Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Reproduction ; 165(4): 347-362, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36633493

ABSTRACT

In brief: Maternal obesity can impair metabolism in the embryo and the resulting offspring. This study shows that metabolic disruptions through α-ketoglutarate may link altered metabolism with epigenetic changes in embryos. Abstract: Maternal obesity can impair offspring metabolic health; however, the precise mechanism underpinning programming is unknown. Ten-Eleven translocase (TET) enzymes demethylate DNA using the TCA cycle intermediary α-ketoglutarate and may be involved in programming offspring health. Whether TETs are disrupted by maternal obesity is unknown. Five to six week-old C57Bl/6 female mice were fed a control diet (CD; 6% fat, n = 175) or a high-fat diet (HFD; 21% fat, n = 158) for 6 weeks. After superovulation, oocytes were collected for metabolic assessment, or females were mated and zygotes were cultured for embryo development, fetal growth, and assessment of global DNA methylation (5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxycytosine (5caC)) in the two-cell embryo. Zygotes collected from superovulated CBAF1 females were cultured in media containing α-ketoglutarate (0, 1.4, 3.5, or 14.0 mM) or with 2-hydroxyglutarate (2HG) (0 or 20 mM), a competitive inhibitor of α-ketoglutarate, with methylation and blastocyst differentiation assessed. After HFD, oocytes showed increased pyruvate oxidation and intracellular ROS, with no changes in Tet3 expression, while two-cell embryo global 5hmC DNA methylation was reduced and 5fC increased. Embryos cultured with 1.4 mM α-ketoglutarate had decreased two-cell 5mC, while 14.0 mM α-ketoglutarate increased the 5hmC:5mC ratio. In contrast, supplementation with 20 mM 2HG increased 5mC and decreased 5fC:5mC and 5caC:5mC ratios. α-ketoglutarate up to 3.5 mM did not alter embryo development, while culturing in 14.0 mM α-ketoglutarate blocked development at the two-cell. Culture with 2HG delayed embryo development past the four-cell and decreased blastocyst total cell number. In conclusion, disruptions in metabolic intermediates in the preimplantation embryo may provide a link between maternal obesity and programming offspring for ill health.


Subject(s)
DNA Methylation , Obesity, Maternal , Animals , Female , Humans , Mice , Pregnancy , 5-Methylcytosine/metabolism , Cytosine/metabolism , Diet, High-Fat , Ketoglutaric Acids/pharmacology , Obesity, Maternal/metabolism , Zygote/metabolism
2.
Endocrinology ; 162(10)2021 10 01.
Article in English | MEDLINE | ID: mdl-34170298

ABSTRACT

Paternal experiences and exposures before conception can influence fetal development and offspring phenotype. The composition of seminal plasma contributes to paternal programming effects through modulating the female reproductive tract immune response after mating. To investigate whether paternal obesity affects seminal plasma immune-regulatory activity, C57Bl/6 male mice were fed an obesogenic high-fat diet (HFD) or control diet (CD) for 14 weeks. Although HFD consumption caused only minor changes to parameters of sperm quality, the volume of seminal vesicle fluid secretions was increased by 65%, and the concentrations and total content of immune-regulatory TGF-ß isoforms were decreased by 75% to 80% and 43% to 55%, respectively. Mating with BALB/c females revealed differences in the strength and properties of the postmating immune response elicited. Transcriptional analysis showed >300 inflammatory genes were similarly regulated in the uterine endometrium by mating independently of paternal diet, and 13 were dysregulated by HFD-fed compared with CD-fed males. Seminal vesicle fluid factors reduced in HFD-fed males, including TGF-ß1, IL-10, and TNF, were among the predicted upstream regulators of differentially regulated genes. Additionally, the T-cell response induced by mating with CD-fed males was blunted after mating with HFD-fed males, with 27% fewer CD4+ T cells, 26% fewer FOXP3+CD4+ regulatory T cells (Treg) cells, and 19% fewer CTLA4+ Treg cells, particularly within the NRP1+ thymic Treg cell population. These findings demonstrate that an obesogenic HFD alters the composition of seminal vesicle fluid and impairs seminal plasma capacity to elicit a favorable pro-tolerogenic immune response in females at conception.


Subject(s)
Plasma/metabolism , Semen/metabolism , Adiposity , Animals , Body Composition , Cytokines/metabolism , Diet, High-Fat , Female , Lymphocyte Subsets , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Phenotype , Pregnancy , Pregnancy, Animal , Protein Isoforms , Reproduction , Semen/physiology , Spermatozoa/physiology , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/metabolism , Uterus/pathology
3.
Nutrients ; 11(9)2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31547309

ABSTRACT

Male obesity, which often co-presents with micronutrient deficiencies, is associated with sub-fertility. Here we investigate whether short-term dietary supplementation of micronutrients (zinc, selenium, lycopene, vitamins E and C, folic acid, and green tea extract) to obese mice for 12 days (designed to span the epididymal transit) could improve sperm quality and fetal outcomes. Five-week-old C57BL6 males were fed a control diet (CD, n = 24) or high fat diet (HFD, n = 24) for 10 weeks before allocation to the 12-day intervention of maintaining their original diets (CD, n = 12, HFD n = 12) or with micronutrient supplementation (CD + S, n = 12, HFD + S, n = 12). Measures of sperm quality (motility, morphology, capacitation, binding), sperm oxidative stress (DCFDA, MSR, and 8OHdG), early embryo development (2-cell cleavage, 8OHdG), and fetal outcomes were assessed. HFD + S males had reduced sperm intracellular reactive oxygen species (ROS) concentrations and 8OHdG lesions, which resulted in reduced 8OHdG lesions in the male pronucleus, increased 2-cell cleavage rates, and partial restoration of fetal weight similar to controls. Sub-fertility associated with male obesity may be restored with very short-term micronutrient supplementation that targets the timing of the transit of sperm through the epididymis, which is the developmental window where sperm are the most susceptible to oxidative damage.


Subject(s)
Dietary Supplements , Infertility, Male/metabolism , Micronutrients/pharmacology , Obesity/metabolism , Oxidative Stress/drug effects , Animals , Diet, High-Fat , Disease Models, Animal , Embryonic Development/drug effects , Infertility, Male/etiology , Infertility, Male/therapy , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/complications , Obesity/therapy , Sperm Motility/drug effects , Spermatozoa/drug effects
4.
Reprod Biomed Soc Online ; 9: 48-63, 2019 Dec.
Article in English | MEDLINE | ID: mdl-32021914

ABSTRACT

This study examined the nature and accuracy of information available across online platforms for couples trying to conceive. A consumer simulation-based investigation of English websites and social media (Facebook, Twitter, Instagram) was undertaken using common search terms identified in a pilot study. Claims about fertility and pregnancy health were then extracted from the results and analysed thematically. The accuracy of each claim was assessed independently by six fertility and conception experts, rated on a scale of 1 (not factual) to 4 (highly factual), with scores collated to produce a median rating. Claims with a median score < 3 were classified as inaccurate. The use of the terms 'trying to conceive' and '#TTC' were common identifiers on online platforms. Claims were extracted predominantly from websites (n = 89) rather than social media, with Twitter and Instagram comprising commercial elements and Facebook focused on community-based support. Thematic analysis revealed three major themes among the claims across all platforms: conception behaviour and monitoring, lifestyle and exposures, and medical. Fact-checking by the experts revealed that 40% of the information assessed was inaccurate, and that inaccuracies were more likely to be present in the conception behaviour and monitoring advice, the topics most amenable to modification. Since online information is a readily accessible and commonly utilized resource, there is opportunity for improved dissemination of evidence-based material to reach interested couples. Further cross-disciplinary and consumer-based research, such as a user survey, is required to understand how best to provide the 'trying to conceive' community with accurate information.

5.
J Endocrinol ; 234(2): F1-F6, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28500085

ABSTRACT

Animal and human studies demonstrate that acquired paternal traits can impair both a male's fertility and the health of his offspring, including advanced age, smoking, stress, trauma, under-nutrition, infection, toxin exposure, and obesity. Many of these factors lead to similar changes to neurological, behavioural, and/or metabolic functioning in offspring. The molecular mechanisms that both respond to the paternal environment and act to transmit traits to offspring are beginning to emerge. This review focuses on three vices of men (alcohol consumption, overweight/obesity, and tobacco smoking) that damage fertility and pose risks to offspring health. These vices are not only the three most prevalent but are also leading risk factors for death and disability adjusted life years (DALYs) worldwide. Moreover, given that these vices are predominantly self-inflicted, interventions aimed at mitigating their consequences are readily identified.


Subject(s)
Alcohol Drinking/adverse effects , Infertility, Male/etiology , Overweight/complications , Paternal Behavior , Smoking/adverse effects , Animals , Epigenesis, Genetic , Humans , Male
6.
Mol Reprod Dev ; 84(9): 914-925, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28452160

ABSTRACT

The reproductive tract environment at conception programs the developmental trajectory of the embryo, sets the course of pregnancy, and impacts offspring phenotype and health. Despite the fundamental importance of this stage of reproduction, the rate-limiting regulatory mechanisms operating locally to control fertility and fecundity are incompletely understood. Emerging studies highlight roles for microRNAs (miRNAs) in regulating reproductive and developmental processes and in modulating the quality and strength of the female immune response. Since endometrial receptivity and robust placentation require specific adaptation of the immune response, we hypothesize that miRNAs participate in establishing pregnancy through effects on key gene networks in immune cells. Our recent studies investigated miRNAs that are induced in the peri-conception environment, focusing on miRNAs that have immune-regulatory roles-particularly miR-223, miR-155, and miR-146a. Genetic mouse models deficient in individual miRNAs are proving informative in defining roles for these miRNAs in the generation and stabilization of regulatory T cells (Treg cells) that confer adaptive immune tolerance. Overlapping and redundant functions between miRNAs that target multiple genes, combined with multiple miRNAs targeting individual genes, indicate complex and sensitive regulatory networks. Although to date most data on miRNA regulation of reproductive events are from mice, conserved functions of miRNAs across species imply similar biological pathways operate in all mammals. Understanding the regulation and roles of miRNAs in the peri-conception immune response will advance our knowledge of how environmental determinants act at conception, and could have practical applications for animal breeding as well as human fertility.


Subject(s)
Fertility/immunology , Immune Tolerance , MicroRNAs/immunology , Placentation/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Female , Humans , Mice , Pregnancy
7.
Nutrients ; 9(2)2017 Feb 09.
Article in English | MEDLINE | ID: mdl-28208792

ABSTRACT

Paternal obesity programs metabolic syndrome in offspring. Low-impact exercise in obese  males improves the metabolic health of female offspring, however whether this occurred in male  offspring remained unknown. C57BL/6NHsd (Harlan) mice were fed a control diet (CD; 6% fat, n =  7) or a high-fat diet (HFD; 21% fat, n = 16) for 18 weeks. After 9 weeks, HFD-fed mice either remained  sedentary (HH, n = 8) or undertook low-moderate exercise (HE, n = 8) for another 9 weeks. Male  offspring were assessed for glucose/insulin tolerance, body composition, plasma lipids, pancreatic  islet cell morphology and microRNA expression. Founder HH induced glucose intolerance, insulin  insensitivity, and hyperlipidaemia in male offspring (p < 0.05). Metabolic health was fully restored  in male offspring by founder exercise to control levels. Founder HH reduced pancreatic ß-cell area  and islet cell size in male offspring, and altered the expression of 13 pancreatic microRNAs (p <  0.05). Founder HE led to partial restoration of pancreatic islet cell morphology and the expression  of two pancreatic microRNAs (let7d-5p, 194-5p) in male offspring. Founder HE reduced male  offspring adiposity, increased muscle mass, reduced plasma free fatty acids (FFAs), and further  altered pancreatic microRNAs (35 vs. HH; 32 vs. CD) (p < 0.05). Low-impact exercise in obese fathers  prior to conception, without dietary change, may be a viable intervention strategy to reduce the illeffects of obesity-induced paternal programming in male offspring.


Subject(s)
Islets of Langerhans/cytology , MicroRNAs/metabolism , Obesity/physiopathology , Obesity/therapy , Physical Conditioning, Animal/methods , Animals , Blood Glucose/physiology , Cellular Reprogramming , Cellular Reprogramming Techniques/methods , Diet, High-Fat/adverse effects , Fathers , Fatty Acids, Nonesterified/blood , Female , Insulin/physiology , Islets of Langerhans/physiopathology , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Paternal Inheritance
8.
PLoS One ; 11(11): e0166076, 2016.
Article in English | MEDLINE | ID: mdl-27814400

ABSTRACT

The prevalence of obesity is increasing worldwide and has tripled in men of reproductive age since the 1970s. Concerningly, obesity is not only comorbid with other chronic diseases, but there is mounting evidence that it increases the non-communicable disease load in their children (eg mortality, obesity, autism). Animal studies have demonstrated that paternal obesity increases the risk of metabolic (eg glucose metabolism defects, obesity) and reproductive disorders in offspring. Epigenetic changes within sperm are clear mechanistic candidates that are associated with both changes to the father's environment and offspring phenotype. Specifically there is emerging evidence that a father's sperm microRNA content both responds to paternal environmental cues and alters the gene expression profile and subsequent development of the early embryo. We used a mouse model of high fat diet (HFD) induced obesity to investigate whether male obesity could modulate sperm microRNA content. We also investigated whether this alteration to a father's sperm microRNA content lead to a similar change in the sperm of male offspring. Our investigations were initially guided by a Taqman PCR array, which indicated the differential abundance of 28 sperm borne microRNAs in HFD mice. qPCR confirmation in a much larger cohort of founder males demonstrated that 13 of these microRNAs were differentially abundant (11 up-regulated; 2 down-regulated) due to HFD feeding. Despite metabolic and reproductive phenotypes also being observed in grand-offspring fathered via the male offspring lineage, there was no evidence that any of the 13 microRNAs were also dysregulated in male offspring sperm. This was presumably due to the variation seen within both groups of offspring and suggests other mechanisms might act between offspring and grand-offspring. Thus 13 sperm borne microRNAs are modulated by a father's HFD and the presumed transfer of this altered microRNA payload to the embryo at fertilisation potentially acts to alter the embryonic molecular makeup post-fertilisation, altering its growth trajectory, ultimately affecting adult offspring phenotype and may contribute to paternal programming.


Subject(s)
MicroRNAs/genetics , Obesity/genetics , Spermatozoa/metabolism , Adiposity/genetics , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Epigenesis, Genetic/genetics , Fathers , Fertilization/genetics , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Reproduction/genetics , Transcriptome/genetics
9.
Sci Rep ; 6: 27010, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27255552

ABSTRACT

There is an ever increasing body of evidence that demonstrates that paternal over-nutrition prior to conception programs impaired metabolic health in offspring. Here we examined whether paternal under-nutrition can also program impaired health in offspring and if any detrimental health outcomes in offspring could be prevented by micronutrient supplementation (vitamins and antioxidants). We discovered that restricting the food intake of male rodents reduced their body weight, fertility, increased sperm oxidative DNA lesions and reduced global sperm methylation. Under-nourished males then sired offspring with reduced postnatal weight and growth but somewhat paradoxically increased adiposity and dyslipidaemia, despite being fed standard chow. Paternal vitamin/antioxidant food fortification during under-nutrition not only normalised founder oxidative sperm DNA lesions but also prevented early growth restriction, fat accumulation and dyslipidaemia in offspring. This demonstrates that paternal under-nutrition reduces postnatal growth but increases the risk of obesity and metabolic disease in the next generation and that micronutrient supplementation during this period of under-nutrition is capable of restoring offspring metabolic health.


Subject(s)
Malnutrition/genetics , Metabolic Syndrome/genetics , Adiposity , Animals , Antioxidants/administration & dosage , Body Composition , Embryonic Development , Female , Food, Fortified , Founder Effect , Infertility, Male/etiology , Infertility, Male/genetics , Insulin/blood , Leptin/blood , Lipids/blood , Male , Malnutrition/complications , Metabolic Syndrome/blood , Metabolic Syndrome/etiology , Mice, Inbred C57BL , Paternal Inheritance , Reactive Oxygen Species/metabolism , Sperm Count , Sperm Motility
10.
Am J Reprod Immunol ; 75(3): 272-80, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26804209

ABSTRACT

To support embryo implantation, the female reproductive tract must provide a tolerogenic immune environment. Seminal fluid contact at conception contributes to activating the endometrial gene expression and immune cell changes required for robust implantation, influencing not only the quality of the ensuing pregnancy but also the health of offspring. miRNAs are small non-coding RNAs that play important regulatory roles in biological processes, including regulation of the immune environment. miRNAs are known to contribute to gene regulation in pregnancy and are altered in pregnancy pathologies. Recent studies indicate that miRNAs participate in establishing immune tolerance at conception, and may contribute to the regulatory effects of seminal fluid in generating tolerogenic dendritic cells and T regulatory cells. This review highlights those miRNAs implicated in programming immune cells that are critical during the peri-conception period and explores how seminal fluid may regulate female tract miRNA expression following coitus.


Subject(s)
Endometrium/immunology , Gene Expression Regulation/immunology , Immune Tolerance/physiology , MicroRNAs/immunology , Pregnancy/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Coitus/physiology , Female , Humans , Male , Semen/immunology
11.
J Assist Reprod Genet ; 32(5): 725-35, 2015 May.
Article in English | MEDLINE | ID: mdl-25854657

ABSTRACT

PURPOSE: To investigate the impacts that a paternal high fat diet (HFD) has on embryology, ovarian/cumulus cell gene expression and COC metabolism from female offspring, using a mouse model. METHODS: Founder male mice were either fed a control diet (CD) or a HFD for 12 weeks. The HFD induced obesity but not diabetes, and founder males were then mated to normal weight CD fed female mice. Female offspring were maintained on a CD, super-ovulated, mated and the resultant zygotes were cultured to the blastocyst stage for embryo morphology, blastocyst cell number and apoptosis assessment. Ovaries and cumulus cells from offspring were collected for gene expression analysis of selected genes that maintain chromatin remodeling and endoplasmic reticulum (ER), metabolic and inflammatory homeostasis. Cumulus/oocyte complexes were also investigated for glucose uptake and lipid accumulation. RESULTS: Female offspring sired by obese fathers produced embryos with delayed development and impaired quality, displayed increases in ovarian expression of Glut1, Glut3 and Glut4, and an increase in cumulus cell expression of Glut4. Interestingly their COCs did take up more glucose, but did accumulate more lipid. CONCLUSIONS: A paternal HFD is associated with subfertility in female offspring despite the offspring being fed a CD and this subfertility is concomitant with ovarian/cumulus cell molecular alterations and increased lipid accumulation.


Subject(s)
Blastocyst/pathology , Cumulus Cells/metabolism , Diet, High-Fat/adverse effects , Obesity/physiopathology , Oocytes/metabolism , Ovary/metabolism , Animals , Blastocyst/metabolism , Cumulus Cells/pathology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Oocytes/pathology , Ovary/pathology , Pregnancy , Real-Time Polymerase Chain Reaction , Semen/chemistry
12.
Physiol Rep ; 3(3)2015 Mar.
Article in English | MEDLINE | ID: mdl-25804263

ABSTRACT

Obesity and related comorbidities are becoming increasingly prevalent globally. In mice preconception paternal exposure to a high fat diet (HFD) impairs the metabolic and reproductive health of male offspring, despite their control diet (CD) consumption. However, offspring share lifestyle, including diet, with parents. We assessed if male offspring from HFD fathers have a heightened susceptibility to HFD-induced metabolic and reproductive derangements. This 2 × 2 design saw founder males (F0) and their offspring (F1) fed either a HFD or a nutritionally matched CD. Regardless of paternal diet, HFD fed male offspring had greater total body weight and adiposity. Offspring sired by a HFD male and fed a HFD were the heaviest, had the greatest adiposity and had the greatest concentration of serum cholesterol, triglyceride, HDL, and NEFA compared with CD sired/fed littermates. A synergistic increase in serum insulin was unmasked by both father/son HFD consumption, concomitant with increased sera glucose. Either a paternal or offspring HFD was associated with similar reductions to offspring sperm motility. Whereas sperm ROS concentrations and sperm-oocyte binding saw detrimental effects of both F0 HFD and F1 HFD with an interaction evident between both, culminating in the most impaired sperm parameters in this group. This indicates that metabolic and fertility disturbances in male offspring sired by HFD fathers are exacerbated by a "second-hit" of exposure to the same obesogenic environment postnatally. If translatable to human health, this suggests that adverse reproductive and metabolic outcomes may be amplified across generations through a shared calorie dense diet, relevant to the current worldwide obesity epidemic.

13.
Biol Reprod ; 92(5): 124, 2015 May.
Article in English | MEDLINE | ID: mdl-25715796

ABSTRACT

The periconceptual environment represents a critical window for programming fetal growth trajectories and susceptibility to disease; however, the underlying mechanism responsible for programming remains elusive. This study demonstrates a causal link between reduction of precompaction embryonic mitochondrial function and perturbed offspring growth trajectories and subsequent metabolic dysfunction. Incubation of embryos with carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP), which uncouples mitochondrial oxidative phosphorylation, significantly reduced mitochondrial membrane potential and ATP production in 8-cell embryos and the number of inner cell mass cells within blastocysts; however, blastocyst development was unchanged. This perturbed embryonic mitochondrial function was concomitant with reduced birth weight in female offspring following embryo transfer, which persisted until weaning. FCCP-treated females also exhibited increased adiposity at 4 wk, increased adiposity gain between 4 and 14 wk, glucose intolerance at 8 wk, and insulin resistance at 14 wk. Although FCCP-treated males also exhibited reduced glucose tolerance, but their insulin sensitivity and adiposity gain between 4 and 14 wk was unchanged. To our knowledge, this is one of the first studies to demonstrate that reducing mitochondrial function and, thus, decreasing ATP output in the precompacting embryo can influence offspring phenotype. This is of great significance as a large proportion of patients requiring assisted reproductive technologies are of advanced maternal age or have a high body mass index, both of which have been independently linked with perturbed early embryonic mitochondrial function.


Subject(s)
Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/toxicity , Cleavage Stage, Ovum/drug effects , Mitochondria/drug effects , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Adiposity/drug effects , Animals , Birth Weight , Embryo Culture Techniques , Embryo Transfer , Embryonic Development/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Litter Size , Male , Metalloproteases/genetics , Metalloproteases/metabolism , Mice , Mitochondria/metabolism , Pregnancy , Prenatal Exposure Delayed Effects
14.
Am J Physiol Endocrinol Metab ; 308(9): E805-21, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25690453

ABSTRACT

Obesity and type 2 diabetes are increasingly prevalent across all demographics. Paternal obesity in humans and rodents can program obesity and impair insulin sensitivity in female offspring. It remains to be determined whether these perturbed offspring phenotypes can be improved through targeted lifestyle interventions in the obese father. Using a mouse model, we demonstrate that diet or exercise interventions for 8 wk (2 rounds of spermatogenesis) in obese founder males restores insulin sensitivity and normalized adiposity in female offspring. Founder diet and/or exercise also normalizes abundance of X-linked sperm microRNAs that target genes regulating cell cycle and apoptosis, pathways central to oocyte and early embryogenesis. Additionally, obesity-associated comorbidities, including inflammation, glucose intolerance, stress, and hypercholesterolemia, were good predictors for sperm microRNA abundance and offspring phenotypes. Interventions aimed at improving paternal metabolic health during specific windows prior to conception can partially normalize aberrant epigenetic signals in sperm and improve the metabolic health of female offspring.


Subject(s)
Animal Nutritional Physiological Phenomena , Fathers , Metabolic Syndrome/prevention & control , MicroRNAs/genetics , Obesity , Physical Conditioning, Animal/physiology , Spermatozoa/metabolism , Animals , Diet , Female , Fertilization/physiology , Infertility, Male/genetics , Infertility, Male/prevention & control , Male , Metabolic Syndrome/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/genetics , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/metabolism , Transcriptome
15.
Ann Nutr Metab ; 64(3-4): 231-8, 2014.
Article in English | MEDLINE | ID: mdl-25300265

ABSTRACT

BACKGROUND: The global rates of male overweight/obesity are rising, approaching 70% of the total adult population in Western nations. Overweight/obesity increases the risk of chronic diseases; however, there is increasing awareness that male obesity negatively impacts fertility, subsequent pregnancy, and the offspring health burden. Developmental programming is well defined in mothers; however, it is becoming increasingly evident that developmental programming can be paternally initiated and mediated through paternal obesity. KEY MESSAGES: Both human and rodent models have established that paternal obesity impairs sex hormones, basic sperm function, and molecular composition. This results in perturbed embryo development and health and an increased subsequent offspring disease burden in both sexes. The reversibility of obesity-induced parental programming has only recently received attention. Promising results in animal models utilizing diet and exercise interventions have shown improvements in sperm function and molecular composition, resulting in restorations of both embryo and fetal health and subsequent male offspring fertility. The direct mode for paternal inheritance is likely mediated via spermatozoa. We propose two main theories for the origin of male obesity-induced paternal programming: (1) accumulation of sperm DNA damage resulting in de novo mutations in the embryo and (2) changes in sperm epigenetic marks (microRNA, methylation, or acetylation) altering the access, transcription, and translation of paternally derived genes during early embryogenesis. CONCLUSIONS: Paternal overweight/obesity induces paternal programming of offspring phenotypes likely mediated through genetic and epigenetic changes in spermatozoa. These programmed changes to offspring health appear to be partially restored via diet/exercise interventions in obese fathers preconception, which have been shown to improve aspects of sperm DNA integrity. However, the majority of data surrounding paternal obesity and offspring phenotypes have come from rodent models; therefore, we contend that it will be increasingly important to study population-based data to determine the likely mode of inheritance in humans.


Subject(s)
Diet/adverse effects , Family Health , Health Promotion , Motor Activity , Obesity/etiology , Obesity/prevention & control , Patient Compliance , Animals , Child , Child Nutritional Physiological Phenomena , DNA Damage , Epigenesis, Genetic , Fathers , Female , Fetal Development , Humans , Life Style , Male , Nutrition Policy , Obesity/epidemiology , Obesity/pathology , Paternal Behavior , Pregnancy , Spermatozoa/pathology
16.
PLoS One ; 9(7): e100832, 2014.
Article in English | MEDLINE | ID: mdl-25006800

ABSTRACT

Paternal health cues are able to program the health of the next generation however the mechanism for this transmission is unknown. Reactive oxygen species (ROS) are increased in many paternal pathologies, some of which program offspring health, and are known to induce DNA damage and alter the methylation pattern of chromatin. We therefore investigated whether a chemically induced increase of ROS in sperm impairs embryo, pregnancy and offspring health. Mouse sperm was exposed to 1500 µM of hydrogen peroxide (H2O2), which induced oxidative damage, however did not affect sperm motility or the ability to bind and fertilize an oocyte. Sperm treated with H2O2 delayed on-time development of subsequent embryos, decreased the ratio of inner cell mass cells (ICM) in the resulting blastocyst and reduced implantation rates. Crown-rump length at day 18 of gestation was also reduced in offspring produced by H2O2 treated sperm. Female offspring from H2O2 treated sperm were smaller, became glucose intolerant and accumulated increased levels of adipose tissue compared to control female offspring. Interestingly male offspring phenotype was less severe with increases in fat depots only seen at 4 weeks of age, which was restored to that of control offspring later in life, demonstrating sex-specific impacts on offspring. This study implicates elevated sperm ROS concentrations, which are common to many paternal health pathologies, as a mediator of programming offspring for metabolic syndrome and obesity.


Subject(s)
Adiposity , Embryonic Development , Glucose/metabolism , Oxidative Stress , Spermatozoa/metabolism , Animals , Blastocyst/cytology , Female , Fertilization , Fetal Development , Glucose Intolerance , Hydrogen Peroxide/pharmacology , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Pregnancy
17.
Fertil Steril ; 101(3): 865-73, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24424359

ABSTRACT

OBJECTIVE: To determine whether dietary and exercise regimes in obese males can provide a novel intervention window for improving the reproductive health of the next generation. DESIGN: Experimental animal study. SETTING: University research facilities. ANIMAL(S): C57BL6 male and female mice. INTERVENTION(S): Mice were fed a control diet (6% fat) or high-fat diet (21% fat) for 9 weeks. After the initial feeding, high-fat-diet males were allocated to diet and/or exercise interventions for a further 9 weeks. After intervention males were mated with females fed standard chow (4% fat) before and during pregnancy. MAIN OUTCOME MEASURE(S): F1 sperm motility, count, morphology, capacitation, mitochondrial function, and sperm binding and weight of reproductive organs. RESULT(S): Our primary finding was that diet intervention alone in founders improved offspring sperm motility and mitochondrial markers of sperm health (decreased reactive oxygen species and mitochondrial membrane potential), ultimately improving sperm binding. Sperm binding and capacitation was also improved in F1 males born to a combined diet and exercise intervention in founders. Founder sperm parameters and metabolic measures as a response to diet and/or exercise (i.e., lipid/glucose homeostasis, sperm count and morphology) correlated with offspring's sperm function, independent of founder treatment. This implicates paternal metabolic and reproductive status in predicting male offspring's reproductive function. CONCLUSION(S): This is the first study to show that improvements to both metabolic (lipids, glucose and insulin sensitivity) and reproductive function (sperm motility and morphology) in obese fathers via diet and exercise interventions can improve subsequent reproductive health in offspring.


Subject(s)
Adiposity/physiology , Diet, High-Fat/adverse effects , Obesity/metabolism , Prenatal Exposure Delayed Effects/metabolism , Reproduction/physiology , Sperm Motility/physiology , Animals , Blood Glucose/metabolism , Diet, High-Fat/methods , Disease Models, Animal , Fathers , Female , Insulin Resistance/physiology , Male , Mice , Obesity/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Random Allocation
18.
FASEB J ; 27(10): 4226-43, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23845863

ABSTRACT

Obesity is highly prevalent, and its incidence is increasing. The previous study showing a major effect of paternal obesity on metabolic health of offspring is confounded by comorbidity with diabetes. Therefore, we investigated the effect of diet-induced paternal obesity, in the absence of diabetes, on the metabolic health of two resultant generations and the molecular profiles of the testes and sperm. Founder (F0) male C57BL6 mice were fed either a high-fat diet (HFD) or a control diet (CD); n = 10/diet for a period of 10 wk. Testis expression of mRNA/microRNAs was analyzed by microarray and qPCR and sperm microRNA abundance by qPCR. Two subsequent generations were generated by mating F0 and then F1 mice to CD mice, and their metabolic health was investigated. All mice, other than F0 males, were maintained on a CD. HFD feeding induced paternal obesity with a 21% increase in adiposity, but not overt diabetes, and initiated intergenerational transmission of obesity and insulin resistance in two generations of offspring. This distinct phenotypic constellation is either partially or fully transmitted to both female and male F1 offspring and further transmitted through both parental lineages to the F2 generation, with a heightened effect on female F1 offspring (+67% in adiposity) and their F2 sons (+24% in adiposity). Founder male obesity altered the testes expression of 414 mRNAs by microarray and 11 microRNAs by qPCR, concomitant with alterations in sperm microRNA content and a 25% reduction in global methylation of germ cell DNA. Diet-induced paternal obesity modulates sperm microRNA content and germ cell methylation status, which are potential signals that program offspring health and initiate the transmission of obesity and impaired metabolic health to future generations. This study implicates paternal obesity in the transgenerational amplification of obesity and type 2 diabetes in humans.


Subject(s)
MicroRNAs/metabolism , Obesity/metabolism , Spermatozoa/metabolism , Testis/metabolism , Transcriptome/physiology , Animals , Energy Metabolism , Female , Gene Expression Regulation/physiology , Glucose Tolerance Test , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Obesity/genetics , Reactive Oxygen Species , Sex Factors
19.
Spermatogenesis ; 2(4): 253-263, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23248766

ABSTRACT

Male obesity in reproductive-age men has nearly tripled in the past 30 y and coincides with an increase in male infertility worldwide. There is now emerging evidence that male obesity impacts negatively on male reproductive potential not only reducing sperm quality, but in particular altering the physical and molecular structure of germ cells in the testes and ultimately mature sperm. Recent data has shown that male obesity also impairs offspring metabolic and reproductive health suggesting that paternal health cues are transmitted to the next generation with the mediator mostly likely occurring via the sperm. Interestingly the molecular profile of germ cells in the testes and sperm from obese males is altered with changes to epigenetic modifiers. The increasing prevalence of male obesity calls for better public health awareness at the time of conception, with a better understanding of the molecular mechanism involved during spermatogenesis required along with the potential of interventions in reversing these deleterious effects. This review will focus on how male obesity affects fertility and sperm quality with a focus on proposed mechanisms and the potential reversibility of these adverse effects.

20.
Reprod Fertil Dev ; 23(7): 929-39, 2011.
Article in English | MEDLINE | ID: mdl-21871212

ABSTRACT

Male obesity is associated with reduced sperm function and increased incidence of sperm DNA damage; however, the underlying molecular mechanisms have not yet been identified. Mammalian SIRT6 protein is involved in caloric-dependant DNA damage repair in other tissue types, yet a possible role for SIRT6 in male obesity and subfertility has not been investigated previously. To assess SIRT6 levels and activity in the testes, male mice (n=12 per diet) were fed either a control diet (CD; 6% fat) or a high-fat diet (HFD; 21% fat) for 16 weeks before the collection of testes and spermatozoa. SIRT6 protein was localised to the nucleus of transitional spermatids and the acrosome of mature spermatozoa, with levels significantly decreased in HFD-fed male mice (P<0.05). This decrease in SIRT6 protein was associated with transitional spermatids having increased levels of acetylated H3K9 in the nucleus (P<0.01) and increased DNA damage (P<0.001). We propose a role for SIRT6 in spermiogenesis and potentially protamination processes, which are known to be compromised by male obesity.


Subject(s)
Obesity/metabolism , Sirtuins/metabolism , Spermatogenesis , Spermatozoa/metabolism , Testis/metabolism , Acetylation , Acrosome/metabolism , Acrosome/ultrastructure , Animals , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , DNA Damage , Dietary Fats/adverse effects , Down-Regulation , Epididymis/metabolism , Epididymis/ultrastructure , Infertility, Male/etiology , Male , Mice , Mice, Inbred C57BL , Obesity/pathology , Obesity/physiopathology , Protein Transport , RNA, Messenger/metabolism , Random Allocation , Sirtuins/genetics , Sperm Capacitation , Spermatids/metabolism , Spermatids/ultrastructure , Spermatozoa/ultrastructure , Testis/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...