Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
2.
Nat Immunol ; 18(11): 1261-1269, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28945244

ABSTRACT

The Zika virus (ZIKV) epidemic has resulted in congenital abnormalities in fetuses and neonates. Although some cross-reactive dengue virus (DENV)-specific antibodies can enhance ZIKV infection in mice, those recognizing the DENV E-dimer epitope (EDE) can neutralize ZIKV infection in cell culture. We evaluated the therapeutic activity of human monoclonal antibodies to DENV EDE for their ability to control ZIKV infection in the brains, testes, placentas, and fetuses of mice. A single dose of the EDE1-B10 antibody given 3 d after ZIKV infection protected against lethality, reduced ZIKV levels in brains and testes, and preserved sperm counts. In pregnant mice, wild-type or engineered LALA variants of EDE1-B10, which cannot engage Fcg receptors, diminished ZIKV burden in maternal and fetal tissues, and protected against fetal demise. Because neutralizing antibodies to EDE have therapeutic potential against ZIKV, in addition to their established inhibitory effects against DENV, it may be possible to develop therapies that control disease caused by both viruses.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Dengue Virus/immunology , Epitopes/immunology , Viral Envelope Proteins/immunology , Zika Virus Infection/immunology , Animals , Brain/immunology , Brain/virology , Chlorocebus aethiops , Cross Reactions/immunology , Dengue Virus/classification , Dengue Virus/metabolism , Female , Fetus/immunology , Fetus/virology , Host-Pathogen Interactions/immunology , Humans , Male , Mice , Neutralization Tests , Pregnancy , Protein Multimerization/immunology , Testis/immunology , Testis/virology , Vero Cells , Viral Envelope Proteins/chemistry , Viral Load/immunology , Zika Virus/immunology , Zika Virus/physiology , Zika Virus Infection/virology
3.
Nature ; 540(7633): 438-442, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27798603

ABSTRACT

Infection of pregnant women with Zika virus (ZIKV) can cause congenital malformations including microcephaly, which has focused global attention on this emerging pathogen. In addition to transmission by mosquitoes, ZIKV can be detected in the seminal fluid of affected males for extended periods of time and transmitted sexually. Here, using a mouse-adapted African ZIKV strain (Dakar 41519), we evaluated the consequences of infection in the male reproductive tract of mice. We observed persistence of ZIKV, but not the closely related dengue virus (DENV), in the testis and epididymis of male mice, and this was associated with tissue injury that caused diminished testosterone and inhibin B levels and oligospermia. ZIKV preferentially infected spermatogonia, primary spermatocytes and Sertoli cells in the testis, resulting in cell death and destruction of the seminiferous tubules. Less damage was caused by a contemporary Asian ZIKV strain (H/PF/2013), in part because this virus replicates less efficiently in mice. The extent to which these observations in mice translate to humans remains unclear, but longitudinal studies of sperm function and viability in ZIKV-infected humans seem warranted.


Subject(s)
Testis/pathology , Testis/virology , Zika Virus Infection/pathology , Zika Virus/pathogenicity , Animals , Cell Death , Dengue Virus/physiology , Epididymis/pathology , Epididymis/virology , Humans , Inhibins/metabolism , Male , Mice , Mice, Inbred C57BL , Oligospermia/pathology , Oligospermia/virology , Seminiferous Tubules/pathology , Seminiferous Tubules/virology , Sertoli Cells/virology , Spermatocytes/virology , Spermatogonia/virology , Testosterone/metabolism , Time Factors
4.
Trends Genet ; 33(6): 399-407, 2017 06.
Article in English | MEDLINE | ID: mdl-28438343

ABSTRACT

The current global obesity pandemic is clearly linked to both the increasing prevalence of, and preference for, foods high in calories, specifically fat and sucrose, and declining levels of daily physical activity. A less commonly discussed possible explanation is that risk of obesity begins in utero as a result of developmental plasticity during early life. This idea fits into the broader Developmental Origins of Health and Diseases (DOHAD) hypothesis, which holds that stressful in utero exposure manifests as disease in adulthood. In this review, we highlight several studies that have revealed the role of epigenetics in multigenerational transmission of developmentally programmed obesity and associated cardiometabolic disease.


Subject(s)
Cardiovascular Diseases/genetics , Epigenesis, Genetic , Metabolic Diseases/genetics , Obesity/genetics , Cardiovascular Diseases/complications , Cardiovascular Diseases/metabolism , Humans , Metabolic Diseases/complications , Metabolic Diseases/metabolism , Obesity/complications , Obesity/metabolism
5.
Biol Reprod ; 102(4): 843-851, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31901086

ABSTRACT

Successful establishment of pregnancy depends on steroid hormone-driven cellular changes in the uterus during the peri-implantation period. To become receptive to embryo implantation, uterine endometrial stromal cells (ESCs) must transdifferentiate into decidual cells that secrete factors necessary for embryo survival and trophoblast invasion. Autophagy is a key homeostatic process vital for cellular homeostasis. Although the uterus undergoes major cellular changes during early pregnancy, the precise role of autophagy in uterine function is unknown. Here, we report that conditional knockout of the autophagy protein FIP200 in the reproductive tract of female mice results in reduced fecundity due to an implantation defect. In the absence of FIP200, aberrant progesterone signaling results in sustained uterine epithelial proliferation and failure of stromal cells to decidualize. Additionally, loss of FIP200 impairs decidualization of human ESCs. We conclude that the autophagy protein FIP200 plays a crucial role in uterine receptivity, decidualization, and fertility. These data establish autophagy as a major cellular pathway required for uterine receptivity and decidualization in both mice and human ESCs.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy/physiology , Progesterone/metabolism , Uterus/metabolism , Animals , Autophagy-Related Proteins/genetics , Decidua/metabolism , Embryo Implantation/physiology , Endometrium/metabolism , Female , Humans , Mice , Mice, Knockout , Signal Transduction/physiology , Stromal Cells/metabolism
6.
Hepatology ; 70(4): 1168-1184, 2019 10.
Article in English | MEDLINE | ID: mdl-31004524

ABSTRACT

The mechanisms by which alterations in intestinal bile acid (BA) metabolism improve systemic glucose tolerance and hepatic metabolic homeostasis are incompletely understood. We examined metabolic adaptations in mice with conditional intestinal deletion of the abetalipoproteinemia (ABL) gene microsomal triglyceride transfer protein (Mttp-IKO), which blocks chylomicron assembly and impairs intestinal lipid transport. Mttp-IKO mice exhibit improved hepatic glucose metabolism and augmented insulin signaling, without weight loss. These adaptations included decreased BA excretion, increased pool size, altered BA composition, and increased fibroblast growth factor 15 production. Mttp-IKO mice absorb fructose normally but are protected against dietary fructose-induced hepatic steatosis, without weight loss or changes in energy expenditure. In addition, Mttp-IKO mice exhibit altered cecal microbial communities, both at baseline and following fructose feeding, including increased abundance of Bacteroides and Lactobacillus genera. Transplantation of cecal microbiota from chow-fed Mttp-IKO mice into antibiotic-treated wild-type recipients conferred transmissible protection against fructose-induced hepatic steatosis in association with a bloom in Akkermansia and increased Clostridium XIVa genera, whose abundance was positively correlated with fecal coprostanol and total neutral sterol excretion in recipient mice. However, antibiotic-treated Mttp-IKO mice were still protected against fructose-induced hepatic steatosis, suggesting that changes in microbiota are not required for this phenotype. Nevertheless, we found increased abundance of fecal Akkermansia from two adult ABL subjects with MTTP mutations compared to their heterozygous parents and within the range noted in six healthy control subjects. Furthermore, Akkermansia abundance across all subjects was positively correlated with fecal coprostanol excretion. Conclusion: The findings collectively suggest multiple adaptive pathways of metabolic regulation following blocked chylomicron assembly, including shifts in BA signaling and altered microbial composition that confer a transmissible phenotype.


Subject(s)
Adaptation, Physiological/genetics , Chylomicrons/genetics , Fatty Liver/metabolism , Gastrointestinal Microbiome/genetics , Lipid Metabolism/genetics , Akkermansia , Animals , Bile Acids and Salts/metabolism , Biological Transport/genetics , Carrier Proteins/metabolism , Disease Models, Animal , Fatty Liver/pathology , Fructose/pharmacology , Glucose Tolerance Test , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Random Allocation , Sensitivity and Specificity , Signal Transduction , Verrucomicrobia/pathogenicity
7.
J Infect Dis ; 220(12): 1904-1914, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31063544

ABSTRACT

BACKGROUND: Zika virus (ZIKV) has become a global concern because infection of pregnant mothers was linked to congenital birth defects. Zika virus is unique from other flaviviruses, because it is transmitted vertically and sexually in addition to by mosquito vectors. Prior studies in mice, nonhuman primates, and humans have shown that ZIKV targets the testis in males, resulting in persistent infection and oligospermia. However, its effects on the corresponding female gonads have not been evaluated. METHODS: In this study, we assessed the effects of ZIKV on the ovary in nonpregnant mice. RESULTS: During the acute phase, ZIKV productively infected the ovary causing accumulation of CD4+ and virus-specific CD8+ T cells. T cells protected against ZIKV infection in the ovary, as higher viral burden was measured in CD8-/- and TCRßδ-/- mice. Increased cell death and tissue inflammation in the ovary was observed during the acute phase of infection, but this normalized over time. CONCLUSIONS: In contrast to that observed with males, minimal persistence and no long-term consequences of ZIKV infection on ovarian follicular reserve or fertility were demonstrated in this model. Thus, although ZIKV replicates in cells of the ovary and causes acute oophoritis, there is rapid resolution and no long-term effects on fertility, at least in mice.


Subject(s)
Fertility , Oophoritis/physiopathology , Oophoritis/virology , Zika Virus Infection/physiopathology , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Biomarkers , Disease Models, Animal , Female , Infertility, Female/etiology , Mice , Mice, Knockout , Oophoritis/complications , Oophoritis/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Viral Load , Viral Tropism , Zika Virus Infection/complications , Zika Virus Infection/pathology
8.
Am J Physiol Endocrinol Metab ; 316(4): E674-E686, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30860882

ABSTRACT

Studies show maternal obesity is a risk factor for metabolic syndrome and nonalcoholic fatty liver disease (NAFLD) in offspring. Here we evaluated potential mechanisms underlying these phenotypes. Female C57Bl6 mice were fed chow or an obesogenic high-fat/high-sucrose (HF/HS) diet with subsequent mating of F1 and F2 female offspring to lean males to develop F2 and F3 generations, respectively. Offspring were fed chow or fibrogenic (high transfat, cholesterol, fructose) diets, and histopathological, metabolic changes, and bile acid (BA) homeostasis was evaluated. Chow-fed F1 offspring from maternal HF/HS lineages (HF/HS) developed periportal fibrosis and inflammation with aging, without differences in hepatic steatosis but increased BA pool size and shifts in BA composition. F1, but not F2 or F3, offspring from HF/HS showed increased steatosis on a fibrogenic diet, yet inflammation and fibrosis were paradoxically decreased in F1 offspring, a trend continued in F2 and F3 offspring. HF/HS feeding leads to increased periportal fibrosis and inflammation in chow-fed offspring without increased hepatic steatosis. By contrast, fibrogenic diet-fed F1 offspring from HF/HS dams exhibited worse hepatic steatosis but decreased inflammation and fibrosis. These findings highlight complex adaptations in NAFLD phenotypes with maternal diet.


Subject(s)
Bile Acids and Salts/metabolism , Cholesterol/metabolism , Diet , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Prenatal Exposure Delayed Effects/metabolism , Triglycerides/metabolism , Animals , Diet, High-Fat , Dietary Fats , Dietary Sucrose , Female , Fibrosis , Fructose , Homeostasis , Inflammation , Liver/pathology , Male , Metabolic Syndrome , Mice , Mice, Inbred C57BL , Obesity , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Trans Fatty Acids
9.
Am J Physiol Heart Circ Physiol ; 316(5): H1202-H1210, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30901280

ABSTRACT

Maternal obesity is correlated with cardiovascular disease in offspring, with a 1.3-fold increase in events observed in offspring of obese women. We have observed that obesity-exposed oocytes demonstrate impaired mitophagy and transmit damaged mitochondria to the offspring. Accordingly, we hypothesized that maternal obesity induces cardiac mitochondrial dysfunction in the offspring via transgenerational inheritance of abnormal oocyte mitochondria. We mated female mice fed a high-fat/high-sucrose (HFS) diet (or chow) with chow-fed males and assessed cardiac structure and function in their descendants that were chow fed in each generation. All F1 to F3 descendants bred via the female in each generation were nonobese and demonstrated cardiac mitochondrial abnormalities with crystal rarefaction and reduced oxygen consumption pointing to a transgenerational effect, while obese F0 dams' hearts were unaffected. Furthermore, male offspring from F1 to F3 generations and female F1 and F2 offspring developed increased left ventricular (LV) mass (vs. chow-fed controls). Increased LV mass was also observed in offspring generated by in vitro fertilization of obesity-exposed oocytes and gestation in nonobese surrogates, ruling out a gestational environment effect. Contrary to our hypothesis, male F1 also transmitted these effects to their offspring, ruling out maternal mitochondria as the primary mode of transmission. We conclude that transmission of obesity-induced effects in the oocyte nucleus rather than abnormal mitochondria underlie transgenerational inheritance of cardiac mitochondrial defects in descendants of obese females. These findings will spur exploration of epigenetic alterations in the oocyte genome as potential mechanisms whereby a family history of maternal obesity predisposes to cardiovascular disease in humans.


Subject(s)
Cell Nucleus/genetics , Diet, High-Fat/adverse effects , Dietary Sucrose/adverse effects , Genes, Mitochondrial , Heart Diseases/genetics , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Obesity, Maternal/genetics , Prenatal Exposure Delayed Effects , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Cell Nucleus/metabolism , Cell Nucleus/pathology , Disease Models, Animal , Female , Gestational Weight Gain , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Heredity , Male , Maternal Nutritional Physiological Phenomena , Mice, Inbred C57BL , Mitochondria, Heart/pathology , Obesity, Maternal/metabolism , Obesity, Maternal/physiopathology , Oocytes/metabolism , Oocytes/pathology , Pregnancy , Risk Factors
10.
FASEB J ; 32(1): 63-72, 2018 01.
Article in English | MEDLINE | ID: mdl-28842431

ABSTRACT

Male exposure to cigarette smoke is associated with seminal defects and with congenital anomalies and childhood cancers in offspring. In mice, paternal exposure to cigarette smoke condensate (CSC) causes molecular defects in germ cells and phenotypic effects in their offspring. Here we used an ex vivo testicular explant model and in vivo exposure to determine the concentration at which CSC impairs spermatogenesis and offspring development. We explanted testis tissue at postnatal day (P)5.5 and cultured it until P11.5. Assessment of growth parameters by analyzing expression of cell-specific markers revealed that the explant system maintained structural and functional integrity. We exposed the P5.5 to -11.5 explants to various concentrations (40-160 µg/ml) of CSC and confirmed that nicotine in the CSC was metabolized to cotinine. We assessed various growth and differentiation parameters, as well as testosterone production, and observed that many spermatogenesis features were impaired at 160 µg/ml CSC. The same parameters were impaired by a similar CSC concentration in vivo Finally, females mated to males that were exposed to 160 µg/ml CSC neonatally had increased rates of pup resorption. We conclude that male exposure to CSC impairs offspring development and that the concentration at which CSC impairs spermatogenesis is similar in vivo and ex vivo. Given that the concentrations of CSC we used contained similar doses of nicotine as human smokers are exposed to, we argue that our model mimics human male reproductive effects of smoking.-Esakky, P., Hansen, D. A., Drury, A. M., Felder, P., Cusumano, A., Moley, K. H. Testicular cells exhibit similar molecular responses to cigarette smoke condensate ex vivo and in vivo.


Subject(s)
Smoking/adverse effects , Spermatogenesis/drug effects , Testis/drug effects , Animals , Animals, Newborn , Cells, Cultured , Cotinine/metabolism , Embryo Loss/chemically induced , Embryonic Development/drug effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Models, Animal , Nicotine/metabolism , Pregnancy , Smoke/adverse effects , Testis/metabolism , Testis/pathology , Testosterone/metabolism , Tissue Culture Techniques
11.
Dev Biol ; 426(1): 126-138, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28438607

ABSTRACT

Mitochondria are the most prominent organelle in the oocyte. Somatic cells maintain a healthy population of mitochondria by degrading damaged mitochondria via mitophagy, a specialized autophagy pathway. However, evidence from previous work investigating the more general macroautophagy pathway in oocytes suggests that mitophagy may not be active in the oocyte. This would leave the vast numbers of mitochondria - poised to be inherited by the offspring - vulnerable to damage. Here we test the hypothesis that inactive mitophagy in the oocyte underlies maternal transmission of dysfunctional mitochondria. To determine whether oocytes can complete mitophagy, we used either CCCP or AntimycinA to depolarize mitochondria and trigger mitophagy. After depolarization, we did not detect co-localization of mitochondria with autophagosomes and mitochondrial DNA copy number remained unchanged, indicating the non-functional mitochondrial population was not removed. To investigate the impact of an absence of mitophagy in oocytes with damaged mitochondria on offspring mitochondrial function, we utilized in vitro fertilization of high fat high sugar (HF/HS)-exposed oocytes, which have lower mitochondrial membrane potential and damaged mitochondria. Here, we demonstrate that blastocysts generated from HF/HS oocytes have decreased mitochondrial membrane potential, lower metabolites involved in ATP generation, and accumulation of PINK1, a mitophagy marker protein. This mitochondrial phenotype in the blastocyst mirrors the phenotype we show in HF/HS exposed oocytes. Taken together, these data suggest that the mechanisms governing oocyte mitophagy are fundamentally distinct from those governing somatic cell mitophagy and that the absence of mitophagy in the setting of HF/HS exposure contributes to the oocyte-to-blastocyst transmission of dysfunctional mitochondria.


Subject(s)
Autophagy/physiology , DNA, Mitochondrial/genetics , Gene Dosage/genetics , Membrane Potential, Mitochondrial/physiology , Mitochondria/pathology , Mitophagy/physiology , Oocytes/metabolism , Animals , Antimycin A/toxicity , Cells, Cultured , Female , Gene Dosage/drug effects , Hydrazones/toxicity , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Obesity/pathology
12.
Physiology (Bethesda) ; 32(1): 51-59, 2017 01.
Article in English | MEDLINE | ID: mdl-27927805

ABSTRACT

Recent human and animal studies investigating the roles of the genome, epigenome, and environmental cues have identified associations between offspring predisposition to life-long obesity/metabolic disease and epigenetic modifications such as DNA methylation. This review explores the mechanisms by which maternal exposures impair the health of not only the next generation but also potentially future generations of offspring.


Subject(s)
Epigenesis, Genetic , Maternal Exposure , Maternal-Fetal Exchange , Metabolic Diseases/genetics , Obesity/genetics , Animals , DNA Methylation , Endocrine Disruptors , Female , Histones/metabolism , Humans , Metabolic Diseases/physiopathology , Mitochondria/genetics , Mitochondria/metabolism , Obesity/physiopathology , Pregnancy
13.
Am J Physiol Endocrinol Metab ; 312(2): E109-E116, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28028036

ABSTRACT

Increased sugar consumption, particularly fructose, in the form of sweetened beverages and sweeteners in our diet adversely affects metabolic health. Because these effects are associated with features of the metabolic syndrome in humans, the direct effect of fructose on pancreatic islet function is unknown. Therefore, we examined the islet phenotype of mice fed excess fructose. Fructose-fed mice exhibited fasting hyperglycemia and glucose intolerance but not hyperinsulinemia, dyslipidemia, or hyperuricemia. Islet function was impaired, with decreased glucose-stimulated insulin secretion and increased glucagon secretion and high fructose consumption leading to α-cell proliferation and upregulation of the fructose transporter GLUT5, which was localized only in α-cells. Our studies demonstrate that excess fructose consumption contributes to hyperglycemia by affecting both ß- and α-cells of islets in mice.


Subject(s)
Fructose/pharmacology , Glucose/metabolism , Homeostasis/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Animals , Cells, Cultured , Dietary Carbohydrates/pharmacology , Down-Regulation/drug effects , Glucose Intolerance/metabolism , Male , Mice , Mice, Inbred C57BL
14.
Hum Reprod ; 31(6): 1315-26, 2016 06.
Article in English | MEDLINE | ID: mdl-27052498

ABSTRACT

STUDY QUESTION: What effect does diet-induced obesity have on endometrial stromal cell (ESC) decidualization? SUMMARY ANSWER: Diet-induced obesity impairs ESC decidualization. WHAT IS KNOWN ALREADY: Decidualization is important for successful implantation and subsequent health of the pregnancy. Compared with normal-weight women, obese women have lower pregnancy rates (both spontaneous and by assisted reproductive technology), higher rates of early pregnancy loss and poorer oocyte quality. STUDY DESIGN, SIZE, DURATION: Beginning at 6 weeks of age, female C57Bl/6J mice were fed either a high-fat/high-sugar diet (HF/HS; 58% Fat Energy/Sucrose) or a diet of standard mouse chow (CON; 13% Fat) for 12 weeks. At this point, metabolic parameters were measured. Some of the mice (n = 9 HF/HS and 9 CON) were mated with reproductively competent males, and implantation sites were assessed. Other mice (n = 11 HF/HS and 10 CON) were mated with vasectomized males, and artificial decidualization was induced. For in vitro human studies of primary ESCs, endometrial tissue was obtained via biopsy from normo-ovulatory patients without history of infertility (obese = BMI > 30 kg/m(2), n = 11 and lean = BMI < 25 kg/m(2), n = 7) and from patients consented for hysterectomies for a benign indication (n = 4). In vitro studies were also performed with immortalized human ESCs. ESCs were decidualized in culture for nine 9 days in the presence or absence of palmitic acid (PA), and the degree of decidualization was assessed by measuring expression of decidualization markers. PARTICIPANTS/MATERIALS, SETTING, METHODS: The sizes of implantation sites and fetuses were analyzed in mice mated with reproductively competent males. In mice mated with vasectomized males, decidualization was induced, and uterine tissues were analyzed via hematoxylin and eosin staining, quantitative RT-PCR (RT-qPCR), and western blots. Human ESCs were cultured in vitro and induced to decidualize by treatment with cAMP and medroxyprogesterone. The level of expression of decidualization markers was assessed by RT-qPCR (mRNA) and western blotting (protein). ATP content of ESCs was measured, and levels of autophagy were assessed by western blotting of the autophagy regulators acetyl coa carboxylase (ACC) and ULK1 (Ser 317). Autophagic flux was measured by western blot of the marker LC3b-II. MAIN RESULTS AND THE ROLE OF CHANCE: Mice exposed to an HF/HS diet became obese and metabolically impaired. HF/HS-exposed mice mated to reproductively competent males had smaller implantation sites in early pregnancy (P <0.001) and larger fetuses at term (P <0.05) than CON-exposed mice. In the artificial decidualization experiments, mice exposed to the HF/HS diet developed 50% smaller deciduomas than mice exposed to CON diet (P< 0.001). Human ESCs cultured in the presence of PA had markedly decreased mRNA expression of the decidualization markers, decidual prolactin (PRL) (P< 0.0001) and insulin-like growth factor binding protein 1 (IGFBP1) (P< 0.0001). Expression of PRL and IGFBP1 by mRNA were also significantly lower in early follicular phase ESCs of obese women than in those of normal-weight women (P< 0.05). Protein expression of phosphorylated ACC and phosphorylated ULK1, both activated forms, were lower in deciduomas of HF/HS mice than in those of control mice (P < 0.01). In immortalized human ESCs, LC3b-II levels were higher in decidualized cells than in controls, indicating increased autophagy. PA treatment abrogated this increase. LIMITATIONS, REASONS FOR CAUTION: Many aspects of obesity and metabolic impairment could contribute to the decidualization defects observed in the HF/HS-exposed mice. Although our findings suggest that both autophagy and decidualization are impaired by exposure to PA, the underlying mechanisms should be elucidated. Finally, our human patient sample size was small. WIDER IMPLICATIONS OF THE FINDINGS: Although many factors contribute to poor reproductive outcome and early pregnancy loss in obese women, our study suggests the importance of decidualization defects. Such defects may contribute to compromised endometrial receptivity and poor implantation. If defects in autophagy contribute to impaired decidualization, therapeutics could be developed to improve this process and thus improve implantation and pregnancy outcomes in obese women. STUDY FUNDING/COMPETING INTERESTS: Grants include NIH 5T32HD040135-12 (J.S.R.), R01 HD065435 (K.H.M.), NIH T32 HD049305 (J.L.S.) and ACOG Research Grant (M.B.S.). The authors report no conflicts of interest.


Subject(s)
Autophagy , Diet, High-Fat , Obesity/pathology , Stromal Cells/pathology , Animals , Biomarkers/metabolism , Decidua , Embryo Implantation , Female , Humans , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Palmitic Acid/pharmacology , Phosphorylation , RNA, Messenger/metabolism
15.
Reproduction ; 151(3): 261-70, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26700938

ABSTRACT

Obesity negatively affects many aspects of the human body including reproductive function. In females, the root of the decline in fertility is linked to problems in the oocyte. Problems seen in oocytes that positively correlate with increasing BMI include changes to the metabolism, lipid accumulation, meiosis, and metaphase II (MII) spindle structure. Studies in mice indicate that dietary interventions fail to reverse these problems. How exercise affects the oocytes has not been addressed. Therefore, we hypothesized an exercise intervention would improve oocyte quality. Here we show that in a mouse model of an exercise, intervention can improve lipid metabolism in germinal vesicle (GV) stage oocytes. Oocytes significantly increased activity and transcription of the ß-oxidation enzyme hydroxyacyl-coenzyme A dehydrogenase in response to exercise training only if the mice had been fed a high-fat diet (HFD). An exercise intervention also reversed the lipid accumulation seen in GV stage oocytes of HFD females. However, delays in meiosis and disorganized MII spindles remained present. Therefore, exercise is able to improve, but not reverse, damage imparted on oocytes as a result of an HFD and obesity. By utilizing an exercise intervention on an HFD, we determined only lipid content, and lipid metabolism is changed in GV oocytes. Moving forward, interventions to improve oocyte quality may need to be more targeted to the oocyte specifically. Because of the HFD-induced deficiency in ß-oxidation, dietary supplementation with substrates to improve lipid utilization may be more beneficial.


Subject(s)
Obesity/therapy , Oocytes/metabolism , Physical Conditioning, Animal , Animals , Diet, High-Fat/adverse effects , Female , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mitochondria/pathology , Mitochondria/ultrastructure , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Obesity/metabolism , Obesity/pathology , Spindle Apparatus/pathology
16.
Cell Mol Life Sci ; 72(2): 251-71, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25280482

ABSTRACT

Obesity, diabetes, and related metabolic disorders are major health issues worldwide. As the epidemic of metabolic disorders continues, the associated medical co-morbidities, including the detrimental impact on reproduction, increase as well. Emerging evidence suggests that the effects of maternal nutrition on reproductive outcomes are likely to be mediated, at least in part, by oocyte metabolism. Well-balanced and timed energy metabolism is critical for optimal development of oocytes. To date, much of our understanding of oocyte metabolism comes from the effects of extrinsic nutrients on oocyte maturation. In contrast, intrinsic regulation of oocyte development by metabolic enzymes, intracellular mediators, and transport systems is less characterized. Specifically, decreased acid transport proteins levels, increased glucose/lipid content and elevated reactive oxygen species in oocytes have been implicated in meiotic defects, organelle dysfunction and epigenetic alteration. Therefore, metabolic disturbances in oocytes may contribute to the diminished reproductive potential experienced by women with metabolic disorders. In-depth research is needed to further explore the underlying mechanisms. This review also discusses several approaches for metabolic analysis. Metabolomic profiling of oocytes, the surrounding granulosa cells, and follicular fluid will uncover the metabolic networks regulating oocyte development, potentially leading to the identification of oocyte quality markers and prevention of reproductive disease and poor outcomes in offspring.


Subject(s)
Energy Metabolism/physiology , Maternal Nutritional Physiological Phenomena/physiology , Metabolome/physiology , Models, Biological , Oocytes/growth & development , Oocytes/metabolism , Reproduction/physiology , Amino Acids/metabolism , Female , Glucose/metabolism , Granulosa Cells/metabolism , Humans , Lipid Metabolism
17.
Diabetologia ; 58(3): 615-24, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25608625

ABSTRACT

AIMS/HYPOTHESIS: Maternal obesity is associated with an increased risk of obesity and impaired glucose homeostasis in offspring. However, it is not known whether a gestational or pre-gestational exposure confers similar risks, and if so, what the underlying mechanisms are. METHODS: We used reciprocal two-cell embryo transfers between mice fed either a control or high-fat diet (HFD) starting at the time of weaning. Gene expression in placenta was assessed by microarray analyses. RESULTS: A pre-gestational exposure to a maternal HFD (HFD/control) impaired fetal and placental growth despite a normal gestational milieu. Expression of imprinted genes and genes regulating vasculogenesis and lipid metabolism was markedly altered in placenta of HFD/control. An exposure to an HFD (control/HFD) only during gestation also resulted in fetal growth restriction and decreased placental weight. Interestingly, only a gestational exposure to an HFD (control/HFD) resulted in obesity and impaired glucose tolerance in adulthood. CONCLUSIONS/INTERPRETATION: An HFD during pregnancy has profound consequences for the offspring later in life. Our data demonstrate that the mechanism underlying this phenomenon is not related to placental dysfunction, intrauterine growth restriction or postnatal weight gain, but rather an inability of the progeny to adapt to the abnormal gestational milieu of an HFD. Thus, the ability to adapt to an adverse intrauterine environment is conferred prior to pregnancy and it is possible that the effects of a maternal HFD may be transmitted to subsequent generations.


Subject(s)
Obesity/complications , Animals , Animals, Newborn , Body Weight/physiology , Diet, High-Fat/adverse effects , Female , Fetal Growth Retardation/etiology , Male , Mice , Placenta/pathology , Pregnancy , Pregnancy Complications , Prenatal Exposure Delayed Effects
18.
J Biol Chem ; 289(16): 10989-10998, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24519932

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in the world, and it is thought to be the hepatic manifestation of the metabolic syndrome. Excess dietary fructose causes both metabolic syndrome and NAFLD in rodents and humans, but the pathogenic mechanisms of fructose-induced metabolic syndrome and NAFLD are poorly understood. GLUT8 (Slc2A8) is a facilitative glucose and fructose transporter that is highly expressed in liver, heart, and other oxidative tissues. We previously demonstrated that female mice lacking GLUT8 exhibit impaired first-pass hepatic fructose metabolism, suggesting that fructose transport into the hepatocyte, the primary site of fructose metabolism, is in part mediated by GLUT8. Here, we tested the hypothesis that GLUT8 is required for hepatocyte fructose uptake and for the development of fructose-induced NAFLD. We demonstrate that GLUT8 is a cell surface-localized transporter and that GLUT8 overexpression or GLUT8 shRNA-mediated gene silencing significantly induces and blocks radiolabeled fructose uptake in cultured hepatocytes. We further show diminished fructose uptake and de novo lipogenesis in fructose-challenged GLUT8-deficient hepatocytes. Finally, livers from long term high-fructose diet-fed GLUT8-deficient mice exhibited attenuated fructose-induced hepatic triglyceride and cholesterol accumulation without changes in hepatocyte insulin-stimulated Akt phosphorylation. GLUT8 is thus essential for hepatocyte fructose transport and fructose-induced macrosteatosis. Fructose delivery across the hepatocyte membrane is thus a proximal, modifiable disease mechanism that may be exploited to prevent NAFLD.


Subject(s)
Cell Membrane/metabolism , Fatty Liver/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Hepatocytes/metabolism , Lipogenesis , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/genetics , Cell Membrane/genetics , Cell Membrane/pathology , Cholesterol/genetics , Cholesterol/metabolism , Fatty Liver/genetics , Fatty Liver/pathology , Female , Fructose/genetics , Fructose/metabolism , Gene Silencing , Glucose/genetics , Glucose/metabolism , Glucose Transport Proteins, Facilitative/genetics , Hep G2 Cells , Hepatocytes/pathology , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Triglycerides/genetics , Triglycerides/metabolism
19.
Reprod Fertil Dev ; 27(4): 716-24, 2015 May.
Article in English | MEDLINE | ID: mdl-25775080

ABSTRACT

Obesity adversely affects reproduction and results in oocyte defects in both mice and humans. In the present study we used a mouse model to examine whether the adverse effects of an obesogenic diet on oocyte metabolism and morphology can be reversed by return to a control diet. The intervention group consisted of C57BL6/J mice placed on a high-fat diet (HFD; 35.8% fat and 20.2% protein by nutritional content) for 6 weeks and then switched to an isocaloric control diet (CD; 13% fat and 25% protein) for 8 weeks (HFD/CD mice). The control group consisted of age-matched C57BL6/J mice maintained on CD for 14 weeks (CD/CD mice). Although metabolic parameters (weight, glucose tolerance and cholesterol levels) of HFD/CD mice returned to normal after this 'diet reversal' period, several oocyte defects were not reversible. These HFD/CD oocytes demonstrated significantly higher percentages of abnormal meiotic spindles, lower mitochondrial membrane potential and lower ATP and citrate levels, and higher percentages of abnormal lipid accumulation and mitochondrial distribution compared with CD/CD mice. These results suggest that the negative effects of an obesogenic diet on oocyte quality are not reversible, despite reversal of metabolic parameters. These data may provide better insight when counselling obese women regarding reproductive options and success.


Subject(s)
Diet, High-Fat , Obesity/metabolism , Oocytes/metabolism , Animals , Body Weight/physiology , Female , Mice , Reproduction/physiology
20.
Biol Reprod ; 90(1): 9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24258214

ABSTRACT

Prior studies in our laboratory have demonstrated that cigarette smoke condensate (CSC) activates arylhydrocarbon receptor (Ahr) leading to upregulation of several antioxidant enzymes in murine spermatocytes. In this study, we show that exposure of the spermatocyte cell line GC-2spd(ts) to CSC induces an increase in Cyp1a1, demonstrating AHR activation, and simultaneous expression and nuclear translocation of nuclear factor erythroid 2-related factor 2 (NRF2), where it is believed to modulate Ahr expression by a feedback mechanism. Pharmacological inhibition by the AHR-antagonist CH223191 and interference by Ahr- and Nrf2-small interfering RNA followed by quantitative real-time PCR implicate the Ahr-Nrf2 pathway in the modulation of DNA damage and growth suppression genes such as Gadd45a and P21 and oxidative stress-related genes Cyp1a1, Nrf2, and Ahrr. Flow cytometry accompanied with cell proliferation assay indicate the CSC induces accumulation of spermatocytes at the S-G2/M phase of the cell cycle. Thus, the data obtained suggest that CSC contains several AHR-agonists that are capable of altering the growth pattern of spermatocytes in vitro through the Ahr-Nrf2 signaling mechanism.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Cycle/drug effects , NF-E2-Related Factor 2/physiology , Nicotiana/toxicity , Receptors, Aryl Hydrocarbon/physiology , Smoke/adverse effects , Spermatocytes/cytology , Spermatocytes/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Male , Mice , Protein Transport/drug effects , Signal Transduction/drug effects , Spermatocytes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL