201
|
Abstract
A key characteristic of cancer cells is the ability to switch from a predominantly oxidative metabolism to glycolysis and the production of lactate even when oxygen is plentiful. This metabolic switch, known as the Warburg effect, was first described in the 1920s, and has fascinated and puzzled researchers ever since. However, a dramatic increase in glycolysis in the presence of oxygen is one of the hallmarks of the development of the early mammalian embryo; a metabolic switch with many parallels to the Warburg effect of cancers. The present review provides a brief overview of this and other similarities between the metabolism in tumours and early embryos and proposes whether knowledge of early embryo metabolism can help us to understand metabolic regulation in cancer cells.
Collapse
|
202
|
O'Gorman A, Wallace M, Cottell E, Gibney MJ, McAuliffe FM, Wingfield M, Brennan L. Metabolic profiling of human follicular fluid identifies potential biomarkers of oocyte developmental competence. Reproduction 2013; 146:389-95. [PMID: 23886995 DOI: 10.1530/rep-13-0184] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The use of metabolomic based techniques to aid oocyte and embryo selection has gained attention in recent years. Previous work from our laboratory has demonstrated that the (1)H NMR-based metabolic profile of follicular fluid correlates with oocyte developmental potential. Patients undergoing IVF at the Merrion Fertility Clinic had follicular fluid collected at the time of oocyte retrieval. The fatty acid composition of follicular fluid from follicles where oocytes fertilised and developed into multi-cell embryos (n=15) and from oocytes that fertilised normally but failed to cleave (n=9) (cleaved vs non-cleaved) was compared. Statistical analysis was performed on the data using univariate and multivariate techniques. Analysis of the fatty acid composition revealed that there were nine fatty acids significantly different between follicular fluid from the cleaved and the non-cleaved sample groups. Of particular interest were the higher concentration of total saturated (P=0.03) and the lower concentration of total polyunsaturated fatty acids in the non-cleaved sample group (P=0.001). Random forest classification models were used to predict successful cleavage in follicular fluid samples producing models with errors rates of <10%. Receiver operating characteristic analysis demonstrated that the model had good predictability with an area under the curve of 0.96. The panel of fatty acid biomarkers identified in this study indicates that the fatty acid composition of follicular fluid may be more predictive in comparison to other previously identified biomarkers. Following validation in a larger cohort, these biomarkers may have the potential to be used in fertility clinics to aid the selection of oocytes in the future.
Collapse
Affiliation(s)
- A O'Gorman
- UCD School of Agriculture and Food Science, UCD Conway Institute, Belfield, Dublin, Ireland
| | | | | | | | | | | | | |
Collapse
|
203
|
Abstract
STUDY QUESTION Are overall and central obesity associated with reduced fecundability in US black women? SUMMARY ANSWER Overall and central obesity--based on self-reported measures of body mass index (BMI, kg/m(2)), waist circumference and waist-to-hip ratio--were independent risk factors for subfertility in our cohort. WHAT IS KNOWN ALREADY Overall obesity (BMI ≥ 30 kg/m(2)) has been associated with infertility in several studies. The role of central obesity is less clear. There are no previous studies of time-to-pregnancy (TTP) in black women. STUDY DESIGN, SIZE, DURATION Data were derived from the Black Women's Health Study, a prospective cohort study. During 1995-2011, there were 2239 planned pregnancy attempts reported by 1697 women, resulting in 2022 births. Cohort retention was greater than 80%. PARTICIPANTS/MATERIALS, SETTING, METHODS Eligible women were aged 21-40 years and reported at least one planned pregnancy attempt during 1995-2011. Height and weight were reported in 1995, with weight updated every two years; waist and hip circumferences were reported in 1995 and updated in 2003. A validation study within the cohort showed high correlations between self-reported and technician-measured weight (r = 0.97), height (r = 0.93), waist circumference (r = 0.75) and hip circumference (r = 0.74). In 2011, TTP was reported in months. Proportional probabilities regression models were used to estimate fecundability ratios (FRs) and 95% confidence intervals (CI), adjusting for covariates. MAIN RESULTS AND THE ROLE OF CHANCE High BMI was associated with delayed conception: relative to BMI 18.5-24.9, FRs for BMI categories of <18.5, 25.0-29.9, 30.0-34.9 and ≥ 35.0 were 0.92 (CI: 0.64-1.32), 0.93 (CI: 0.84-1.03), 0.92 (CI: 0.79-1.06) and 0.73 (CI: 0.61-0.87), respectively. Associations were stronger among nulliparous women (P-interaction = 0.003). After controlling for BMI, reduced fecundability was observed among women with large waist circumferences (≥ 33 versus <26 inches: FR = 0.73, CI: 0.60-0.88) and large waist-to-hip ratios (≥ 0.85 versus <0.71: FR = 0.83, CI: 0.71-0.97). LIMITATIONS, REASONS FOR CAUTION TTP was reported retrospectively and error in recall is likely, particularly as time since the pregnancy increases. However, results were similar when based on the most recent versus first pregnancies. Confounding may have been introduced by the lack of control for important determinants of TTP. Nevertheless, control for maternal age and education, which are highly correlated with TTP determinants such as paternal age and persistence in trying, should reduce the extent of confounding. The analysis was confined to planned pregnancies. If pregnancy intention was related both to body size and fecundability, our results could be biased. Bias is likely to be small because we found little difference in body size and other measured characteristics between pregnancy planners and non-planners. WIDER IMPLICATIONS OF THE FINDINGS Our findings add to the growing body of literature showing that excess BMI is associated with reduced fecundability and further suggest that central obesity is an important independent risk factor for infertility. The relation of obesity to infertility is especially relevant to US black women because they have higher rates of obesity and infertility. Reductions in overall and central obesity may offer the potential to improve fertility outcomes. STUDY FUNDING/COMPETING INTEREST(S) This work was funded by National Cancer Institute grant CA58420. We have no competing interests to report.
Collapse
Affiliation(s)
- Lauren A Wise
- Slone Epidemiology Center at Boston University, 1010 Commonwealth Avenue, Boston, MA 02215, USA
| | | | | |
Collapse
|
204
|
Sharma R, Biedenharn KR, Fedor JM, Agarwal A. Lifestyle factors and reproductive health: taking control of your fertility. Reprod Biol Endocrinol 2013; 11:66. [PMID: 23870423 PMCID: PMC3717046 DOI: 10.1186/1477-7827-11-66] [Citation(s) in RCA: 372] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 07/10/2013] [Indexed: 12/16/2022] Open
Abstract
Approximately 10 to 15% of couples are impacted by infertility. Recently, the pivotal role that lifestyle factors play in the development of infertility has generated a considerable amount of interest. Lifestyle factors are the modifiable habits and ways of life that can greatly influence overall health and well-being, including fertility. Many lifestyle factors such as the age at which to start a family, nutrition, weight, exercise, psychological stress, environmental and occupational exposures, and others can have substantial effects on fertility; lifestyle factors such as cigarette smoking, illicit drug use, and alcohol and caffeine consumption can negatively influence fertility while others such as preventative care may be beneficial. The present literature review encompasses multiple lifestyle factors and places infertility in context for the couple by focusing on both males and females; it aims to identify the roles that lifestyle factors play in determining reproductive status. The growing interest and amount of research in this field have made it evident that lifestyle factors have a significant impact on fertility.
Collapse
Affiliation(s)
- Rakesh Sharma
- Center for Reproductive Medicine, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Kelly R Biedenharn
- Center for Reproductive Medicine, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Jennifer M Fedor
- Center for Reproductive Medicine, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Ashok Agarwal
- Center for Reproductive Medicine, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, USA
| |
Collapse
|
205
|
Abstract
Obesity is associated with multiple adverse reproductive outcomes, but the mechanisms involved are largely unknown. Obesity has been referred to as a "complex system," defined as a system of heterogeneous parts interacting in nonlinear ways to influence the behavior of the parts as a whole. Human reproduction is also a complex system; hence the difficulty in identifying the mechanisms linking obesity and adverse reproductive function. This review discusses the adverse reproductive outcomes associated with obesity and the mechanisms involved and concludes with a discussion of public health policy with respect to the treatment of infertility in obese women.
Collapse
Affiliation(s)
- Emily S Jungheim
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Washington University in St Louis, St Louis, MO 63108, USA.
| | | | | | | |
Collapse
|
206
|
Merhi Z, Buyuk E, Berger DS, Zapantis A, Israel DD, Chua S, Jindal S. Leptin suppresses anti-Mullerian hormone gene expression through the JAK2/STAT3 pathway in luteinized granulosa cells of women undergoing IVF. Hum Reprod 2013; 28:1661-1669. [DOI: 10.1093/humrep/det072] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
207
|
Cordier AG, Léveillé P, Dupont C, Tarrade A, Picone O, Larcher T, Dahirel M, Poumerol E, Mandon-Pepin B, Lévy R, Chavatte-Palmer P. Dietary lipid and cholesterol induce ovarian dysfunction and abnormal LH response to stimulation in rabbits. PLoS One 2013; 8:e63101. [PMID: 23690983 PMCID: PMC3653923 DOI: 10.1371/journal.pone.0063101] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 03/28/2013] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND/AIM Excess of fat intake is dramatically increasing in women of childbearing age and results in numerous health complications, including reproductive disorders. Using rabbit does as a biomedical model, the aim of this study was to evaluate onset of puberty, endocrine responses to stimulation and ovarian follicular maturation in females fed a high fat high cholesterol diet (HH diet) from 10 weeks of age (i.e., 2 weeks before normal onset of puberty) or a control diet (C diet). METHODOLOGY/PRINCIPAL FINDINGS Three experiments were performed, each including 8 treated (HH group) and 8 control (C group) does. In experiment 1, the endocrine response to Gonadotropin releasing hormone (GnRH) was evaluated at 13, 18 and 22 weeks of age. In experiment 2, the follicular population was counted in ovaries of adult females (18 weeks of age). In experiment 3, the LH response to mating and steroid profiles throughout gestation were evaluated at 18 weeks of age. Fetal growth was monitored by ultrasound and offspring birth weight was recorded. Data showed a significantly higher Luteinizing hormone (LH) response after induction of ovulation at 13 weeks of age in the HH group. There was no difference at 18 weeks, but at 22 weeks, the LH response to GnRH was significantly reduced in the HH group. The number of atretic follicles was significantly increased and the number of antral follicles significantly reduced in HH does vs. controls. During gestation, the HH diet induced intra-uterine growth retardation (IUGR). CONCLUSION The HH diet administered from before puberty onwards affected onset of puberty, follicular growth, hormonal responses to breeding and GnRH stimulation in relation to age and lead to fetal IUGR.
Collapse
Affiliation(s)
- Anne-Gaël Cordier
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- APHP, Hosp Antoine Béclère, Service de Gynécologie-Obstétrique et Médecine de la Reproduction, Clamart, France
| | - Pauline Léveillé
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- Université Paris 13, Sorbonne Paris Cité, Unité de Recherche en Epidémiologie Nutritionnelle (UREN), Bobigny, France
- APHP, Hôpital Jean-Verdier, Bondy, France
| | - Charlotte Dupont
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- Université Paris 13, Sorbonne Paris Cité, Unité de Recherche en Epidémiologie Nutritionnelle (UREN), Bobigny, France
- APHP, Hôpital Jean-Verdier, Bondy, France
| | - Anne Tarrade
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- PremUp foundation, Paris, France
| | - Olivier Picone
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
| | | | - Michèle Dahirel
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- PremUp foundation, Paris, France
| | - Elodie Poumerol
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
| | | | - Rachel Lévy
- Université Paris 13, Sorbonne Paris Cité, Unité de Recherche en Epidémiologie Nutritionnelle (UREN), Bobigny, France
- APHP, Hôpital Jean-Verdier, Bondy, France
| | - Pascale Chavatte-Palmer
- INRA, UMR1198 Biologie du Développement et Reproduction, Jouy-en-Josas, France
- PremUp foundation, Paris, France
| |
Collapse
|
208
|
Mouzat K, Baron S, Marceau G, Caira F, Sapin V, Volle DH, Lumbroso S, Lobaccaro JM. Emerging roles for LXRs and LRH-1 in female reproduction. Mol Cell Endocrinol 2013; 368:47-58. [PMID: 22750099 DOI: 10.1016/j.mce.2012.06.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 06/18/2012] [Accepted: 06/19/2012] [Indexed: 01/05/2023]
Abstract
Nutritional status is known to control female reproductive physiology. Many reproductive pathologies such as anorexia nervosa, dystocia and preeclampsia, have been linked to body mass index and to metabolic syndrome. Lipid metabolism has also been associated with ovarian, uterine and placental functions. Among the regulators of lipid homeostasis, the Liver X Receptors (LXRs) and the Liver Receptor Homolog-1 (LRH-1), two members of the nuclear receptor superfamily, play a central role. LXRs are sensitive to intracellular cholesterol concentration and decrease plasma cholesterol, allowing to considering them as "cholesterol sensors". LRH-1 shares many target-genes with LXRs and has been considered for a long time as a real orphan nuclear receptor, but recent findings showed that phospholipids are ligands for this nuclear receptor. Acting in concert, LXRs and LRH-1 could thus be sensitive to slight modifications in cellular lipid balance, tightly maintaining their cellular concentrations. These last years, the use of transgenic mice clarified the roles of these nuclear receptors in many physiological functions. This review will be focused on the roles of LXRs and LRH-1 on female reproduction. Their contribution to ovarian endocrine and exocrine functions, as well as uterine and placental physiology will be discussed. The future challenge will thus be to target these nuclear receptors to prevent lipid-associated reproductive diseases in women.
Collapse
Affiliation(s)
- Kevin Mouzat
- Laboratoire de Biochimie, Centre Hospitalier Universitaire de Nîmes, Hôpital Carémeau, Place du Pr. Robert Debré, F-30029 Nimes, France.
| | | | | | | | | | | | | | | |
Collapse
|
209
|
Michalakis K, Mintziori G, Kaprara A, Tarlatzis BC, Goulis DG. The complex interaction between obesity, metabolic syndrome and reproductive axis: a narrative review. Metabolism 2013; 62:457-78. [PMID: 22999785 DOI: 10.1016/j.metabol.2012.08.012] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 08/13/2012] [Accepted: 08/18/2012] [Indexed: 12/16/2022]
Abstract
The aim of this narrative review is to provide current evidence for the interaction between obesity, metabolic syndrome (MS) and reproductive axis. Gonadotropin-releasing hormone (GnRH) pulses and, consequently, normal function of reproductive (hypothalamus-pituitary-gonadal) axis depend on normal energy balance, which presupposes sufficient food intake, reasonable energy consumption and average thermoregulatory costs. In case of an energy imbalance, reproductive dysfunction may occur. In young women, excessive leanness is accompanied by puberty delay, whereas premature puberty might be a manifestation of obesity. In a similar way, obesity in men affects fertility. Excess adipose tissue results in increased conversion of testosterone to estradiol, which may lead to secondary hypogonadism through reproductive axis suppression. Moreover, oxidative stress at the level of the testicular micro-environment may result in decreased spermatogenesis and sperm damage. Products of the adipocyte, such as leptin, adiponectin and resistin, and gut peptides, such as ghrelin, are considered to be crucial in the interaction between energy balance and reproduction. Finally, an indirect evidence for the interplay between MS and reproductive axis is the fact that when treating components of one, parameters of the other can be improved as well. These therapeutic interventions include lifestyle modifications, pharmacological agents, such as sex hormone replacement therapy, and surgical procedures. Although many issues remain unclear, the elucidation of the complex interaction between MS and reproductive axis will have obvious clinical implications in the therapeutic approach of both entities.
Collapse
Affiliation(s)
- Konstantinos Michalakis
- First Department of Internal Medicine, Laikon University Hospital, Athens University Medical School, Greece
| | | | | | | | | |
Collapse
|
210
|
A novel approach to quantifying ovarian cell lipid content and lipid accumulation in vitro by confocal microscopy in lean women undergoing ovarian stimulation for in vitro fertilization (IVF). J Assist Reprod Genet 2013; 30:733-40. [PMID: 23512091 DOI: 10.1007/s10815-013-9976-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 03/06/2013] [Indexed: 01/21/2023] Open
Abstract
PURPOSE To quantify intracellular lipid levels in cumulus cells (CCs) and mural granulosa cells (MGCs) of lean women undergoing gonadotropin therapy for in vitro fertilization (IVF), based upon different cell preparation methods. METHODS CCs and MGCs from 16 lean women undergoing ovarian stimulation for IVF were studied. Cells were pooled by cell type, with each type of cell separated into two groups for determination of initial lipid content (Method 1) and subsequent lipid accumulation in vitro (Method 2). Cells for initial lipid content were immediately fixed at the time of the oocyte retrieval with 4% paraformaldehyde in suspension, while those for subsequent lipid accumulation in vitro were cultured for 4 h with 5% fetal calf serum and then fixed. Cells were treated with lipid fluorescent dye BODIPY® FL C16 and nuclear marker DAPI. Intracellular lipid was quantified by confocal microscopy, using ImageJ software analysis. RESULTS There was no significant effect of cell type (P = 0.2) or cell type-cell preparation method interaction (P = 0.8) on cell area (Method 1: CC 99.7 ± 5.1, MGC 132.8 ± 5.8; Method 2: CC 221.9 ± 30.4, MGC 265.1 ± 48.5 μm(2)). The mean area of all cells combined was significantly less for cells prepared by Method 1 (116.2 ± 4.9 μm(2)) vs. Method 2 (243.5 ± 22.5 μm(2), P < 0.00005). Intracellular lipid level, however, was significantly altered by cell preparation method (P < 0.05; cell preparation method-cell type interaction, P < 0.00001). Initial lipid content was significantly lower in CC (74.5 ± 9.3) than MGC (136.3 ± 16.7 fluorescence/cell area, P < 0.00005), while subsequent lipid accumulation in vitro was significantly higher in CC (154.0 ± 9.1) than MGC (104.6 ± 9.9 fluorescence/cell area, P < 0.00001). The relatively diminished initial CC lipid content compared to subsequent CC lipid accumulation in vitro (P < 0.00001), and the opposite pattern for MGC (P < 0.05), significantly lowered the CC/MGC lipid ratio in Method 1 (0.55 ± 0.04) vs. Method 2 (1.58 ± 0.10, P < 0.00001). CONCLUSIONS Differential uptake or utilization of lipid by CC and MGC occurs during oocyte maturation and steroidogenesis, respectively, with the amount of lipid present in ovarian cells a function of both the follicular microenvironment at the time of the oocyte retrieval and the capacity of these cells to accumulate lipid in vitro over time.
Collapse
|
211
|
Butler MS, Yang X, Ricciardelli C, Liang X, Norman RJ, Tilley WD, Hickey TE. Small glutamine-rich tetratricopeptide repeat-containing protein alpha is present in human ovaries but may not be differentially expressed in relation to polycystic ovary syndrome. Fertil Steril 2013; 99:2076-83.e1. [PMID: 23433514 DOI: 10.1016/j.fertnstert.2013.01.140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Revised: 01/25/2013] [Accepted: 01/25/2013] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate the expression and function of small glutamine-rich tetratricopeptide repeat-containing protein alpha (SGTA), an androgen receptor (AR) molecular chaperone, in human ovarian tissues. DESIGN Examine the effect of SGTA on AR subcellular localization in granulosa tumor cells (KGN) and SGTA expression in ovarian tissues. SETTING University-based research laboratory. PATIENT(S) Archived tissues from premenopausal women and granulosa cells from infertile women receiving assisted reproduction. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) AR subcellular localization and SGTA protein or mRNA levels. RESULT(S) SGTA and AR proteins were expressed in the cytoplasm of KGN cells and exposure to androgen stimulated AR nuclear localization. SGTA protein knockdown increased AR nuclear localization at low (0-0.1 nmol/L) but not high (1-10 nmol/L) concentrations of androgen hormone. In ovarian tissues, SGTA was localized to the cytoplasm of granulosa cells at all stages of folliculogenesis and in thecal cells of antral follicles. SGTA protein levels were similar when comparing primordial and primary follicles within core biopsies (n = 40) from women with and without polycystic ovary syndrome (PCOS). Likewise, SGTA mRNA levels were not significantly different in granulosa cells from preovulatory follicles after hyperstimulation of women with and without PCOS. CONCLUSION(S) SGTA is present in human ovaries and has the potential to modulate AR signalling, but it may not be differentially expressed in PCOS.
Collapse
Affiliation(s)
- Miriam S Butler
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | | | | | | | | | | | | |
Collapse
|
212
|
Van Hoeck V, Leroy JLMR, Arias Alvarez M, Rizos D, Gutierrez-Adan A, Schnorbusch K, Bols PEJ, Leese HJ, Sturmey RG. Oocyte developmental failure in response to elevated nonesterified fatty acid concentrations: mechanistic insights. Reproduction 2013; 145:33-44. [PMID: 23108110 DOI: 10.1530/rep-12-0174] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Elevated plasma nonesterified fatty acid (NEFA) concentrations are associated with negative energy balance and metabolic disorders such as obesity and type II diabetes. Such increased plasma NEFA concentrations induce changes in the microenvironment of the ovarian follicle, which can compromise oocyte competence. Exposing oocytes to elevated NEFA concentrations during maturation affects the gene expression and phenotype of the subsequent embryo, notably prompting a disrupted oxidative metabolism. We hypothesized that these changes in the embryo are a consequence of modified energy metabolism in the oocyte. To investigate this, bovine cumulus oocyte complexes were matured under elevated NEFA conditions, and energy metabolism-related gene expression, mitochondrial function, and ultrastructure evaluated. It was found that expression of genes related to REDOX maintenance was modified in NEFA-exposed oocytes, cumulus cells, and resultant blastocysts. Moreover, the expression of genes related to fatty acid synthesis in embryos that developed from NEFA-exposed oocytes was upregulated. From a functional perspective, inhibition of fatty acid β-oxidation in maturing oocytes exposed to elevated NEFA concentrations restored developmental competence. There were no clear differences in mitochondrial morphology or oxygen consumption between treatments, although there was a trend for a higher mitochondrial membrane potential in zygotes derived from NEFA-exposed oocytes. These data show that the degree of mitochondrial fatty acid β-oxidation has a decisive impact on the development of NEFA-exposed oocytes. Furthermore, the gene expression data suggest that the resulting embryos adapt through altered metabolic strategies, which might explain the aberrant energy metabolism previously observed in these embryos originating from NEFA-exposed maturing oocytes.
Collapse
Affiliation(s)
- V Van Hoeck
- Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, Gamete Research Center, University of Antwerp, Universiteitsplein 1-Gebouw U, B-2610 Wilrijk, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Obesity at age 20 and the risk of miscarriages, irregular periods and reported problems of becoming pregnant: the Adventist Health Study-2. Eur J Epidemiol 2012; 27:923-31. [PMID: 23224589 PMCID: PMC3539069 DOI: 10.1007/s10654-012-9749-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 11/06/2012] [Indexed: 11/12/2022]
Abstract
In a group of 46,000 North-American Adventist women aged 40 and above, we investigated the relationships between body mass index (BMI, kg/m2) at age 20 and the proportion of women who reported at least one miscarriage, periods with irregular menstruation or failing to become pregnant even if trying for more than one straight year. Approximately 31, 14 and 17 %, respectively, reported the three different problems related to reproduction. Positive age- and marital status adjusted relationships were found between BMI at age 20 and periods with irregular menstruation or failing to become pregnant even if trying for more than 1 year, but not with the risk of miscarriages. Women with BMI ≥ 32.5 kg/m2 when aged 20 had approximately 2.0 (95 % CI: 1.6, 2.4) and 1.5 (95 % CI: 1.3, 1.9) higher odds for irregular periods or failing to get pregnant, respectively, than women with BMI in the 20–24.9 kg/m2 bracket. These relationships were consistently found in a number of strata of the population, including the large proportion of the women who never had smoked or never used alcohol. Underweight (BMI < 18.5 kg/m2) when aged 20 marginally (approximately 15 %) increased the risk of failing to get pregnant within a year. Thus, obesity at age 20 increases the risk of reporting some specific reproductive problems, but not the risk of miscarriages.
Collapse
|
214
|
Binder NK, Mitchell M, Gardner DK. Parental diet-induced obesity leads to retarded early mouse embryo development and altered carbohydrate utilisation by the blastocyst. Reprod Fertil Dev 2012; 24:804-12. [PMID: 22781931 DOI: 10.1071/rd11256] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 12/03/2011] [Indexed: 02/04/2023] Open
Abstract
Maternal obesity results in reproductive complications, whereas the impact of paternal obesity is unclear. In the present study, the effects of parental obesity on preimplantation embryo cell cycle length and carbohydrate utilisation were investigated. Maternal and paternal obesity were assessed independently by deriving zygotes from normal or obese C57BL/6 female mice mated with normal Swiss male mice (maternal obesity), or from normal Swiss female mice mated with normal or obese C57BL/6 male mice (paternal obesity). Zygotes were cultured in vitro and development was then assessed by time-lapse microscopy and metabolism determined using ultramicrofluorescence. Maternal obesity was associated with a significant delay in precompaction cell cycle kinetics from the 1-cell stage. A significant increase in glucose consumption by embryos from obese compared with normal females occurred after compaction, although glycolysis remained unchanged. Similarly, paternal obesity led to significant delays in cell cycle progression during preimplantation embryo development. However, this developmental delay was observed from the second cleavage stage onwards, following embryonic genome activation. Blastocysts from obese males showed disproportionate changes in carbohydrate metabolism, with significantly increased glycolysis. Overall, metabolic changes were not inhibitory to blastocyst formation; however, blastocyst cell numbers were significantly lower when either parent was obese. These data suggest that both maternal and paternal obesity significantly impacts preimplantation embryo physiology.
Collapse
Affiliation(s)
- Natalie K Binder
- Department of Zoology, University of Melbourne, Parkville 3010, Victoria, Australia
| | | | | |
Collapse
|
215
|
Valckx SDM, De Pauw I, De Neubourg D, Inion I, Berth M, Fransen E, Bols PEJ, Leroy JLMR. BMI-related metabolic composition of the follicular fluid of women undergoing assisted reproductive treatment and the consequences for oocyte and embryo quality. Hum Reprod 2012; 27:3531-9. [PMID: 23019302 DOI: 10.1093/humrep/des350] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION Is the metabolic composition of the follicular fluid of women undergoing assisted reproductive treatment (ART) related to serum composition and BMI and is it associated with oocyte and embryo quality? SUMMARY ANSWER We showed that metabolic alterations in the serum are reflected in the follicular fluid and that some of these alterations may affect oocyte quality, irrespective of BMI. WHAT IS KNOWN ALREADY Many studies have focused on the effect of metabolic disorders, such as obesity and type 2 diabetes, on assisted reproduction outcomes. There are, however, only few studies focusing on the importance of the correlation between serum and follicular fluid compositions and the composition of the follicular fluid as the oocyte's micro-environment, affecting its development and subsequent embryo quality. DESIGN, PARTICIPANTS AND SETTING In this prospective cohort study, patient information, fertility treatment outcome data, follicular fluid and serum were obtained from women undergoing ART. Patients were categorized according to their BMI (kg/m(2)) as normal (n = 60), overweight (n = 26) or obese (n = 20). Serum and follicular fluid samples were analyzed for urea, total protein, albumin, cholesterol, high-density lipoprotein cholesterol, triglycerides, non-esterified fatty acids, apolipoprotein A1, apolipoprotein B, glucose, lactate, C-reactive protein, insulin-like growth factor -1 (IGF-1), IGF-binding protein 3 (only in follicular fluid), free carnitine and total carnitine. Metabolite concentrations in serum and follicular fluid samples were correlated and were associated with BMI and fertility treatment outcome. MAIN RESULTS Most serum metabolite differences between patients were reflected in the follicular fluid (P < 0.05). Follicular fluid apolipoprotein A1 and follicular fluid total protein concentrations negatively affected oocyte quality parameters (P < 0.05). However, overall BMI-related associations were poor. BIAS, CONFOUNDING AND OTHER REASONS FOR CAUTION In this study, we included every patient willing to participate. Within this cohort, women with a BMI transcending 35 kg/m(2) were scarce (n = 2), because extremely overweight women are mostly advised to lose weight before starting ART. Furthermore, the number of patients in each BMI group was different, possibly masking associations between the metabolic composition of serum and follicular fluid and oocyte quality parameters. GENERALIZABILITY TO OTHER POPULATIONS There were significant associations indicating that metabolic changes in the serum are reflected in the follicular fluid, potentially affecting oocyte quality, irrespective of the patient's BMI. For ethical reasons, this study only focused on women already in need of artificial reproductive treatment. From a metabolic point of view, we consider this cohort as a representative sample of all women of reproductive age. STUDY FUNDING This study was funded by the special research fund, university of Antwerp (BOF UA). None of the authors has any conflict of interest to declare.
Collapse
Affiliation(s)
- S D M Valckx
- Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
216
|
Bermejo-Alvarez P, Rosenfeld CS, Roberts RM. Effect of maternal obesity on estrous cyclicity, embryo development and blastocyst gene expression in a mouse model. Hum Reprod 2012; 27:3513-22. [PMID: 23001779 DOI: 10.1093/humrep/des327] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
STUDY QUESTION Does maternal obesity affect estrous cyclicity, embryo development and blastocyst gene expression in mice? SUMMARY ANSWER Maternal obesity alters estrous cyclicity and causes the down-regulation of two key metabolite receptors (Slc2a1 and Ldlr) in blastocysts recovered from diet-induced obese females, but embryo development is not affected. WHAT IS KNOWN ALREADY Maternal obesity reduces fertility because of effects in the periconception period, but its negative influence is on estrous cyclicity, oocyte quality or embryo development. STUDY DESIGN, SIZE AND DURATION This was a randomized study based on a mouse model for obesity. Twenty-one outbred NIH Swiss mice were used and obesity was induced by a diet high in fat administered for 12 weeks prior to breeding to control males. MATERIAL, SETTING AND METHODS Females were fed either a control diet (C, n = 9) or a diet high in fat [diet-induced obesity (DiO), n = 12] for 12 weeks, and were then co-housed with fertile males. Mice that failed to breed during 20 consecutive days were considered infertile. Control and diet-induced obese females that demonstrated vaginal plugs were euthanized 3.5 days after mating, blood was sampled for glucose and hormone measurements, corpora lutea counted and embryos recovered; the relative mRNA abundance of 11 candidate genes was determined in blastocysts by qPCR. MAIN RESULTS AND THE ROLE OF CHANCE Five DiO females failed to breed and displayed anovulatory ovaries (DiOI), whereas the other seven DiO females (DiOF) could breed, albeit over an extended period compared with controls. DiOF weighed significantly less than DiOI. Both groups had elevated serum insulin compared with C, although blood glucose level was only significantly higher than that in controls in the infertile DiOI group. Adiponectin was lower in the DiOI and leptin higher in both the DiOI and DiOF mice than in C. DiOF ovulated the same number of oocytes as C, and embryo development to blastocyst was normal. The expression of genes encoding metabolic hormone receptors (Insr, Igf1r, Igf2r, Adipor1, Adipor2 and Lepr) and key metabolic enzymes (Gapdh, Cpt1a and Sod2) did not differ between DiOF and C blastocysts, but that of metabolite receptors (Slc2a1 and Ldr) was down-regulated in DiOF. To limit the role of chance, the experiments were conducted in a defined laboratory setting with the proper controls, and the animals were randomly assigned to each experimental group. Moreover, a P-value of < 0.05 was chosen to determine whether the differences observed between the groups were statistically significant. LIMITATIONS AND REASONS FOR CAUTION The results obtained may not fully extrapolate to humans. Also, as follicular activity was not monitored while breeding, so the extended breeding period for DiOF group might be explained by behavioral abnormalities occurring in normal cycling animals. WIDER IMPLICATIONS OF THE FINDINGS DiO alters the estrous cycle in the mouse model and demonstrates a role of obesity in infertility. The data also suggest that in an outbred, genetically diverse population, such as the human, individual susceptibility to obesity and associated infertility induced by diet exists. The apparently normal development to blastocyst observed in fertile, obese females suggests that preimplantation embryos can resist potentially adverse outcomes caused by an oversupply of fatty acids and glucose under in vivo conditions. This metabolic plasticity may, in part, be due to an ability to down-regulate metabolite transporters, thereby preventing excessive nutrient uptake. STUDY FUNDING/COMPETING INTEREST(S) The research was supported by funds from the University of Missouri, grants from the National Institutes of Health and by a fellowship from the Lalor Foundation. There were no competing interests. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Pablo Bermejo-Alvarez
- Bond Life Sciences Center, University of Missouri, 263a Life Sciences Center, 1201 Rollins St, Columbia, MO 65211, USA.
| | | | | |
Collapse
|
217
|
Machtinger R, Combelles CMH, Missmer SA, Correia KF, Fox JH, Racowsky C. The association between severe obesity and characteristics of failed fertilized oocytes. Hum Reprod 2012; 27:3198-207. [PMID: 22968161 DOI: 10.1093/humrep/des308] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION Is the cytoskeletal and chromosomal organization of failed fertilized oocytes from severely obese patients (BMI ≥ 35 kg/m²) altered compared with that in patients with normal BMI (BMI 18.5-24.9 kg/m²)? SUMMARY ANSWER Compared with normal BMI patients, severe obesity was associated with a greater prevalence of spindle anomalies and non-aligned chromosomes in failed fertilized oocytes. WHAT IS KNOWN AND WHAT THIS PAPER ADDS Obesity is associated with poor reproductive outcomes, but little is known regarding the underlying mechanisms. To address potential mechanisms, our study compared the cytoskeletal and chromosome organization in failed fertilized oocytes from severely obese and normal BMI patients. DESIGN The study population was drawn from IVF patients treated in a hospital-based infertility clinic between February 2010 and July 2011. The prevalence of meiotic spindle and chromosome alignment anomalies in failed fertilized oocytes from patients with severe obesity (i.e. Class II and III; BMI 35.0-50.1 kg/m²) was compared with those from patients with normal BMI (BMI 18.5-24.9 kg/m²). Oocytes were fixed and then labeled for tubulin, actin and chromatin. Spindle number and integrity, as well as chromosome alignment, were assessed using immunofluorescence microscopy and, in some cases, confocal microscopy. Generalized estimating equations were applied, which account for the correlation among oocytes from the same patient to estimate odds ratio (OR), 95% confidence intervals (CIs) and two-sided Wald P-values. Models were adjusted for continuous age at cycle start, cycle type (IVF or ICSI) and polycystic ovarian syndrome (PCOS) a priori. PARTICIPANTS AND SETTING University-affiliated infertility clinic. A total of 276 oocytes that failed to fertilize from 137 patients were evaluated: 105 oocytes from severely obese women (n = 47) and 171 oocytes from normal BMI patients (n = 90). MAIN RESULTS AND THE ROLE OF CHANCE (i) Significantly more oocytes from the severely obese group exhibited two spindles compared with those from the normal BMI group (58.9 versus 35.1%; OR = 2.68, CI = 1.39-5.15, P-value = 0.003). (ii) Among oocytes with a single spindle, those from severely obese patients showed a significantly higher prevalence of disarranged spindles with non-aligned chromosomes compared with those from normal BMI patients (28.6 versus 8.6%; OR = 4.58, CI = 1.05-19.86, P-value = 0.04). BIAS, CONFOUNDING AND OTHER REASONS FOR CAUTION Inclusion of only failed fertilized oocytes, small sample size, unknown factors such as non-PCOS comorbidity. GENERALIZABILITY TO OTHER POPULATIONS For this study, by design, it is unclear whether the findings are generalizable to successfully fertilized oocytes, and whether this oocyte-level influence of obesity is generalizable to infertile women who do not undergo stimulation or, more broadly, to spontaneous conceptions in fertile women. STUDY FUNDING/COMPETING INTEREST(S) none. TRIAL REGISTRATION NUMBER n/a.
Collapse
Affiliation(s)
- Ronit Machtinger
- Department of Obstetrics, Gynecology and Reproductive Biology, Brigham & Women's Hospital and Harvard Medical School, 02115 Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
218
|
Dunning KR, Robker RL. Promoting lipid utilization with l-carnitine to improve oocyte quality. Anim Reprod Sci 2012; 134:69-75. [DOI: 10.1016/j.anireprosci.2012.08.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
219
|
Wu LLY, Norman RJ, Robker RL. The impact of obesity on oocytes: evidence for lipotoxicity mechanisms. Reprod Fertil Dev 2012; 24:29-34. [PMID: 22394715 DOI: 10.1071/rd11904] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Obesity can have detrimental effects on pregnancy rates in natural conceptions and also in women undergoing IVF or intracytoplasmic sperm injection (ICSI). This review summarises the most recent clinical literature investigating whether obesity impacts oocyte quality and early embryo growth. In other tissues, obesity leads to lipotoxicity responses including endoplasmic reticulum stress, mitochondrial dysfunction and apoptosis. Recent reports indicate that lipotoxicity is a mechanism by which obesity may impact oocyte quality.
Collapse
Affiliation(s)
- Linda L-Y Wu
- The Robinson Institute, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia
| | | | | |
Collapse
|
220
|
Women with reduced ovarian reserve or advanced maternal age have an altered follicular environment. Fertil Steril 2012; 98:986-94.e1-2. [PMID: 22818290 DOI: 10.1016/j.fertnstert.2012.06.025] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 06/13/2012] [Accepted: 06/13/2012] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To determine whether altered follicular environment is associated with ovarian reserve or maternal age. DESIGN Prospective study examining follicular fluid (FF) composition and follicular cell metabolism. SETTING University research department and private IVF clinic. PATIENT(S) Women (n = 54) undergoing routine IVF treatment were allocated to one of three groups based on ovarian reserve and maternal age. INTERVENTION(S) Surplus FF, granulosa cells (GC), and cumulus cells (CC) were collected. MAIN OUTCOME MEASURE(S) Follicular fluid concentrations of carbohydrates, hormones, and selected ions. Metabolic analysis and gene expression of GCs and CCs. RESULT(S) Compared to women <35 years with normal ovarian reserve, FF glucose levels were significantly decreased and lactate and progesterone (P4) concentrations significantly increased in women with reduced ovarian reserve or advanced maternal age, whereas GC and CC glucose uptake, lactate production, and phosphofructokinase platelet gene expression were significantly increased. Granulosa cell P4 production from women with reduced ovarian reserve or advanced maternal age was decreased; however, in CCs the reverse was observed with increased gene expression in P4 receptor, prostaglandin E receptor-2, cytosolic phospholipase A2, and tumor protein 53. CONCLUSION(S) Women with either reduced ovarian reserve or advanced maternal age have altered follicular cell metabolism, FF metabolites, and P4 production. This perturbed environment may be responsible for impaired oocyte developmental competence and subsequent embryo development.
Collapse
|
221
|
Agarwal A, Aponte-Mellado A, Premkumar BJ, Shaman A, Gupta S. The effects of oxidative stress on female reproduction: a review. Reprod Biol Endocrinol 2012; 10:49. [PMID: 22748101 PMCID: PMC3527168 DOI: 10.1186/1477-7827-10-49] [Citation(s) in RCA: 894] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 06/06/2012] [Indexed: 12/16/2022] Open
Abstract
Oxidative stress (OS), a state characterized by an imbalance between pro-oxidant molecules including reactive oxygen and nitrogen species, and antioxidant defenses, has been identified to play a key role in the pathogenesis of subfertility in both males and females. The adverse effects of OS on sperm quality and functions have been well documented. In females, on the other hand, the impact of OS on oocytes and reproductive functions remains unclear. This imbalance between pro-oxidants and antioxidants can lead to a number of reproductive diseases such as endometriosis, polycystic ovary syndrome (PCOS), and unexplained infertility. Pregnancy complications such as spontaneous abortion, recurrent pregnancy loss, and preeclampsia, can also develop in response to OS. Studies have shown that extremes of body weight and lifestyle factors such as cigarette smoking, alcohol use, and recreational drug use can promote excess free radical production, which could affect fertility. Exposures to environmental pollutants are of increasing concern, as they too have been found to trigger oxidative states, possibly contributing to female infertility. This article will review the currently available literature on the roles of reactive species and OS in both normal and abnormal reproductive physiological processes. Antioxidant supplementation may be effective in controlling the production of ROS and continues to be explored as a potential strategy to overcome reproductive disorders associated with infertility. However, investigations conducted to date have been through animal or in vitro studies, which have produced largely conflicting results. The impact of OS on assisted reproductive techniques (ART) will be addressed, in addition to the possible benefits of antioxidant supplementation of ART culture media to increase the likelihood for ART success. Future randomized controlled clinical trials on humans are necessary to elucidate the precise mechanisms through which OS affects female reproductive abilities, and will facilitate further explorations of the possible benefits of antioxidants to treat infertility.
Collapse
Affiliation(s)
- Ashok Agarwal
- Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | | | - Beena J Premkumar
- Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Amani Shaman
- Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Sajal Gupta
- Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
222
|
Zander-Fox DL, Henshaw R, Hamilton H, Lane M. Does obesity really matter? The impact of BMI on embryo quality and pregnancy outcomes after IVF in women aged ≤38 years. Aust N Z J Obstet Gynaecol 2012; 52:270-6. [DOI: 10.1111/j.1479-828x.2012.01453.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
223
|
Ou XH, Li S, Wang ZB, Li M, Quan S, Xing F, Guo L, Chao SB, Chen Z, Liang XW, Hou Y, Schatten H, Sun QY. Maternal insulin resistance causes oxidative stress and mitochondrial dysfunction in mouse oocytes. Hum Reprod 2012; 27:2130-45. [DOI: 10.1093/humrep/des137] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
224
|
Wallace M, Cottell E, Gibney MJ, McAuliffe FM, Wingfield M, Brennan L. An investigation into the relationship between the metabolic profile of follicular fluid, oocyte developmental potential, and implantation outcome. Fertil Steril 2012; 97:1078-84.e1-8. [DOI: 10.1016/j.fertnstert.2012.01.122] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 01/01/2012] [Accepted: 01/25/2012] [Indexed: 12/01/2022]
|
225
|
Purcell SH, Chi MM, Lanzendorf S, Moley KH. Insulin-stimulated glucose uptake occurs in specialized cells within the cumulus oocyte complex. Endocrinology 2012; 153:2444-54. [PMID: 22408172 PMCID: PMC3339650 DOI: 10.1210/en.2011-1974] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The oocyte exists within the mammalian follicle surrounded by somatic cumulus cells. These cumulus cells metabolize the majority of the glucose within the cumulus oocyte complex and provide energy substrates and intermediates such as pyruvate to the oocyte. The insulin receptor is present in cumulus cells and oocytes; however, it is unknown whether insulin-stimulated glucose uptake occurs in either cell type. Insulin-stimulated glucose uptake is thought to be unique to adipocytes, skeletal and cardiac muscle, and the blastocyst. Here, we show for the first time that many of the components required for insulin signaling are present in both cumulus cells and oocytes. We performed a set of experiments on mouse cumulus cells and oocytes and human cumulus cells using the nonmetabolizable glucose analog 2-deoxy-d-glucose to measure basal and insulin-stimulated glucose uptake. We show that insulin-stimulated glucose uptake occurs in both compact and expanded cumulus cells of mice, as well as in human cumulus cells. Oocytes, however, do not display insulin-stimulated glucose uptake. Insulin-stimulated glucose uptake in cumulus cells is mediated through phosphatidylinositol 3-kinase signaling as shown by inhibition of insulin-stimulated glucose uptake and Akt phosphorylation with the specific phosphatidylinositol 3-kinase inhibitor, LY294002. To test the effect of systemic in vivo insulin resistance on insulin sensitivity in the cumulus cell, cumulus cells from high fat-fed, insulin-resistant mice and women with polycystic ovary syndrome were examined. Both sets of cells displayed blunted insulin-stimulated glucose uptake. Our studies identify another tissue that, through a classical insulin-signaling pathway, demonstrates insulin-stimulated glucose uptake. Moreover, these findings suggest insulin resistance occurs in these cells under conditions of systemic insulin resistance.
Collapse
Affiliation(s)
- Scott H Purcell
- Department of Obstetrics and Gynecology, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA.
| | | | | | | |
Collapse
|
226
|
Yang X, Wu LL, Chura LR, Liang X, Lane M, Norman RJ, Robker RL. Exposure to lipid-rich follicular fluid is associated with endoplasmic reticulum stress and impaired oocyte maturation in cumulus-oocyte complexes. Fertil Steril 2012; 97:1438-43. [PMID: 22440252 DOI: 10.1016/j.fertnstert.2012.02.034] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 02/23/2012] [Accepted: 02/23/2012] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To determine whether the high lipid content of human follicular fluid influences oocyte maturation. DESIGN Mouse oocytes as substitutes for human oocytes were exposed to follicular fluids of differing lipid content with outcome monitoring. SETTING Private infertility clinic and university laboratory. PATIENT(S) Seventy-four women seeking assisted reproduction, and gonadotropin-stimulated mice. INTERVENTION(S) Assay of follicular fluids for triglyceride and free fatty acids, and stimulation of mouse cumulus-oocyte complexes (COCs) to maturity in vitro in the presence of lipid-rich or lipid-poor follicular fluid. MAIN OUTCOME MEASURE(S) Oocyte lipid content, expression of endoplasmic reticulum stress marker genes, and oocyte maturation assessed in mouse COCs exposed to lipid-rich follicular fluid were compared with complexes exposed to lipid-poor follicular fluid and complexes matured in vivo. RESULT(S) Follicular fluids were obtained from women of known body mass index undergoing oocyte aspiration at a private infertility clinic, and the follicular fluids were assayed for triglyceride and free fatty acids; those with the highest and lowest levels of these lipids were selected. The mouse COCs exposed to lipid-rich follicular fluid during their maturation had increased oocyte lipid content, induction of endoplasmic reticulum stress markers, and impaired oocyte nuclear maturation. CONCLUSION(S) Increased body mass index is associated with elevated triglycerides and free fatty acids in ovarian follicular fluid. Maturation within this lipid-rich environment is detrimental to oocytes.
Collapse
Affiliation(s)
- Xing Yang
- Reproductive Medical Center, Sixth Affiliated Hospital of Sun Yan-sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
227
|
Wu LL, Russell DL, Norman RJ, Robker RL. Endoplasmic reticulum (ER) stress in cumulus-oocyte complexes impairs pentraxin-3 secretion, mitochondrial membrane potential (DeltaPsi m), and embryo development. Mol Endocrinol 2012; 26:562-73. [PMID: 22383462 DOI: 10.1210/me.2011-1362] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Fatty acids such as palmitic acid at high levels are known to induce endoplasmic reticulum (ER) stress and lipotoxicity in numerous cell types and thereby contribute to cellular dysfunctions in obesity. To understand the impact of high fatty acids on oocytes, ER stress and lipotoxicity were induced in mouse cumulus-oocyte complexes during in vitro maturation using the ER Ca(2+) channel blocker thapsigargin or high physiological levels of palmitic acid; both of which significantly induced ER stress marker genes (Atf4, Atf6, Xbp1s, and Hspa5) and inositol-requiring protein-1α phosphorylation, demonstrating an ER stress response that was reversible with the ER stress inhibitor salubrinal. Assessment of pentraxin-3, an extracellular matrix protein essential for fertilization, by immunocytochemistry and Western blotting showed dramatically impaired secretion concurrent with ER stress. Mitochondrial activity in oocytes was assessed by 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide staining of inner mitochondrial membrane potential, and oocytes matured in thapsigargin or high-dose palmitic acid had significantly reduced mitochondrial activity, reduced in vitro fertilization rates, and were slower to develop to blastocysts. The deficiencies in protein secretion, mitochondrial activity, and oocyte developmental competence were each normalized by salubrinal, demonstrating that ER stress is a key mechanism mediating fatty acid-induced defects in oocyte developmental potential.
Collapse
Affiliation(s)
- Linda L Wu
- The Robinson Institute, School of Paediatrics and Reproductive Health, Research Centre for Reproductive Health, University of Adelaide, Adelaide, South Australia 5005, Australia
| | | | | | | |
Collapse
|
228
|
Silva E, Paczkowski M, Krisher RL. The effect of leptin on maturing porcine oocytes is dependent on glucose concentration. Mol Reprod Dev 2012; 79:296-307. [PMID: 22368147 DOI: 10.1002/mrd.22029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 01/27/2012] [Indexed: 12/23/2022]
Abstract
Increased body weight is often accompanied by increased circulating levels of leptin and glucose, which alters glucose metabolism in various tissues, including perhaps the oocyte. Alteration of glucose metabolism impacts oocyte function and may contribute to the subfertility often associated with obese individuals. The objective of this study was to determine the effect of leptin (0, 10, and 100 ng/ml) on the oocyte and cumulus cells during in vitro maturation under differing glucose concentrations. We examined the effects of leptin on oocyte maturation, blastocyst development, and/or gene expression in oocytes and cumulus cells (IRS1, IGF1, PPARγ, IL6, GLUT1) in a physiological glucose (2 mM) and high glucose (50 mM) environment. We also evaluated the effect of leptin on glucose metabolism via glycolysis and the pentose phosphate pathway. In a physiological glucose environment, leptin did not have an influence on oocyte maturation, blastocyst development, or oocyte gene expression. Expression of GLUT1 in cumulus cells was downregulated with 100 ng/ml leptin treatment, but did not affect oocyte glucose metabolism. In a high glucose environment, oocyte maturation and glycolysis were decreased, but in the presence of 100 ng/ml leptin, these parameters were improved to levels similar to control. This effect is potentially mediated by an upregulation of oocyte IRS1 and a correction of cumulus cell IGF1 expression. The present study demonstrates that in a physiological glucose concentration, leptin plays a negligible role in oocyte function. However, leptin appears to modulate the deleterious impact of a high glucose environment on oocyte function.
Collapse
Affiliation(s)
- Elena Silva
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | | | | |
Collapse
|
229
|
Fleming TP, Lucas ES, Watkins AJ, Eckert JJ. Adaptive responses of the embryo to maternal diet and consequences for post-implantation development. Reprod Fertil Dev 2012; 24:35-44. [DOI: 10.1071/rd11905] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Maternal periconceptional (PC) nutrition, coupled with maternal physiological condition, can impact on reproductive performance and potential across mammalian species. Oocyte quality and embryo development are affected adversely by either nutrient restriction or excess. Moreover, the quality of maternal PC nutrition can have lasting effects through fetal development and postnatally into adulthood. Chronic disease, notably cardiovascular and metabolic disease, and abnormal behaviour have been identified in adult offspring in small and large animal models of PC nutrient restriction. These long-term effects associate with compensatory responses that begin from the time of early embryo development. This review assesses the field of PC nutrition in vivo on short- and long-term developmental consequences in rodent and ruminant models and considers the implications for human health.
Collapse
|
230
|
Leroy JLMR, Rizos D, Sturmey R, Bossaert P, Gutierrez-Adan A, Van Hoeck V, Valckx S, Bols PEJ. Intrafollicular conditions as a major link between maternal metabolism and oocyte quality: a focus on dairy cow fertility. Reprod Fertil Dev 2012; 24:1-12. [DOI: 10.1071/rd11901] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Reduced oocyte and embryo quality are recognised as major factors in the problem of disappointing fertility in high producing dairy cows. This review aims to shed more light on the importance of the intrafollicular environment in the subfertility problem in dairy cows. Metabolic disturbances associated with negative energy balance (NEB) early postpartum are associated with ovarian dysfunction. Changes in the growth pattern of the ovarian follicle during a period of NEB can indirectly affect oocyte quality. Furthermore, a maternal metabolic disorder (linked with NEB or nutritionally induced) may alter the endocrine and biochemical composition of the follicular fluid, the micro-environment of the growing and maturing female gamete. The maturing oocyte is very sensitive to any perturbation in its direct environment and in vitro maturation models revealed that some of these metabolic changes reduce the oocyte’s developmental competence. Also, embryo quality is significantly reduced due to maturation in adverse conditions. Well balanced and timed oocyte metabolism and gene expression are crucial to safeguard an optimal oocyte development. In that perspective, metabolome and transcriptome parameters of the oocyte may serve to predict reproductive success rates. Finally, there is growing evidence that adverse conditions for oocyte growth and maturation may also jeopardise the health and performance of the offspring.
Collapse
|
231
|
OLA SI, SUN QY. Factors Influencing the Biochemical Markers for Predicting Mammalian Oocyte Quality. J Reprod Dev 2012; 58:385-92. [DOI: 10.1262/jrd.11-084h] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Safiriyu Idowu OLA
- Department of Animal Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Qing-Yuan SUN
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
232
|
Poston L, Igosheva N, Mistry HD, Seed PT, Shennan AH, Rana S, Karumanchi SA, Chappell LC. Role of oxidative stress and antioxidant supplementation in pregnancy disorders. Am J Clin Nutr 2011; 94:1980S-1985S. [PMID: 21613560 DOI: 10.3945/ajcn.110.001156] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Oxidative stress is widely implicated in failed reproductive performance, including infertility, miscarriage, diabetes-related congenital malformations, and preeclampsia. Maternal obesity is a strong risk factor for preeclampsia, and in a recent study we observed oxidative stress in the oocytes of obese animals before pregnancy as well as in early-stage embryos. This adds to the growing evidence that investigators need to focus more on the preconceptual period in efforts to prevent pregnancy disorders, including those related to oxidative stress. Our research has also focused on the role of free radicals and antioxidant capacity in preeclampsia. By measuring markers of lipid peroxidation and antioxidant capacity, we obtained unequivocal evidence for oxidative stress in this disorder. Partial failure of the process of placentation has been implicated, and recent findings suggest that ischemia-reperfusion in the placenta may contribute to oxidative stress in trophoblasts. Endoplasmic reticulum stress in the placenta may also play a role. Randomized controlled trials have been conducted by our group as well as others to determine whether early supplementation with vitamins C and E in women at risk of preeclampsia is beneficial, but these trials have shown no evidence that these supplements can prevent preeclampsia. Whether this indicates that an inappropriate antioxidant strategy was used or supplementation was administered too late in gestation to be beneficial is not known. Other potential approaches for preventing preeclampsia through amelioration of oxidative stress include the use of supplements in the preconceptual period, selenium supplements, antiperoxynitrite strategies, and statins.
Collapse
Affiliation(s)
- Lucilla Poston
- Division of Women's Health, King's College London, Kings Health Partners-Women's Health, Clinical Academic Grouping, London, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
233
|
Moran LJ, Dodd J, Nisenblat V, Norman RJ. Obesity and reproductive dysfunction in women. Endocrinol Metab Clin North Am 2011; 40:895-906. [PMID: 22108286 DOI: 10.1016/j.ecl.2011.08.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Overweight and obesity are significant and increasing health problems associated with increased risks of morbidity, quality of life, and metabolic and reproductive health consequences. In women, being overweight or obese is associated with impaired fertility and decreased chance of conception both in natural and assisted reproductive technology births. During pregnancy, overweight and obesity are associated with increased risk of adverse maternal and infant health outcomes. Attention to weight loss before conception may improve fertility and maternal and infant health outcomes during pregnancy.
Collapse
Affiliation(s)
- Lisa J Moran
- The Robinson Institute, Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, University of Adelaide, South Australia.
| | | | | | | |
Collapse
|
234
|
Purcell SH, Moley KH. The impact of obesity on egg quality. J Assist Reprod Genet 2011; 28:517-24. [PMID: 21625966 DOI: 10.1007/s10815-011-9592-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 05/23/2011] [Indexed: 11/29/2022] Open
Abstract
Obesity in women is a concern in many countries. This causes numerous health issues; however, this review focuses on the impact of obesity on women's reproduction, and in particular the oocyte. Data from infertility clinics and experimental animal models that address the effects of obesity are presented. Bidirectional communication and metabolic support from the surrounding cumulus cells are critical for oocyte development, and the impact of obesity on these cells is also addressed. Both oocyte maturation and metabolism are impaired due to obesity, negatively impacting further development. In addition to reproductive hormones, obesity induced elevations in insulin, glucose, or free fatty acids, and changes in adipokines appear to impact the developmental competence of the oocyte. The data indicate that any one of these hormones or metabolites can impair oocyte developmental competence in vivo, and the combination of all of these factors and their interactions are the subject of ongoing investigations.
Collapse
Affiliation(s)
- Scott H Purcell
- Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | | |
Collapse
|
235
|
Abstract
Weight, fat mass and obesity have been shown to play a major role in female reproduction. Obese women have a greater risk than nonobese women of infertility and they fail to become pregnant in both natural and assisted conception cycles. This cannot be explained only by their lack of ovulation. There are several potential mechanisms. On one hand, the endometrium seems to be partially responsible for this low fecundity in obese women. On the other hand, the oocyte seems to be implied. In a model of obese mouse, maternal obesity prior to conception is associated with altered mitochondria in mouse oocytes and an increased generation of reactive oxygen species (ROS). Furthermore, compared with controls, obese mice have significantly more decreased embryonic IGF-IR staining, smaller fetuses and smaller pups. In this model, all weaned pups have been fed with a regular diet. At 13 weeks, pups delivered from obese mice were significantly larger, and these pups demonstrated early development of a metabolic-type syndrome. These findings suggest that maternal obesity has adverse effects as early as the oocyte and preimplantation embryo stages and that these effects may contribute to lasting morbidity in offspring, underscoring the importance of optimal maternal weight and nutrition before conception.
Collapse
|
236
|
Jungheim ES, Macones GA, Odem RR, Patterson BW, Lanzendorf SE, Ratts VS, Moley KH. Associations between free fatty acids, cumulus oocyte complex morphology and ovarian function during in vitro fertilization. Fertil Steril 2011; 95:1970-4. [PMID: 21353671 PMCID: PMC3080431 DOI: 10.1016/j.fertnstert.2011.01.154] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 01/25/2011] [Accepted: 01/26/2011] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To determine if follicular free fatty acid (FFA) levels are associated with cumulus oocyte complex (COC) morphology. DESIGN Prospective cohort study. SETTING University in vitro fertilization (IVF) practice. PATIENT(S) A total of 102 women undergoing IVF. INTERVENTION(S) Measurement of FFAs in serum and ovarian follicular fluid. MAIN OUTCOME MEASURE(S) Total and specific follicular and serum FFA levels, correlations between follicular and serum FFAs, and associations between follicular FFA levels and markers of oocyte quality, including COC morphology. RESULT(S) Predominant follicular fluid and serum FFAs were oleic, palmitic, linoleic, and stearic acids. Correlations between follicular and serum FFA concentrations were weak (r=0.252, 0.288, 0.236, 0.309, respectively for specific FFAs; r=0.212 for total FFAs). A receiver operating characteristic curve determined total follicular FFAs≥0.232 μmol/mL distinguished women with a lower versus higher percentage of COCs with favorable morphology. Women with elevated follicular FFAs (n=31) were more likely to have COCs with poor morphology than others (n=71; OR 3.3, 95% CI1.2-9.2). This relationship held after adjusting for potential confounders, including age, body mass index, endometriosis, and amount of gonadotropin administered (β=1.2; OR 3.4, 95% CI 1.1-10.4). CONCLUSION(S) Elevated follicular FFA levels are associated with poor COC morphology. Further work is needed to determine what factors influence follicular FFA levels and if these factors impact fertility.
Collapse
Affiliation(s)
- Emily S Jungheim
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri 63108, USA.
| | | | | | | | | | | | | |
Collapse
|
237
|
Cardozo E, Pavone ME, Hirshfeld-Cytron JE. Metabolic syndrome and oocyte quality. Trends Endocrinol Metab 2011; 22:103-9. [PMID: 21277789 DOI: 10.1016/j.tem.2010.12.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 12/27/2010] [Accepted: 12/28/2010] [Indexed: 11/17/2022]
Abstract
Metabolic syndrome affects one in four women in the USA, and the incidence is rising every year. Metabolic syndrome is strongly associated with development of coronary artery disease and diabetes. Women of reproductive age are not spared from the complications of metabolic syndrome, which overlaps with obesity and polycystic ovary syndrome (PCOS), both of which are linked to infertility and poor reproductive outcome. Therefore, the relationship between the metabolic syndrome and reproductive dysfunction is an active area of study. In this review, we discuss the animal and human data available to determine if the abnormality is at the level of the ovary and/or endometrium, and discuss the underlying mechanisms causing the associated poor reproductive outcomes.
Collapse
Affiliation(s)
- Eden Cardozo
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 676 N St Clair Street Suite 1845 Chicago, IL 60611, Evanston, IL, USA
| | | | | |
Collapse
|
238
|
van den Berg IM, Eleveld C, van der Hoeven M, Birnie E, Steegers EAP, Galjaard RJ, Laven JSE, van Doorninck JH. Defective deacetylation of histone 4 K12 in human oocytes is associated with advanced maternal age and chromosome misalignment. Hum Reprod 2011; 26:1181-90. [PMID: 21349858 DOI: 10.1093/humrep/der030] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Chromosome segregation errors during human oocyte meiosis are associated with low fertility in humans and the incidence of these errors increases with advancing maternal age. Studies of mitosis and meiosis suggest that defective remodeling of chromatin plays a causative role in aneuploidy. We analyzed the histone deacetylation pattern during the final stages of human oocyte maturation to investigate whether defective epigenetic regulation of chromatin remodeling in human oocytes is related to maternal age and leads to segregation errors. METHODS Human surplus oocytes of different meiotic maturation stages [germinal vesicle (GV), metaphase (M)I and MII] were collected from standard IVF/ICSI treatments. Oocytes were analyzed for acetylation of different lysines of histone 4 (H4K5, H4K8, H4K12 and H4K16) and for α-tubulin. RESULTS Human GV oocytes had an intense staining of the chromatin for all four histone 4 lysine acetylations. MI and MII stage oocytes showed either normal deacetylation or various amounts of defective histone deacetylation. Residual H4K12 acetylation was more frequently found in oocytes obtained from older women, with a significant correlation between defective deacetylation and maternal age (r = 0.185, P = 0.007). Eighty-eight percent of the oocytes with residual acetylation had misaligned chromosomes, whereas only 33% of the oocytes that showed correct deacetylated chromatin had misaligned chromosomes (P < 0.001). CONCLUSIONS We conclude that defective deacetylation during human female meiosis increases with maternal age and is correlated with misaligned chromosomes. As chromosome misalignment predisposes to segregation errors, our data imply that defective regulation of histone deacetylation could be an important factor in age-related aneuploidy.
Collapse
Affiliation(s)
- I M van den Berg
- Department of Obstetrics and Gynaecology, Subdivision of Reproductive Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
239
|
Robker RL, Wu LLY, Yang X. Inflammatory pathways linking obesity and ovarian dysfunction. J Reprod Immunol 2011; 88:142-8. [PMID: 21333359 DOI: 10.1016/j.jri.2011.01.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 12/20/2010] [Accepted: 01/16/2011] [Indexed: 01/03/2023]
Abstract
This review summarizes some of the recent advances in obesity research and describes how we and others have built upon these findings to better understand the impact of obesity on granulosa cells, cumulus cells and oocytes within the ovaries of obese females. Obesity is associated with lipid accumulation in non-adipose tissue cells and the induction of oxidative stress and endoplasmic reticulum stress responses that are tightly linked with systemic inflammation. Analysis of ovarian cells and fluid of obese women indicates that these same mechanisms are activated in the ovary in response to obesity. Studies in mice support this and allow further dissection of the pathways by which diet-induced obesity contributes to changes in mitochondria and the endoplasmic reticulum. These studies are in their infancy but cumulatively provide basic information about the cellular mechanisms that may lead to the impaired ovulation and reduced oocyte developmental potential that is observed in obese females.
Collapse
Affiliation(s)
- Rebecca L Robker
- School of Paediatrics and Reproductive Health, Robinson Institute, University of Adelaide, Adelaide, South Australia 5005, Australia.
| | | | | |
Collapse
|
240
|
Yang X, Dunning KR, Wu LLY, Hickey TE, Norman RJ, Russell DL, Liang X, Robker RL. Identification of perilipin-2 as a lipid droplet protein regulated in oocytes during maturation. Reprod Fertil Dev 2010; 22:1262-71. [PMID: 20883652 DOI: 10.1071/rd10091] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 06/28/2010] [Indexed: 12/16/2022] Open
Abstract
Lipid droplet proteins regulate the storage and utilisation of intracellular lipids. Evidence is emerging that oocyte lipid utilisation impacts embryo development, but lipid droplet proteins have not been studied in oocytes. The aim of the present study was to characterise the size and localisation of lipid droplets in mouse oocytes during the periovulatory period and to identify lipid droplet proteins as potential biomarkers of oocyte lipid content. Oocyte lipid droplets, visualised using a novel method of staining cumulus-oocyte complexes (COCs) with BODIPY 493/503, were small and diffuse in oocytes of preovulatory COCs, but larger and more centrally located after maturation in response to ovulatory human chorionic gonadotrophin (hCG) in vivo, or FSH + epidermal growth factor in vitro. Lipid droplet proteins perilipin, perilipin-2, cell death-inducing DNA fragmentation factor 45-like effector (CIDE)-A and CIDE-B were detected in the mouse ovary by immunohistochemistry, but only perilipin-2 was associated with lipid droplets in the oocyte. In COCs, perilipin-2 mRNA and protein increased in response to ovulatory hCG. IVM failed to induce perilipin-2 mRNA, yet oocyte lipid content was increased in this context, indicating that perilipin-2 is not necessarily reflective of relative oocyte lipid content. Thus, perilipin-2 is a lipid droplet protein in oocytes and its induction in the COC concurrent with dynamic reorganisation of lipid droplets suggests marked changes in lipid utilisation during oocyte maturation.
Collapse
Affiliation(s)
- Xing Yang
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510080, China
| | | | | | | | | | | | | | | |
Collapse
|
241
|
Jungheim ES, Moley KH. Current knowledge of obesity's effects in the pre- and periconceptional periods and avenues for future research. Am J Obstet Gynecol 2010; 203:525-30. [PMID: 20739012 PMCID: PMC3718032 DOI: 10.1016/j.ajog.2010.06.043] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 05/18/2010] [Accepted: 06/17/2010] [Indexed: 11/21/2022]
Abstract
The prevalence of obesity is growing among reproductive-age women. This is concerning because obesity has significant health-related consequences. Aside from the long-term risks of diabetes, heart disease, and some types of cancer, obesity poses immediate threats for young women including subfertility and adverse early and late pregnancy outcomes. Epidemiologic and experimental studies demonstrate associations between prepregnancy obesity and poor reproductive outcomes; however, the mechanisms involved are poorly understood. We discuss current knowledge of the pathophysiology of obesity in early reproductive events and how these events may affect reproductive outcomes including fertility and miscarriage risk. We also discuss avenues for future research and interventions to improve reproductive outcomes for obese women.
Collapse
Affiliation(s)
- Emily S Jungheim
- Department of Obstetrics and Gynecology, Washington University, St Louis, MO, USA
| | | |
Collapse
|
242
|
Varghese AC, Ly KD, Corbin C, Mendiola J, Agarwal A. Oocyte developmental competence and embryo development: impact of lifestyle and environmental risk factors. Reprod Biomed Online 2010; 22:410-20. [PMID: 21388885 DOI: 10.1016/j.rbmo.2010.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 10/19/2010] [Accepted: 11/10/2010] [Indexed: 11/24/2022]
Abstract
Oocyte development is the end result of a sophisticated biological process that is hormonally regulated and produced by highly specialized cellular lines that differentiate in early embryo/fetal development. Embryo development is initially regulated by maternal transcripts until replaced by embryonic genomic expression. Then, an assortment of hormones and local environmental factors in various concentrations along the reproductive tract (e.g. fallopian tube, endometrial lining) provide the protection, nutrients and means of communication for the embryo to implant and develop. Both oocytes and embryos are susceptible to environmental, occupational and lifestyle exposures that can exert direct toxic effects and disrupt hormones. While some exposures may produce reversible changes, others, especially those damaging germinal cells in utero or during prepuberty, may result in permanent sequelae that continue in future generations. This article reviews the main factors that affect female fertility and their possible influence on human reproduction. Some lifestyles, xeno-oestrogens and heavy metals are already known to compromise female reproductive function. Nonetheless, many questions remain and little is known about the effect of many other factors on female fertility.
Collapse
Affiliation(s)
- Alex C Varghese
- Fertility Clinic and IVF Department, AMRI Medical Centre, Kolkata, India
| | - Kim D Ly
- Center for Reproductive Medicine, Glickman Urological and Kidney Institute and Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Cresandra Corbin
- Center for Reproductive Medicine, Glickman Urological and Kidney Institute and Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jaime Mendiola
- Public Health and Epidemiology Research Group, School of Medicine, University of Murcia, Murcia, Spain
| | - Ashok Agarwal
- Center for Reproductive Medicine, Glickman Urological and Kidney Institute and Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
243
|
Marquard KL, Stephens SM, Jungheim ES, Ratts VS, Odem RR, Lanzendorf S, Moley KH. Polycystic ovary syndrome and maternal obesity affect oocyte size in in vitro fertilization/intracytoplasmic sperm injection cycles. Fertil Steril 2010; 95:2146-9, 2149.e1. [PMID: 21071018 DOI: 10.1016/j.fertnstert.2010.10.026] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 09/09/2010] [Accepted: 10/13/2010] [Indexed: 11/30/2022]
Abstract
To determine the impact of maternal metabolic state on oocyte development in women undergoing in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI), we retrospectively analyzed a cohort of women with PCOS undergoing IVF/ICSI from 2008-2009 in a university-based fertility center. We determined that women with PCOS and obesity have smaller oocytes than control subjects, and that when further subdivided by body mass index, both PCOS and obesity independently influence oocyte size.
Collapse
Affiliation(s)
- Kerri L Marquard
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri 63108, USA
| | | | | | | | | | | | | |
Collapse
|
244
|
Wu LLY, Dunning KR, Yang X, Russell DL, Lane M, Norman RJ, Robker RL. High-fat diet causes lipotoxicity responses in cumulus-oocyte complexes and decreased fertilization rates. Endocrinology 2010; 151:5438-45. [PMID: 20861227 DOI: 10.1210/en.2010-0551] [Citation(s) in RCA: 231] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In obesity, accumulation of lipid in nonadipose tissues, or lipotoxicity, is associated with endoplasmic reticulum (ER) stress, mitochondrial dysfunction, and ultimately apoptosis. We have previously shown that obese women have increased triglycerides in follicular fluid; thus, the present study examined whether high-fat diet-induced obesity causes lipotoxicity in granulosa cells and the cumulus-oocyte complex (COC). Oocytes of mice fed a high-fat diet had dramatically increased lipid content and reduced mitochondrial membrane potential compared to those of mice fed a control diet. COCs from mice fed a high-fat diet had increased expression of ER stress marker genes ATF4 and GRP78. Apoptosis was increased in granulosa and cumulus cells of mice fed a high-fat diet. Mice fed a high-fat diet also exhibited increased anovulation and decreased in vivo fertilization rates. Thus, lipid accumulation, ER stress, mitochondrial dysfunction, and apoptosis are markedly increased in ovarian cells of mice fed a high-fat diet. ER stress markers were also analyzed in granulosa cells and follicular fluid from women with varying body mass indices (BMI). ATF4 was increased in granulosa cells and [Ca(2+)] in follicular fluid from obese women compared to nonobese women. These results indicate that lipotoxicity may be occurring in ovarian cells of obese women and may contribute to the reduced pregnancy rates observed in response to obesity.
Collapse
Affiliation(s)
- Linda Lin-Yan Wu
- School of Pediatrics and Reproductive Health, Robinson Institute, University of Adelaide, Adelaide, South Australia 5005, Australia
| | | | | | | | | | | | | |
Collapse
|
245
|
Farhi J, Ben-Haroush A, Sapir O, Fisch B, Ashkenazi J. High-quality embryos retain their implantation capability in overweight women. Reprod Biomed Online 2010; 21:706-11. [PMID: 20880747 DOI: 10.1016/j.rbmo.2010.06.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 06/23/2010] [Accepted: 06/30/2010] [Indexed: 11/30/2022]
Abstract
To assess the effect of obesity on implantation rate, pregnancy rate and course of pregnancy in young women undergoing IVF in whom only high-quality embryos were transferred, a cohort study included women attending the IVF unit in 2006-2007 with favourable parameters to achieve pregnancy (<38years, fewer than three IVF cycles, transfer of two high-quality embryos), grouped by body mass index (BMI). Of 230 women, 160 had a BMI ⩽25kg/m(2) (mean 21.6±2.2) and 73 had BMI >25kg/m(2) (mean 29.5±3.7). The overweight group had a higher consumption of gonadotrophins during stimulation. There were no between-group differences in treatment protocols, duration of gonadotrophin stimulation, maximal oestradiol concentrations, endometrial thickness and number of oocytes retrieved/fertilized, or in rates of pregnancy (51.3%, 52.1%), implantation (34.5%, 37.5%), multiple pregnancy, and abortion. Rate of gestational diabetes or pregnancy-induced hypertension was higher in the overweight group (23.3%, 8.2%; P=0.045). Within the overweight group, those with multiple pregnancies were at highest risk (31.3%, 6.9%; P=0.031). In conclusion, implantation and pregnancy rates are not compromised in overweight women when high-quality embryos are transferred. However, in overweight women, pregnancy complications remain high, mainly in those with multiple pregnancies.
Collapse
Affiliation(s)
- Jacob Farhi
- Infertility and IVF Unit, Department of Obstetrics and Gynecology, Helen Schneider Hospital for Women, Rabin Medical Center, Beilinson Campus, Petach Tikva 49100, Israel.
| | | | | | | | | |
Collapse
|
246
|
Dunning KR, Cashman K, Russell DL, Thompson JG, Norman RJ, Robker RL. Beta-oxidation is essential for mouse oocyte developmental competence and early embryo development. Biol Reprod 2010; 83:909-18. [PMID: 20686180 DOI: 10.1095/biolreprod.110.084145] [Citation(s) in RCA: 293] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Oocyte and embryo metabolism are closely linked with their subsequent developmental capacity. Lipids are a potent source of cellular energy, yet little is known about lipid metabolism during oocyte maturation and early embryo development. Generation of ATP from lipids occurs within mitochondria via beta-oxidation of fatty acids, with the rate-limiting step catalyzed by carnitine palmitoyl transferase I (CPT1B), a process also requiring carnitine. We sought to investigate the regulation and role of beta-oxidation during oocyte maturation and preimplantation development. Expression of Cpt1b mRNA, assessed by real-time RT-PCR in murine cumulus-oocyte complexes (COCs), increased following hormonal induction of oocyte maturation and ovulation in vivo with human chorionic gonadotropin (5 IU) and in embryos reaching the blastocyst stage. Beta-oxidation, measured by the production of (3)H(2)O from [(3)H]palmitic acid, was significantly increased over that in immature COCs following induction of maturation in vitro with epidermal growth factor (3 ng/ml) and follicle-stimulating hormone (50 mIU/ml). The importance of lipid metabolism for oocyte developmental competence and early embryo development was demonstrated by assessing the rate of embryo development following inhibition or upregulation of beta-oxidation with etomoxir (an inhibitor of CPT1B) or L-carnitine, respectively. Inhibition of beta-oxidation during oocyte maturation or zygote cleavage impaired subsequent blastocyst development. In contrast, L-carnitine supplementation during oocyte maturation significantly increased beta-oxidation, improved developmental competence, and in the absence of a carbohydrate energy supply, significantly increased 2-cell cleavage. Thus, carnitine is an important cofactor for developing oocytes, and fatty acids are an important energy source for oocyte and embryo development.
Collapse
Affiliation(s)
- Kylie R Dunning
- The Robinson Institute, School of Paediatrics and Reproductive Health, The University of Adelaide, Adelaide, South Australia, Australia.
| | | | | | | | | | | |
Collapse
|
247
|
Jungheim ES, Schoeller EL, Marquard KL, Louden ED, Schaffer JE, Moley KH. Diet-induced obesity model: abnormal oocytes and persistent growth abnormalities in the offspring. Endocrinology 2010; 151:4039-46. [PMID: 20573727 PMCID: PMC2940512 DOI: 10.1210/en.2010-0098] [Citation(s) in RCA: 257] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 05/20/2010] [Indexed: 12/25/2022]
Abstract
Associations between maternal obesity and adverse fetal outcomes are well documented, but the mechanisms involved are largely unknown. Most previous work has focused on postconceptional events, however, our laboratory has shown pre- and periconceptional aberrations in maternal glucose metabolism have adverse effects on oocytes and embryos that carry on to the fetus. To demonstrate effects of maternal obesity in the pre- and periconceptional periods, we compared reproductive tissues from diet-induced obese female mice to those of control mice. Ovaries were either stained for follicular apoptosis or dissected and evaluated for oocyte size and meiotic maturation. Mice were also mated and followed for reproductive outcomes including preimplantation embryonic IGF-I receptor (IGF-IR) immunostaining, midgestation fetal growth, and midgestational placental IGF receptor 2 (Igf2r) mRNA. Delivered pups were followed for growth and development of markers of metabolic syndrome. Compared with controls, obese mice had significantly more apoptotic ovarian follicles, smaller and fewer mature oocytes, decreased embryonic IGF-IR staining, smaller fetuses, increased placental Igf2r mRNA, and smaller pups. All weaned pups were fed a regular diet. At 13 wk pups delivered from obese mice were significantly larger, and these pups demonstrated glucose intolerance and increased cholesterol and body fat suggesting early development of a metabolic-type syndrome. Together, our findings suggest maternal obesity has adverse effects as early as the oocyte and preimplantation embryo stage and that these effects may contribute to lasting morbidity in offspring, underscoring the importance of optimal maternal weight and nutrition before conception.
Collapse
Affiliation(s)
- Emily S Jungheim
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | |
Collapse
|
248
|
Serke H, Bausenwein J, Hirrlinger J, Nowicki M, Vilser C, Jogschies P, Hmeidan FA, Blumenauer V, Spanel-Borowski K. Granulosa cell subtypes vary in response to oxidized low-density lipoprotein as regards specific lipoprotein receptors and antioxidant enzyme activity. J Clin Endocrinol Metab 2010; 95:3480-90. [PMID: 20444928 DOI: 10.1210/jc.2009-2654] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT The oxidized low-density lipoprotein (oxLDL) and its lectin-like oxLDL receptor-1 (LOX-1) are found in the follicular fluid and in granulosa cells. Lipoprotein receptors and antioxidant enzymes could differ in granulosa cell subtypes. OBJECTIVE Our aim was to reveal cell-specific responses under oxLDL treatment. DESIGN AND SETTING We conducted basic research at the Institute of Anatomy and the Clinic of Reproductive Medicine. PATIENTS Women undergoing in vitro fertilization therapy participated in the study. MAIN OUTCOME MEASURES Cultures of cytokeratin-positive/negative (CK(+)/CK(-)) granulosa cells and of cumulus cells were treated with 150 microg/ml oxLDL or native LDL under serum-free conditions for up to 36 h. Dead cells were determined by uptake of propidium iodide. LOX-1, toll-like receptor 4, and cluster of differentiation 36 (CD36) were examined in lysates by Western blots. The enzyme activities were determined in lysates and in supernatants. RESULTS Under oxLDL treatment, predominantly CK(+) cells underwent nonapoptotic cell death. Receptors showed a cell-specific pattern of up-regulation: toll-like receptor 4 in CK(+) cells, LOX-1 in CK(-) cells, and CD36 in cumulus cells. An antioxidant ranking occurred: superoxide dismutase activity in CK(+) cells, total glutathione in CK(-) cells, and catalase activity in cumulus cells. The supernatants of oxLDL-treated CK(+) cell cultures contained more catalase activity than in controls, whereas a moderate increase was noted for glutathione peroxidase (GPx) in supernatants of CK(-) and cumulus cells. CONCLUSIONS Catalase/GPx activity in the supernatants may be due to cell death or to secretion. Oxidative stress could be sensed by CK(+) cells and indicated by changes in catalase/GPx activity in the follicular fluid during ovarian disorders.
Collapse
Affiliation(s)
- H Serke
- Institute of Anatomy, University of Leipzig, Liebigstrasse 13, D-04103 Leipzig, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Current Opinion in Endocrinology, Diabetes & Obesity. Current world literature. Curr Opin Endocrinol Diabetes Obes 2010; 17:293-312. [PMID: 20418721 DOI: 10.1097/med.0b013e328339f31e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
250
|
Igosheva N, Abramov AY, Poston L, Eckert JJ, Fleming TP, Duchen MR, McConnell J. Maternal diet-induced obesity alters mitochondrial activity and redox status in mouse oocytes and zygotes. PLoS One 2010; 5:e10074. [PMID: 20404917 PMCID: PMC2852405 DOI: 10.1371/journal.pone.0010074] [Citation(s) in RCA: 351] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Accepted: 03/17/2010] [Indexed: 11/23/2022] Open
Abstract
The negative impact of obesity on reproductive success is well documented but the stages at which development of the conceptus is compromised and the mechanisms responsible for the developmental failure still remain unclear. Recent findings suggest that mitochondria may be a contributing factor. However to date no studies have directly addressed the consequences of maternal obesity on mitochondria in early embryogenesis. Using an established murine model of maternal diet induced obesity and a live cell dynamic fluorescence imaging techniques coupled with molecular biology we have investigated the underlying mechanisms of obesity-induced reduced fertility. Our study is the first to show that maternal obesity prior to conception is associated with altered mitochondria in mouse oocytes and zygotes. Specifically, maternal diet-induced obesity in mice led to an increase in mitochondrial potential, mitochondrial DNA content and biogenesis. Generation of reactive oxygen species (ROS) was raised while glutathione was depleted and the redox state became more oxidised, suggestive of oxidative stress. These altered mitochondrial properties were associated with significant developmental impairment as shown by the increased number of obese mothers who failed to support blastocyst formation compared to lean dams. We propose that compromised oocyte and early embryo mitochondrial metabolism, resulting from excessive nutrient exposure prior to and during conception, may underlie poor reproductive outcomes frequently reported in obese women.
Collapse
Affiliation(s)
- Natalia Igosheva
- Division of Reproduction and Endocrinology, King's College London, London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|