fetal development. Evidence is now emerging that poor paternal lifestyle factors such as diet, can modify these essential uterine responses, altering fetal development and ultimately long-term offspring health. The use of animal models has enhanced our understanding of the effects of seminal plasma on maternal uterine physiology, embryo development and offspring health. However, further studies are needed to define the interaction between seminal plasma components and female reproductive tissues in humans. Such studies will be central in providing better information and infertility treatments to intending parents.


Assuntos
Desenvolvimento Embrionário/genética , Sêmen/química , Humanos , Masculino
3.
Metabolomics ; 18(2): 13, 2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-35141784

RESUMO

BACKGROUND: The paternal diet affects lipid metabolism in offspring for at least two generations through nutritional programming. However, we do not know how this is propagated to the offspring. OBJECTIVES: We tested the hypothesis that the changes in lipid metabolism that are driven by paternal diet are propagated through spermatozoa and not seminal plasma. METHODS: We applied an updated, purpose-built computational network analysis tool to characterise control of lipid metabolism systemically (Lipid Traffic Analysis v2.3) on a known mouse model of paternal nutritional programming. RESULTS: The analysis showed that the two possible routes for programming effects, the sperm (genes) and seminal plasma (influence on the uterine environment), both have a distinct effect on the offspring's lipid metabolism. Further, the programming effects in offspring suggest that changes in lipid distribution are more important than alterations in lipid biosynthesis. CONCLUSIONS: These results show how the uterine environment and genes both affect lipid metabolism in offspring, enhancing our understanding of the link between parental diet and metabolism in offspring.


Assuntos
Metabolismo dos Lipídeos , Sêmen , Animais , Pai , Humanos , Masculino , Metabolômica , Camundongos , Espermatozoides/metabolismo
4.
Reproduction ; 162(5): F101-F109, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-33544695

RESUMO

Pregnancy represents a time of dramatic physiological adaptation by the mother in which dramatic changes in maternal cardiovascular, metabolic and immune systems occur. These adaptations, initiated from the earliest stages of gestation, are crucial for the implantation and continued development of the embryo, the establishment of the placenta and the growth of the fetus. Impairments in the normal adaptation of the maternal cardiovascular, metabolic and immune systems underlie the aetiology of gestational disorders such as preeclampsia and gestational diabetes. Studies have shown that the development of such gestational complications not only affects the well-being of the mother but also the short- and long-term health of her offspring. While the connection between maternal lifestyle factors and the development of gestational disorders such as preeclampsia and gestational diabetes has been studied in detail, the link between a father's lifestyle and the well-being of the mother during pregnancy has received less attention. In this review, we will explore the evidence that a range of paternal factors, such as age and diet, at the time of conception can not only affect the development of his offspring, but also the well-being of the mother during pregnancy. In addition, we will examine the sperm- and seminal plasma-specific mechanisms that connect the health of the father with that of the mother and his offspring.


Assuntos
Saúde Materna , Complicações na Gravidez , Dieta , Pai , Feminino , Humanos , Masculino , Gravidez , Sêmen
5.
Anal Bioanal Chem ; 413(19): 4763-4773, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34254158

RESUMO

Detailed molecular analysis is of increasing importance in research into the regulation of biochemical pathways, organismal growth and disease. Lipidomics in particular is increasingly sought after as it provides insight into molecular species involved in energy storage, signalling and fundamental cellular structures. This has led to the use of a range of tools and techniques to acquire lipidomics data. 31P NMR for lipidomics offers well-resolved head group/lipid class analysis, structural data that can be used to inform and strengthen interpretation of mass spectrometry data and part of a priori structural determination. In the present study, we codify the use of 31P NMR for lipidomics studies to make the technique more accessible to new users and more useful for a wider range of questions. The technique can be used in isolation (phospholipidomics) or as a part of determining lipid composition (lipidomics). We describe the process from sample extraction to data processing and analysis. This pipeline is important because it allows greater thoroughness in lipidomics studies and increases scope for answering scientific questions about lipid-containing systems.


Assuntos
Lipidômica/métodos , Lipídeos/química , Espectroscopia de Ressonância Magnética/métodos , Isótopos de Fósforo/química , Animais , Camundongos
6.
Proc Natl Acad Sci U S A ; 115(40): 10064-10069, 2018 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-30150380

RESUMO

The association between poor paternal diet, perturbed embryonic development, and adult offspring ill health represents a new focus for the Developmental Origins of Health and Disease hypothesis. However, our understanding of the underlying mechanisms remains ill-defined. We have developed a mouse paternal low-protein diet (LPD) model to determine its impact on semen quality, maternal uterine physiology, and adult offspring health. We observed that sperm from LPD-fed male mice displayed global hypomethylation associated with reduced testicular expression of DNA methylation and folate-cycle regulators compared with normal protein diet (NPD) fed males. Furthermore, females mated with LPD males display blunted preimplantation uterine immunological, cell signaling, and vascular remodeling responses compared to controls. These data indicate paternal diet impacts on offspring health through both sperm genomic (epigenetic) and seminal plasma (maternal uterine environment) mechanisms. Extending our model, we defined sperm- and seminal plasma-specific effects on offspring health by combining artificial insemination with vasectomized male mating of dietary-manipulated males. All offspring derived from LPD sperm and/or seminal plasma became heavier with increased adiposity, glucose intolerance, perturbed hepatic gene expression symptomatic of nonalcoholic fatty liver disease, and altered gut bacterial profiles. These data provide insight into programming mechanisms linking poor paternal diet with semen quality and offspring health.


Assuntos
Exposição Dietética , Proteínas Alimentares/administração & dosagem , Exposição Paterna , Sêmen/metabolismo , Espermatozoides/metabolismo , Testículo/metabolismo , Animais , Epigênese Genética/efeitos dos fármacos , Feminino , Masculino , Camundongos , Análise do Sêmen , Útero/metabolismo
7.
J Physiol ; 598(4): 699-715, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31617219

RESUMO

KEY POINTS: A low protein diet had minimal effects on paternal cardiovascular function or renin-angiotensin system activity. Paternal low protein diet modified F1 neonatal and adult offspring renin-angiotensin system activity and cardiovascular function in a sperm and/or seminal plasma specific manner. Paternal low protein diet modified F1 male offspring testicular expression of central epigenetic regulators. Significant changes in F2 neonatal offspring growth and tissue angiotensin-converting enzyme activity were programmed by paternal low protein diet in a sperm and/or seminal plasma specific manner. ABSTRACT: Although the impact of maternal diet on adult offspring health is well characterized, the role that a father's diet has on his offspring's health remains poorly defined. We establish the significance of a sup-optimal paternal low protein diet for offspring vascular homeostasis and define the sperm and seminal plasma specific programming effects on cardiovascular health. Male C57BL6 mice were fed either a control normal protein diet (NPD; 18% protein) or an isocaloric low protein diet (LPD; 9% protein) for a minimum of 7 weeks. Using artificial insemination, in combination with vasectomized male mating, we generated offspring derived from either NPD or LPD sperm (devoid of seminal plasma) but in the presence of NPD or LPD seminal plasma (devoid of sperm). We observed that either LPD sperm or seminal fluid at conception impaired adult offspring vascular function in response to both vasoconstrictors and dilators. Underlying these changes in vascular function were significant changes in serum, lung and kidney angiotensin-converting enzyme (ACE) activity, established in F1 offspring from 3 weeks of age, maintained into adulthood and present also within juvenile F2 offspring. Furthermore, we observed differential expression of multiple central renin-angiotensin system regulators in adult offspring kidneys. Finally, paternal diet modified the expression profiles of central epigenetic regulators of DNA methylation, histone modifications and RNA methylation in adult F1 male testes. These novel data reveal the impact of sub-optimal paternal nutrition on offspring cardiovascular well-being, programming offspring cardiovascular function through both sperm and seminal plasma specific mechanisms over successive generations.


Assuntos
Sistema Cardiovascular/fisiopatologia , Dieta com Restrição de Proteínas , Pai , Fenômenos Fisiológicos da Nutrição , Sêmen , Espermatozoides , Animais , Epigênese Genética , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
Hum Reprod ; 35(11): 2497-2514, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-33020802

RESUMO

STUDY QUESTION: Do the long-term health outcomes following IVF differ depending upon the duration of embryo culture before transfer? SUMMARY ANSWER: Using a mouse model, we demonstrate that in male but not female offspring, adverse cardiovascular (CV) health was more likely with prolonged culture to the blastocyst stage, but metabolic dysfunction was more likely if embryo transfer (ET) occurred at the early cleavage stage. WHAT IS KNOWN ALREADY: ART associate with increased risk of adverse CV and metabolic health in offspring, and these findings have been confirmed in animal models in the absence of parental infertility issues. It is unclear which specific ART treatments may cause these risks. There is increasing use of blastocyst, versus cleavage-stage, transfer in clinical ART which does not appear to impair perinatal health of children born, but the longer-term health implications are unknown. STUDY DESIGN, SIZE, DURATION: Five mouse groups were generated comprising: (i) natural mating (NM)-naturally mated, non-superovulated and undisturbed gestation; (ii) IV-ET-2Cell-in-vivo derived two-cell embryos collected from superovulated mothers, with immediate ET to recipients; (iii) IVF-ET-2Cell-IVF generated embryos, from oocytes from superovulated mothers, cultured to the two-cell stage before ET to recipients; (iv) IV-ET-BL-in-vivo derived blastocysts collected from superovulated mothers, with immediate ET to recipients; (v) IVF-ET-BL-IVF generated embryos, from oocytes from superovulated mothers, cultured to the blastocyst stage before ET to recipients. Both male and female offspring were analysed for growth, CV and metabolic markers of health. There were 8-13 litters generated for each group for analyses; postnatal data were analysed by multilevel random effects regression to take account of between-mother and within-mother variation and litter size. PARTICIPANTS/MATERIALS, SETTINGS, METHODS: C57/BL6 female mice (3-4 weeks old) were used for oocyte production; CBA males for sperm with human tubal fluid medium were used for IVF. Embryos were transferred (ET) to MF1 pseudo-pregnant recipients at the two-cell stage or cultured in synthetic oviductal medium enriched with potassium medium to the blastocyst stage before ET. Control in-vivo embryos from C57BL6 × CBA matings were collected and immediately transferred at the two-cell or blastocyst stage. Postnatal assays included growth rate up to 27 weeks; systolic blood pressure (SBP) at 9, 15 and 21 weeks; lung and serum angiotensin-converting enzyme (ACE) activity at time of cull (27 weeks); glucose tolerance test (GTT; 27 weeks); basal glucose and insulin levels (27 weeks); and lipid accumulation in liver cryosections using Oil Red O imaging (27 weeks). MAIN RESULTS AND THE ROLE OF CHANCE: Blastocysts formed by IVF developed at a slower rate and comprised fewer cells that in-vivo generated blastocysts without culture (P < 0.05). Postnatal growth rate was increased in all four experimental treatments compared with NM group (P < 0.05). SBP, serum and lung ACE and heart/body weight were higher in IVF-ET-BL versus IVF-ET-2Cell males (P < 0.05) and higher than in other treatment groups, with SBP and lung ACE positively correlated (P < 0.05). Glucose handling (GTT AUC) was poorer and basal insulin levels were higher in IVF-ET-2Cell males than in IVF-ET-BL (P < 0.05) with the glucose:insulin ratio more negatively correlated with body weight in IVF-ET-2Cell males than in other groups. Liver/body weight and liver lipid droplet diameter and density in IVF-ET-2Cell males were higher than in IVF-ET-BL males (P < 0.05). IVF groups had poorer health characteristics than their in-vivo control groups, indicating that outcomes were not caused specifically by background techniques (superovulation, ET). No consistent health effects from duration of culture were identified in female offspring. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Results from experimental animal models cannot be extrapolated to humans. Nevertheless, they are valuable to develop conceptual models, in this case, in the absence of confounding parental infertility, in assessing the safety of ART manipulations. WIDER IMPLICATIONS OF THE FINDINGS: The study indicates that longer duration of embryo culture after IVF up to blastocyst before ET leads to increased dysfunction of CV health in males compared with IVF and shorter cleavage-stage ET. However, the metabolic health of male offspring was poorer after shorter versus longer culture duration. This distinction indicates that the origin of CV and metabolic health phenotypes after ART may be different. The poorer metabolic health of males after cleavage-stage ET coincides with embryonic genome activation occurring at the time of ET. STUDY FUNDING/COMPETING INTEREST(S): This work was supported through the European Union FP7-CP-FP Epihealth programme (278418) and FP7-PEOPLE-2012-ITN EpiHealthNet programme (317146) to T.P.F., the Biotechnology and Biological Sciences Research Council (BBSRC) (BB/F007450/1) to T.P.F., and the Saudi government, University of Jeddah and King Abdulaziz University to A.A. The authors have no conflicts of interest to declare.


Assuntos
Blastocisto , Técnicas de Cultura Embrionária , Animais , Transferência Embrionária , Feminino , Fertilização in vitro , Masculino , Camundongos , Camundongos Endogâmicos CBA , Gravidez
9.
Reproduction ; 159(5): 627-641, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32163913

RESUMO

The link between male diet and sperm quality has received significant investigation. However, the impact diet and dietary supplements have on the testicular environment has been examined to a lesser extent. Here, we establish the impact of a sub-optimal low protein diet (LPD) on testicular morphology, apoptosis and serum fatty acid profiles. Furthermore, we define whether supplementing a LPD with specific methyl donors abrogates any detrimental effects of the LPD. Male C57BL6 mice were fed either a control normal protein diet (NPD; 18% protein; n = 8), an isocaloric LPD (LPD; 9% protein; n = 8) or an LPD supplemented with methyl donors (MD-LPD; choline chloride, betaine, methionine, folic acid, vitamin B12; n = 8) for a minimum of 7 weeks. Analysis of male serum fatty acid profiles by gas chromatography revealed elevated levels of saturated fatty acids and lower levels of mono- and polyunsaturated fatty acids in MD-LPD males when compared to NPD and/or LPD males. Testes of LPD males displayed larger seminiferous tubule cross section area when compared to NPD and MD-LPD males, while MD-LPD tubules displayed a larger luminal area. Furthermore, TUNNEL staining revealed LPD males possessed a reduced number of tubules positive for apoptosis, while gene expression analysis showed MD-LPD testes displayed decreased expression of the pro-apoptotic genes Bax, Csap1 and Fas when compared to NPD males. Finally, testes from MD-LPD males displayed a reduced telomere length but increased telomerase activity. These data reveal the significance of sub-optimal nutrition for paternal metabolic and reproductive physiology.


Assuntos
Dieta com Restrição de Proteínas , Suplementos Nutricionais , Testículo/efeitos dos fármacos , Testículo/fisiologia , Animais , Betaína/administração & dosagem , Colina/administração & dosagem , Ácidos Graxos/sangue , Ácido Fólico/administração & dosagem , Masculino , Metionina/administração & dosagem , Camundongos , Vitamina B 12/administração & dosagem
10.
Molecules ; 25(14)2020 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-32668693

RESUMO

Extraction of the lipid fraction is a key part of acquiring lipidomics data. High-throughput lipidomics, the extraction of samples in 96w plates that are then run on 96 or 384w plates, has particular requirements that mean special development work is needed to fully optimise an extraction method. Several methods have been published as suitable for it. Here, we test those methods using four liquid matrices: milk, human serum, homogenised mouse liver and homogenised mouse heart. In order to determine the difference in performance of the methods as objectively as possible, we used the number of lipid variables identified, the total signal strength and the coefficient of variance to quantify the performance of the methods. This showed that extraction methods with an aqueous component were generally better than those without for these matrices. However, methods without an aqueous fraction in the extraction were efficient for milk samples. Furthermore, a mixture containing a chlorinated solvent (dichloromethane) appears to be better than an ethereal solvent (tert-butyl methyl ether) for extracting lipids. This study suggests that a 3:1:0.005 mixture of dichloromethane, methanol and triethylammonium chloride, with an aqueous wash, is the most efficient of the currently reported methods for high-throughput lipid extraction and analysis. Further work is required to develop non-aqueous extraction methods that are both convenient and applicable to a broad range of sample types.


Assuntos
Lipídeos/isolamento & purificação , Fígado/química , Leite/química , Miocárdio/química , Soro/química , Manejo de Espécimes/métodos , Animais , Humanos , Lipidômica , Lipídeos/análise , Camundongos , Solventes/química
11.
Lancet ; 391(10132): 1842-1852, 2018 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-29673874

RESUMO

Parental environmental factors, including diet, body composition, metabolism, and stress, affect the health and chronic disease risk of people throughout their lives, as captured in the Developmental Origins of Health and Disease concept. Research across the epidemiological, clinical, and basic science fields has identified the period around conception as being crucial for the processes mediating parental influences on the health of the next generation. During this time, from the maturation of gametes through to early embryonic development, parental lifestyle can adversely influence long-term risks of offspring cardiovascular, metabolic, immune, and neurological morbidities, often termed developmental programming. We review periconceptional induction of disease risk from four broad exposures: maternal overnutrition and obesity; maternal undernutrition; related paternal factors; and the use of assisted reproductive treatment. Studies in both humans and animal models have demonstrated the underlying biological mechanisms, including epigenetic, cellular, physiological, and metabolic processes. We also present a meta-analysis of mouse paternal and maternal protein undernutrition that suggests distinct parental periconceptional contributions to postnatal outcomes. We propose that the evidence for periconceptional effects on lifetime health is now so compelling that it calls for new guidance on parental preparation for pregnancy, beginning before conception, to protect the health of offspring.


Assuntos
Desenvolvimento Embrionário/fisiologia , Epigênese Genética , Efeitos Tardios da Exposição Pré-Natal , Fenômenos Fisiológicos da Nutrição Pré-Natal , Animais , Dieta , Feminino , Fertilização , Humanos , Camundongos , Obesidade/fisiopatologia , Gravidez
12.
Adv Exp Med Biol ; 1200: 71-89, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31471795

RESUMO

The ability to adapt to changing environmental conditions is critical for any species to survive. Many environmental changes occur too rapidly for an organism's genome to adapt in time. Accordingly, being able to modify either its own phenotype, or the phenotype of its offspring to better suit future anticipated environmental conditions could afford an organism a significant advantage. However, a range of animal models and human epidemiological data sets are now showing that environmental factors such as changes in the quality or quantity of an individual's diet, temperature, stress or exposure to pollutants can all adversely affect the quality of parental gametes, the development of the preimplantation embryo and the health and wellbeing of offspring over multiple generations. This chapter will examine transgenerational effects of both maternal and paternal environmental factors on offspring development and wellbeing in both human and animal model studies. Changes in the epigenetic status of either parental or grand-parental gametes provide one candidate mechanism through which the impacts of environmental experience can be passed from one generation to another. This chapter will therefore also focus on the impact of parental and grand-parental diet on epigenetic transgenerational inheritance and offspring phenotype.


Assuntos
Adaptação Biológica/genética , Dieta , Meio Ambiente , Epigênese Genética , Animais , Humanos , Modelos Animais , Fenótipo
13.
Biochim Biophys Acta ; 1859(7): 906-13, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27060415

RESUMO

Cells and organisms respond to nutrient deprivation by decreasing global rates of transcription, translation and DNA replication. To what extent such changes can be reversed is largely unknown. We examined the effect of maternal dietary restriction on RNA synthesis in the offspring. Low protein diet fed either throughout gestation or for the preimplantation period alone reduced cellular RNA content across fetal somatic tissues during challenge and increased it beyond controls in fetal and adult tissues after challenge release. Changes in transcription of ribosomal RNA, the major component of cellular RNA, were responsible for this phenotype as evidenced by matching alterations in RNA polymerase I density and DNA methylation at ribosomal DNA loci. Cellular levels of the ribosomal transcription factor Rrn3 mirrored the rRNA expression pattern. In cell culture experiments, Rrn3 overexpression reduced rDNA methylation and increased rRNA expression; the converse occurred after inhibition of Rrn3 activity. These observations define novel mechanism where poor nutrition before implantation irreversibly alters basal rates of rRNA transcription thereafter in a process mediated by rDNA methylation and Rrn3 factor.


Assuntos
Implantação do Embrião/genética , Regulação da Expressão Gênica no Desenvolvimento , Fenômenos Fisiológicos da Nutrição Materna , Efeitos Tardios da Exposição Pré-Natal/genética , RNA Ribossômico/genética , Animais , Dieta , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Feminino , Células HEK293 , Humanos , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia
14.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1371-1381, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28189722

RESUMO

Defining the mechanisms underlying the programming of early life growth is fundamental for improving adult health and wellbeing. While the association between maternal diet, offspring growth and adult disease risk is well-established, the effect of father's diet on offspring development is largely unknown. Therefore, we fed male mice an imbalanced low protein diet (LPD) to determine the impact on post-fertilisation development and fetal growth. We observed that in preimplantation embryos derived from LPD fed males, expression of multiple genes within the central metabolic AMPK pathway was reduced. In late gestation, paternal LPD programmed increased fetal weight, however, placental weight was reduced, resulting in an elevated fetal:placental weight ratio. Analysis of gene expression patterns revealed increased levels of transporters for calcium, amino acids and glucose within LPD placentas. Furthermore, placental expression of the epigenetic regulators Dnmt1 and Dnmt3L were increased also, coinciding with altered patterns of maternal and paternal imprinted genes. More strikingly, we observed fetal skeletal development was perturbed in response to paternal LPD. Here, while offspring of LPD fed males possessed larger skeletons, their bones comprised lower volumes of high mineral density in combination with reduced maturity of bone apatite. These data offer new insight in the underlying programming mechanisms linking poor paternal diet at the time of conception with the development and growth of his offspring.


Assuntos
Blastocisto/metabolismo , Proteínas Alimentares , Epigênese Genética , Desenvolvimento Fetal , Regulação da Expressão Gênica no Desenvolvimento , Desenvolvimento Musculoesquelético , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Deficiência de Proteína/metabolismo , Animais , Blastocisto/patologia , Feminino , Masculino , Camundongos , Gravidez , Complicações na Gravidez/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Deficiência de Proteína/patologia
15.
Adv Exp Med Biol ; 1014: 137-154, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28864989

RESUMO

The number of adults afflicted with heart disease, obesity and diabetes, central components of metabolic disorder, has grown rapidly in recent decades, affecting up to one quarter of the world's population. Typically, these diseases are attributed to lifestyle factors such as poor diet, lack of exercise and smoking. However, studies have now identified strong associations between patterns of growth during foetal and neonatal life and an increase predisposition towards developing heart disease, obesity and diabetes in adult life. While the connection between a mother's diet and the long-term health of her offspring has been studied in great detail, our understanding of whether offspring health might be affected by a father's diet remains limited. Greater insight into the impact that paternal nutrition has on sperm quality, epigenetic status and potential offspring programming mechanisms is needed to redress this parental-programming knowledge imbalance. Disturbances in paternal reproductive epigenetic status represents one key mechanism linking paternal diet with the programing of offspring development and adult health, as many enzymatic processes involved in epigenetic regulation use metabolic intermediates to modify DNA and histones. Here, poor paternal nutrition could result in perturbed sperm and testicular epigenetic status, impacting on post-fertilisation gene transcriptional regulation and protein expression in offspring tissues, resulting in increased incidences of metabolic disorder in adult life.


Assuntos
Desenvolvimento Embrionário , Epigênese Genética , Fertilização , Feminino , Humanos , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Estado Nutricional , Exposição Paterna , Fenótipo , Gravidez
17.
Reproduction ; 149(6): 563-75, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25755287

RESUMO

Mammalian placentation is dependent upon the action of trophoblast cells at the time of implantation. Appropriate fetal growth, regulated by maternal nutrition and nutrient transport across the placenta, is a critical factor for adult offspring long-term health. We have demonstrated that a mouse maternal low-protein diet (LPD) fed exclusively during preimplantation development (Emb-LPD) increases offspring growth but programmes adult cardiovascular and metabolic disease. In this study, we investigate the impact of maternal nutrition on post-implantation trophoblast phenotype and fetal growth. Ectoplacental cone explants were isolated at day 8 of gestation from female mice fed either normal protein diet (NPD: 18% casein), LPD (9% casein) or Emb-LPD and cultured in vitro. We observed enhanced spreading and cell division within proliferative and secondary trophoblast giant cells (TGCs) emerging from explants isolated from LPD-fed females when compared with NPD and Emb-LPD explants after 24 and 48 h. Moreover, both LPD and Emb-LPD explants showed substantial expansion of TGC area during 24-48 h, not observed in NPD. No difference in invasive capacity was observed between treatments using Matrigel transwell migration assays. At day 17 of gestation, LPD- and Emb-LPD-fed conceptuses displayed smaller placentas and larger fetuses respectively, resulting in increased fetal:placental ratios in both groups compared with NPD conceptuses. Analysis of placental and yolk sac nutrient signalling within the mammalian target of rapamycin complex 1 pathway revealed similar levels of total and phosphorylated downstream targets across groups. These data demonstrate that early post-implantation embryos modify trophoblast phenotype to regulate fetal growth under conditions of poor maternal nutrition.


Assuntos
Desenvolvimento Fetal/fisiologia , Células Gigantes/citologia , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Placentação/fisiologia , Trofoblastos/citologia , Animais , Movimento Celular/fisiologia , Dieta com Restrição de Proteínas , Feminino , Células Gigantes/metabolismo , Camundongos , Fosforilação , Gravidez , Transdução de Sinais/fisiologia , Trofoblastos/metabolismo
18.
Reprod Fertil Dev ; 27(4): 684-92, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25730413

RESUMO

Periconceptional environment may influence embryo development, ultimately affecting adult health. Here, we review the rodent model of maternal low-protein diet specifically during the preimplantation period (Emb-LPD) with normal nutrition during subsequent gestation and postnatally. This model, studied mainly in the mouse, leads to cardiovascular, metabolic and behavioural disease in adult offspring, with females more susceptible. We evaluate the sequence of events from diet administration that may lead to adult disease. Emb-LPD changes maternal serum and/or uterine fluid metabolite composition, notably with reduced insulin and branched-chain amino acids. This is sensed by blastocysts through reduced mammalian target of rapamycin complex 1 signalling. Embryos respond by permanently changing the pattern of development of their extra-embryonic lineages, trophectoderm and primitive endoderm, to enhance maternal nutrient retrieval during subsequent gestation. These compensatory changes include stimulation in proliferation, endocytosis and cellular motility, and epigenetic mechanisms underlying them are being identified. Collectively, these responses act to protect fetal growth and likely contribute to offspring competitive fitness. However, the resulting growth adversely affects long-term health because perinatal weight positively correlates with adult disease risk. We argue that periconception environmental responses reflect developmental plasticity and 'decisions' made by embryos to optimise their own development, but with lasting consequences.


Assuntos
Dieta com Restrição de Proteínas , Proteínas Alimentares , Desenvolvimento Embrionário/fisiologia , Desenvolvimento Fetal/fisiologia , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Animais , Blastocisto/metabolismo , Feminino , Camundongos , Gravidez
19.
Am J Physiol Heart Circ Physiol ; 306(10): H1444-52, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24658019

RESUMO

Although the association between maternal periconceptional diet and adult offspring health is well characterised, our understanding of the impact of paternal nutrition at the time of conception on offspring phenotype remains poorly defined. Therefore, we determined the effect of a paternal preconception low protein diet (LPD) on adult offspring cardiovascular and metabolic health in mice. Male C57BL/6 mice were fed either normal protein diet (NPD; 18% casein) or LPD (9% casein) for 7 wk before mating. At birth, a reduced male-to-female ratio (P = 0.03) and increased male offspring weight (P = 0.009) were observed in litters from LPD compared with NPD stud males with no differences in mean litter size. LPD offspring were heavier than NPD offspring at 2 and 3 wk of age (P < 0.02). However, no subsequent differences in body weight were observed. Adult male offspring derived from LPD studs developed relative hypotension (decreased by 9.2 mmHg) and elevated heart rate (P < 0.05), whereas both male and female offspring displayed vascular dysfunction and impaired glucose tolerance relative to NPD offspring. At cull (24 wk), LPD males had elevated adiposity (P = 0.04), reduced heart-to-body weight ratio (P = 0.04), and elevated circulating TNF-α levels (P = 0.015) compared with NPD males. Transcript expression in offspring heart and liver tissue was reduced for genes involved in calcium signaling (Adcy, Plcb, Prkcb) and metabolism (Fto) in LPD offspring (P < 0.03). These novel data reveal the impact of suboptimal paternal nutrition on adult offspring cardiovascular and metabolic homeostasis, and provide some insight into the underlying regulatory mechanisms.


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Filho de Pais com Deficiência , Dieta com Restrição de Proteínas/efeitos adversos , Desnutrição/complicações , Metabolismo/fisiologia , Fatores Sexuais , Animais , Feminino , Intolerância à Glucose/epidemiologia , Hipotensão/epidemiologia , Incidência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Fenômenos Fisiológicos da Nutrição/fisiologia , Razão de Masculinidade
20.
Eur J Nutr ; 53(7): 1523-31, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24481689

RESUMO

PURPOSE: Prenatal undernutrition followed by postweaning feeding of a high-fat diet results in obesity in the adult offspring. In this study, we investigated whether diet-induced thermogenesis is altered as a result of such nutritional mismatch. METHODS: Female MF-1 mice were fed a normal protein (NP, 18% casein) or a protein-restricted (PR, 9% casein) diet throughout pregnancy and lactation. After weaning, male offspring of both groups were fed either a high-fat diet (HF; 45% kcal fat) or standard chow (C, 7% kcal fat) to generate the NP/C, NP/HF, PR/C and PR/HF adult offspring groups (n = 7-11 per group). RESULTS: PR/C and NP/C offspring have similar body weights at 30 weeks of age. Postweaning HF feeding resulted in significantly heavier NP/HF offspring (P < 0.01), but not in PR/HF offspring, compared with their chow-fed counterparts. However, the PR/HF offspring exhibited greater adiposity (P < 0.01) v the NP/HF group. The NP/HF offspring had increased energy expenditure and increased mRNA expression of uncoupling protein-1 and ß-3 adrenergic receptor in the interscapular brown adipose tissue (iBAT) compared with the NP/C mice (both at P < 0.01). No such differences in energy expenditure and iBAT gene expression were observed between the PR/HF and PR/C offspring. CONCLUSIONS: These data suggest that a mismatch between maternal diet during pregnancy and lactation, and the postweaning diet of the offspring, can attenuate diet-induced thermogenesis in the iBAT, resulting in the development of obesity in adulthood.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Dieta com Restrição de Proteínas/efeitos adversos , Fenômenos Fisiológicos da Nutrição Materna , Termogênese/fisiologia , Adiposidade , Animais , Glicemia/metabolismo , Pressão Sanguínea , Peso Corporal , Calorimetria Indireta , Gorduras na Dieta , Proteínas Alimentares/administração & dosagem , Ingestão de Energia , Metabolismo Energético , Feminino , Canais Iônicos/genética , Canais Iônicos/metabolismo , Lactação , Metabolismo dos Lipídeos , Masculino , Camundongos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Tamanho do Órgão , Gravidez , Receptores Adrenérgicos beta 3/genética , Receptores Adrenérgicos beta 3/metabolismo , Proteína Desacopladora 1 , Desmame
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA