1
|
Xu YP, Fu JC, Hong ZL, Zeng DF, Guo CQ, Li P, Wu JX. Psychological stressors involved in the pathogenesis of premature ovarian insufficiency and potential intervention measures. Gynecol Endocrinol 2024; 40:2360085. [PMID: 38813955 DOI: 10.1080/09513590.2024.2360085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/14/2024] [Indexed: 05/31/2024] Open
Abstract
Premature ovarian insufficiency (POI) is a common gynecological endocrine disease, which seriously affects women's physical and mental health and fertility, and its incidence is increasing year by year. With the development of social economy and technology, psychological stressors such as anxiety and depression caused by social, life and environmental factors may be one of the risk factors for POI. We used PubMed to search peer-reviewed original English manuscripts published over the last 10 years to identify established and experimental studies on the relationship between various types of stress and decreased ovarian function. Oxidative stress, follicular atresia, and excessive activation of oocytes, caused by Stress-associated factors may be the main causes of ovarian function damage. This article reviews the relationship between psychological stressors and hypoovarian function and the possible early intervention measures in order to provide new ideas for future clinical treatment and intervention.
Collapse
Affiliation(s)
- Ying-Pei Xu
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Ji-Chun Fu
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Zhi-Lin Hong
- Clinical Laboratory Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - De-Fei Zeng
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Chao-Qin Guo
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Ping Li
- Department of Reproductive Medicine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Jin-Xiang Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Reproductive Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
2
|
Oestreich AK, Harasymowicz NS, Savadipour A, Harissa Z, Rashidi N, Luhmann MK, Kuziez M, Moley KH, Guilak F. Maternal high-fat high-sugar diet impairs bone quality and strength but not cartilage in aging mice. J Orthop Res 2024. [PMID: 39342461 DOI: 10.1002/jor.25980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 07/15/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024]
Abstract
Osteoarthritis (OA) is a prevalent aging disorder of synovial joints and recent work suggests that a parental high-fat diet increases OA severity following joint injury in offspring. We hypothesized that a maternal high-fat high-sugar (HFHS) diet would promote spontaneous osteoarthritis-related cartilage and bone changes in 1-year-old offspring. Female C57BL/6 J mice were placed on either a chow control or HFHS diet for 6 weeks before mating to a chow-fed C57BL/6 J male and maintained on their assigned diets throughout pregnancy and lactation. Male and female offspring were weaned onto a chow diet, raised to 1 year of age, and evaluated for cartilage and bone changes indicative of OA. However, offspring did not show early signs of OA as measured by histological Mankin scoring, mechanical testing of the pericellular matrix, histological synovitis scoring, or subchondral bone thickening as measured by microcomputed Tomography. On the other hand, male offspring from HFHS-fed dams had reduced trabecular bone quality in the tibial metaphysis and decreased cortical thickness. Although maternal HFHS diet did not impact trabecular or cortical bone quality in tibias of female offspring, the radii of these animals had decreased cortical thickness, increased medullary area, and impaired breaking strength compared to those of control-fed dams. Finally, we evaluated bone quality and strength in male and female F2 offspring and found that the grandmaternal diet modestly impacted radial bone geometry but not strength. Together these results suggest that maternal HFHS diet impairs F1 offspring skeletal integrity in a sex and bone site-specific manner.
Collapse
Affiliation(s)
- Arin K Oestreich
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Natalia S Harasymowicz
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Alireza Savadipour
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, USA
| | - Zainab Harissa
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Neda Rashidi
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, USA
| | - Meredith K Luhmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Mohammed Kuziez
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Kelle H Moley
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University, St. Louis, Missouri, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
3
|
Xhonneux I, Marei WFA, Meulders B, Slootmans J, Pintelon I, Leroy JLMR. The impact of offspring and maternal obesogenic diets on adult offspring oocyte mitochondrial morphology in primordial and preantral follicles. PLoS One 2024; 19:e0305912. [PMID: 38935642 PMCID: PMC11210809 DOI: 10.1371/journal.pone.0305912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024] Open
Abstract
Diet-induced obesity reduces oocyte quality mainly by impacting oocyte mitochondrial functions. Moreover, maternal obesity is associated with mitochondrial dysfunction in oocytes of their adult offspring. However, these effects were reported only in fully grown oocytes, mainly in the form of abnormal mitochondrial ultrastructure. It is unknown if obesogenic (OB) diets or maternal obesity already impact the primordial and preantral follicles. Considering the long duration and dynamics of folliculogenesis, determining the stage at which oocytes are affected and the extent of the damage is crucial for optimal reproductive management of obese patients and their daughters. Potential interaction between maternal and offspring diet effects are also not described, yet pivotal in our contemporary society. Therefore, here we examined the impact of OB diets on oocyte mitochondrial ultrastructure in primordial and activated preantral follicles in offspring from diet-induced obese or lean mothers. We used an outbred Swiss mouse model to increase the pathophysiological relevance to humans. Female mice were fed control or OB diets for 7 weeks, then mated with control males. Their female offspring were fed control or OB diets after weaning for 7 weeks (2-by-2 factorial design). Adult offspring ovarian sections were examined using transmission electron microscopy. We characterised and classified unique features of oocyte mitochondrial ultrastructure in the preantral follicles. An increase in mitochondrial matrix density was the most predominant change during follicle activation in secondary follicles, a feature that is linked with a higher mitochondrial activity. Maternal obesity increased mitochondrial density already in the primordial follicles suggesting an earlier increase in bioenergetic capacity. Maternal obesity did not induce abberant ultrastructure (abnormalities and defects) in primordial or preantral follicles. In contrast, offspring OB diet increased mitochondrial abnormalities in the primordial follicles. Further investigation of the consequences of these changes on oocyte metabolic regulation and stress levels during folliculogenesis is needed.
Collapse
Affiliation(s)
- Inne Xhonneux
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Waleed F. A. Marei
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
- Faculty of Veterinary Medicine, Department of Theriogenology, Cairo University, Giza, Egypt
| | - Ben Meulders
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Jens Slootmans
- Department of Biosystems, University of Louvain, Louvain, Belgium
- Flanders Research Institute for Agriculture, Fisheries and Food, Merelbeke, Belgium
| | - Isabel Pintelon
- Department of Veterinary Sciences, Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Belgium
- Antwerp Centre for Advanced Microscopy (ACAM), University of Antwerp, Wilrijk, Belgium
| | - Jo L. M. R. Leroy
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
4
|
Pinckard KM, Félix-Soriano E, Hamilton S, Terentyeva R, Baer LA, Wright KR, Nassal D, Esteves JV, Abay E, Shettigar VK, Ziolo MT, Hund TJ, Wold LE, Terentyev D, Stanford KI. Maternal exercise preserves offspring cardiovascular health via oxidative regulation of the ryanodine receptor. Mol Metab 2024; 82:101914. [PMID: 38479548 PMCID: PMC10965826 DOI: 10.1016/j.molmet.2024.101914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
OBJECTIVE The intrauterine environment during pregnancy is a critical factor in the development of obesity, diabetes, and cardiovascular disease in offspring. Maternal exercise prevents the detrimental effects of a maternal high fat diet on the metabolic health in adult offspring, but the effects of maternal exercise on offspring cardiovascular health have not been thoroughly investigated. METHODS To determine the effects of maternal exercise on offspring cardiovascular health, female mice were fed a chow (C; 21% kcal from fat) or high-fat (H; 60% kcal from fat) diet and further subdivided into sedentary (CS, HS) or wheel exercised (CW, HW) prior to pregnancy and throughout gestation. Offspring were maintained in a sedentary state and chow-fed throughout 52 weeks of age and subjected to serial echocardiography and cardiomyocyte isolation for functional and mechanistic studies. RESULTS High-fat fed sedentary dams (HS) produced female offspring with reduced ejection fraction (EF) compared to offspring from chow-fed dams (CS), but EF was preserved in offspring from high-fat fed exercised dams (HW) throughout 52 weeks of age. Cardiomyocytes from HW female offspring had increased kinetics, calcium cycling, and respiration compared to CS and HS offspring. HS offspring had increased oxidation of the RyR2 in cardiomyocytes coupled with increased baseline sarcomere length, resulting in RyR2 overactivity, which was negated in female HW offspring. CONCLUSIONS These data suggest a role for maternal exercise to protect against the detrimental effects of a maternal high-fat diet on female offspring cardiac health. Maternal exercise improved female offspring cardiomyocyte contraction, calcium cycling, respiration, RyR2 oxidation, and RyR2 activity. These data present an important, translatable role for maternal exercise to preserve cardiac health of female offspring and provide insight on mechanisms to prevent the transmission of cardiovascular diseases to subsequent generations.
Collapse
Affiliation(s)
- Kelsey M Pinckard
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Elisa Félix-Soriano
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shanna Hamilton
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Radmila Terentyeva
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Lisa A Baer
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Katherine R Wright
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Drew Nassal
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, Cardiovascular Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Joao Victor Esteves
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Eaman Abay
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Vikram K Shettigar
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Mark T Ziolo
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Thomas J Hund
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Internal Medicine, Cardiovascular Medicine, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Biomedical Engineering, The Ohio State University College of Engineering, Columbus, OH, USA
| | - Loren E Wold
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of Cardiac Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Dmitry Terentyev
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Surgery, Division of General and Gastrointestinal Surgery, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
5
|
Meulders B, Marei WFA, Xhonneux I, Loier L, Smits A, Leroy JLMR. Preconception Diet Interventions in Obese Outbred Mice and the Impact on Female Offspring Metabolic Health and Oocyte Quality. Int J Mol Sci 2024; 25:2236. [PMID: 38396912 PMCID: PMC10888670 DOI: 10.3390/ijms25042236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Obese individuals often suffer from metabolic health disorders and reduced oocyte quality. Preconception diet interventions in obese outbred mice restore metabolic health and oocyte quality and mitochondrial ultrastructure. Also, studies in inbred mice have shown that maternal obesity induces metabolic alterations and reduces oocyte quality in offspring (F1). Until now, the effect of maternal high-fat diet on F1 metabolic health and oocyte quality and the potential beneficial effects of preconception dietary interventions have not been studied together in outbred mice. Therefore, we fed female mice a high-fat/high-sugar (HF/HS) diet for 7 weeks and switched them to a control (CONT) or caloric-restriction (CR) diet or maintained them on the HF/HS diet for 4 weeks before mating, resulting in three treatment groups: diet normalization (DN), CR, and HF/HS. In the fourth group, mice were fed CONT diet for 11 weeks (CONT). HF/HS mice were fed an HF/HS diet from conception until weaning, while all other groups were then fed a CONT diet. After weaning, offspring were kept on chow diet and sacrificed at 11 weeks. We observed significantly elevated serum insulin concentrations in female HF/HS offspring and a slightly increased percentage of mitochondrial ultrastructural abnormalities, mitochondrial size, and mitochondrial mean gray intensity in HF/HS F1 oocytes. Also, global DNA methylation was increased and cellular stress-related proteins were downregulated in HF/HS F1 oocytes. Mostly, these alterations were prevented in the DN group, while, in CR, this was only the case for a few parameters. In conclusion, this research has demonstrated for the first time that a maternal high-fat diet in outbred mice has a moderate impact on female F1 metabolic health and oocyte quality and that preconception DN is a better strategy to alleviate this compared to CR.
Collapse
Affiliation(s)
- Ben Meulders
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
| | - Waleed F. A. Marei
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
- Faculty of Veterinary Medicine, Department of Theriogenology, Cairo University, Giza 12211, Egypt
| | - Inne Xhonneux
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
| | - Lien Loier
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
| | - Anouk Smits
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
| | - Jo L. M. R. Leroy
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium; (B.M.); (W.F.A.M.); (I.X.); (L.L.); (A.S.)
| |
Collapse
|
6
|
Xhonneux I, Marei WFA, Meulders B, Andries S, Leroy JLMR. The impact of a maternal and offspring obesogenic diet on daughter's oocyte mitochondrial ultrastructure and bioenergetic responses. Insights from an outbred mouse model. Front Physiol 2023; 14:1288472. [PMID: 37965107 PMCID: PMC10642210 DOI: 10.3389/fphys.2023.1288472] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Obesity affects oocyte mitochondrial functions and reduces oocyte quality and fertility. Obesity may also increase the risk of metabolic disorders in the offspring. Children are likely to follow their parents lifestyle and diet, which also contributes to the increased prevelance of obesity across generations. We hypothesise that the impact of obesogenic (OB) diet and obesity on oocyte mitochondrial functions is different in offspring born to obese mothers compared to those born to healthy mothers. To test this hypothesis, we fed a control (C, 10% fat, 7% sugar) or an OB diet (60% fat, 20% sugar) to female mice (for 7 weeks (w)) and then to their female offspring (for 7w after weaning) in a 2 × 2 factorial design (C » C, n = 35, C » OB, n = 35, OB » C n = 49 and OB » OB, n = 50). Unlike many other studies, we used an outbred Swiss mouse model to increase the human pathophysiological relevance. Offspring were sacrificed at 10w and their oocytes were collected. Offspring OB diet increased oocyte lipid droplet content, mitochondrial activity and reactive oxygen species (ROS) levels, altered mitochondrial ultrastructure and reduced oocyte pyruvate consumption. Mitochondrial DNA copy numbers and lactate production remained unaffected. Mitochondrial ultrastructure was the only factor where a significant interaction between maternal and offspring diet effect was detected. The maternal OB background resulted in a small but significant increase in offspring's oocyte mitochondrial ultrastructural abnormalities without altering mitochondrial inner membrane potential, active mitochondrial distribution, mitochondrial DNA copy numbers, or ROS production. This was associated with reduced mitochondrial complex III and V expression and reduced pyruvate consumption which may be compensatory mechanisms to control mitochondrial inner membrane potential and ROS levels. Therefore, in this Swiss outbred model, while offspring OB diet had the largest functional impact on oocyte mitochondrial features, the mitochondrial changes due to the maternal background appear to be adaptive and compensatory rather than dysfunctional.
Collapse
Affiliation(s)
- Inne Xhonneux
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Waleed F. A. Marei
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Ben Meulders
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Silke Andries
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Jo L. M. R. Leroy
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
7
|
Elías-López AL, Vázquez-Mena O, Sferruzzi-Perri AN. Mitochondrial dysfunction in the offspring of obese mothers and it's transmission through damaged oocyte mitochondria: Integration of mechanisms. Biochim Biophys Acta Mol Basis Dis 2023:166802. [PMID: 37414229 DOI: 10.1016/j.bbadis.2023.166802] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/23/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023]
Abstract
In vivo and in vitro studies demonstrate that mitochondria in the oocyte, are susceptible to damage by suboptimal pre/pregnancy conditions, such as obesity. These suboptimal conditions have been shown to induce mitochondrial dysfunction (MD) in multiple tissues of the offspring, suggesting that mitochondria of oocytes that pass from mother to offspring, can carry information that can programme mitochondrial and metabolic dysfunction of the next generation. They also suggest that transmission of MD could increase the risk of obesity and other metabolic diseases in the population inter- and trans-generationally. In this review, we examined whether MD observed in offspring tissues of high energetic demand, is the result of the transmission of damaged mitochondria from obese mothers' oocytes to the offspring. The contribution of genome-independent mechanisms (namely mitophagy) in this transmission were also explored. Finally, potential interventions aimed at improving oocyte/embryo health were investigated, to see if they may provide an opportunity to halter the generational effects of MD.
Collapse
Affiliation(s)
- A L Elías-López
- Dirección de Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Mexico.
| | | | - A N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, UK.
| |
Collapse
|
8
|
Leroy JLMR, Meulders B, Moorkens K, Xhonneux I, Slootmans J, De Keersmaeker L, Smits A, Bogado Pascottini O, Marei WFA. Maternal metabolic health and fertility: we should not only care about but also for the oocyte! Reprod Fertil Dev 2022; 35:1-18. [PMID: 36592978 DOI: 10.1071/rd22204] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Metabolic disorders due to obesity and unhealthy lifestyle directly alter the oocyte's microenvironment and impact oocyte quality. Oxidative stress and mitochondrial dysfunction play key roles in the pathogenesis. Acute effects on the fully grown oocytes are evident, but early follicular stages are also sensitive to metabolic stress leading to a long-term impact on follicular cells and oocytes. Improving the preconception health is therefore of capital importance but research in animal models has demonstrated that oocyte quality is not fully recovered. In the in vitro fertilisation clinic, maternal metabolic disorders are linked with disappointing assisted reproductive technology results. Embryos derived from metabolically compromised oocytes exhibit persistently high intracellular stress levels due to weak cellular homeostatic mechanisms. The assisted reproductive technology procedures themselves form an extra burden for these defective embryos. Minimising cellular stress during culture using mitochondrial-targeted therapy could rescue compromised embryos in a bovine model. However, translating such applications to human in vitro fertilisation clinics is not simple. It is crucial to consider the sensitive epigenetic programming during early development. Research in humans and relevant animal models should result in preconception care interventions and in vitro strategies not only aiming at improving fertility but also safeguarding offspring health.
Collapse
Affiliation(s)
- J L M R Leroy
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - B Meulders
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - K Moorkens
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - I Xhonneux
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - J Slootmans
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - L De Keersmaeker
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - A Smits
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - O Bogado Pascottini
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - W F A Marei
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
9
|
Di Berardino C, Peserico A, Capacchietti G, Zappacosta A, Bernabò N, Russo V, Mauro A, El Khatib M, Gonnella F, Konstantinidou F, Stuppia L, Gatta V, Barboni B. High-Fat Diet and Female Fertility across Lifespan: A Comparative Lesson from Mammal Models. Nutrients 2022; 14:nu14204341. [PMID: 36297035 PMCID: PMC9610022 DOI: 10.3390/nu14204341] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022] Open
Abstract
Female reproduction focuses mainly on achieving fully grown follicles and competent oocytes to be successfully fertilized, as well as on nourishing the developing offspring once pregnancy occurs. Current evidence demonstrates that obesity and/or high-fat diet regimes can perturbate these processes, leading to female infertility and transgenerational disorders. Since the mechanisms and reproductive processes involved are not yet fully clarified, the present review is designed as a systematic and comparative survey of the available literature. The available data demonstrate the adverse influences of obesity on diverse reproductive processes, such as folliculogenesis, oogenesis, and embryo development/implant. The negative reproductive impact may be attributed to a direct action on reproductive somatic and germinal compartments and/or to an indirect influence mediated by the endocrine, metabolic, and immune axis control systems. Overall, the present review highlights the fragmentation of the current information limiting the comprehension of the reproductive impact of a high-fat diet. Based on the incidence and prevalence of obesity in the Western countries, this topic becomes a research challenge to increase self-awareness of dietary reproductive risk to propose solid and rigorous preventive dietary regimes, as well as to develop targeted pharmacological interventions.
Collapse
Affiliation(s)
- Chiara Di Berardino
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alessia Peserico
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Correspondence:
| | - Giulia Capacchietti
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alex Zappacosta
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council, A. Buzzati-Traverso Campus, via E. Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy
| | - Valentina Russo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Annunziata Mauro
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Mohammad El Khatib
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Francesca Gonnella
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Fani Konstantinidou
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Barbara Barboni
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
10
|
Programming by maternal obesity: a pathway to poor cardiometabolic health in the offspring. Proc Nutr Soc 2022; 81:227-242. [DOI: 10.1017/s0029665122001914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is an ever increasing prevalence of maternal obesity worldwide such that in many populations over half of women enter pregnancy either overweight or obese. This review aims to summarise the impact of maternal obesity on offspring cardiometabolic outcomes. Maternal obesity is associated with increased risk of adverse maternal and pregnancy outcomes. However, beyond this exposure to maternal obesity during development also increases the risk of her offspring developing long-term adverse cardiometabolic outcomes throughout their adult life. Both human studies and those in experimental animal models have shown that maternal obesity can programme increased risk of offspring developing obesity and adipose tissue dysfunction; type 2 diabetes with peripheral insulin resistance and β-cell dysfunction; CVD with impaired cardiac structure and function and hypertension via impaired vascular and kidney function. As female offspring themselves are therefore likely to enter pregnancy with poor cardiometabolic health this can lead to an inter-generational cycle perpetuating the transmission of poor cardiometabolic health across generations. Maternal exercise interventions have the potential to mitigate some of the adverse effects of maternal obesity on offspring health, although further studies into long-term outcomes and how these translate to a clinical context are still required.
Collapse
|
11
|
Yang F, Wu S, Yang W, Yu J, Liu B, Zeng S. Effects of exercise on donkey semen parameters and some physiological indices. J Equine Vet Sci 2022; 117:104079. [PMID: 35843388 DOI: 10.1016/j.jevs.2022.104079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022]
Abstract
To study the effects of exercise on donkey semen parameters, Dezhou donkey (Equus asinus) jackasses (n=18) were assigned to three groups: control (CN, n=6), one-hour exercise (1 h EX, n=6) and two-hour exercise (2 h EX, n=6) groups. They were exercised with a walking machine for 3 days/week for 8 weeks. Semen parameters and reproductive hormones were evaluated weekly. The heart rate, rectal temperature, blood haematology indexes and the body condition score (BCS) were also measured. Results showed that the ejaculation volume increased while the sperm concentration decreased after exercise. However, other sperm parameters were unaffected. Reproductive hormones at resting period were rarely affected by exercise. The red blood cell number increased (P=0.023), while the mean erythrocyte hemoglobin concentration decreased (P=0.045) after the 8-week exercise in the 2 h EX group compared with the CN. After the 8-week exercise, the resting heart rate of the two exercise groups decreased significantly compared with the CN. The BCS in the CN group increased after the experiment (P=0.024). In conclusion, we found that exercise had positive effects on some physiological indices but did not affect semen parameters expect for volume and concentration of the jackass.
Collapse
Affiliation(s)
- Feng Yang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China; National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| | - Shuaishuai Wu
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, China
| | - Weigang Yang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jie Yu
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co. Ltd., Dong'e, China
| | - Bing Liu
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co. Ltd., Dong'e, China
| | - Shenming Zeng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| |
Collapse
|
12
|
Tocantins C, Diniz MS, Grilo LF, Pereira SP. The birth of cardiac disease: Mechanisms linking gestational diabetes mellitus and early onset of cardiovascular disease in offspring. WIREs Mech Dis 2022; 14:e1555. [PMID: 35304833 DOI: 10.1002/wsbm.1555] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 02/10/2022] [Accepted: 03/09/2022] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is the biggest killer worldwide, composing a major economic burden for health care systems. Obesity and diabetes are dual epidemics on the rise and major risk factors predisposing for CVD. Increased obesity- and diabetes-related incidence is now observed among children, adolescents, and young adults. Gestational diabetes mellitus (GDM) is the most common metabolic pregnancy disorder, and its prevalence is rapidly increasing. During pregnancies complicated by GDM, the offspring are exposed to a compromised intrauterine environment characterized by hyperglycemic periods. Unfavorable in utero conditions at critical periods of fetal cardiac development can produce developmental adaptations that remodel the cardiovascular system in a way that can contribute to adult-onset of heart disease due to the programming during fetal life. Epidemiological studies have reported increased cardiovascular complications among GDM-descendants, highlighting the urgent need to investigate and understand the mechanisms modulated during fetal development of in utero GDM-exposed offspring that predispose an individual to increased CVD during life. In this manuscript, we overview previous studies in this area and gather evidence linking GDM and CVD development in the offspring, providing new insights on novel mechanisms contributing to offspring CVD programming by GDM, from the role of maternal-fetal interactions to their impact on fetal cardiovascular development, how the perpetuation of cardiac programming is maintained in postnatal life, and advance the intergenerational implications contributing to increased CVD premature origin. Understanding the perpetuation of CVD can be the first step to manage and reverse this leading cause of morbidity and mortality. This article is categorized under: Reproductive System Diseases > Molecular and Cellular Physiology Cardiovascular Diseases > Molecular and Cellular Physiology Metabolic Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Carolina Tocantins
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Mariana S Diniz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Luís F Grilo
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Susana P Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Laboratory of Metabolism and Exercise (LametEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
13
|
Gonzalez MB, Robker RL, Rose RD. Obesity and oocyte quality: Significant implications for ART and Emerging mechanistic insights. Biol Reprod 2021; 106:338-350. [PMID: 34918035 DOI: 10.1093/biolre/ioab228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/22/2021] [Accepted: 12/07/2021] [Indexed: 11/14/2022] Open
Abstract
The prevalence of obesity in adults worldwide, and specifically in women of reproductive age, is concerning given the risks to fertility posed by the increased risk of type 2 diabetes, metabolic syndrome and other non-communicable diseases. Obesity has a multi-systemic impact in female physiology that is characterized by the presence of oxidative stress, lipotoxicity, and the activation of pro-inflammatory pathways, inducing tissue-specific insulin resistance and ultimately conducive to abnormal ovarian function. A higher body mass is linked to Polycystic Ovary Syndrome, dysregulated menstrual cycles, anovulation, and longer time to pregnancy, even in ovulatory women. In the context of ART, compared to women of normal BMI, obese women have worse outcomes in every step of their journey, resulting in reduced success measured as live birth rate. Even after pregnancy is achieved, obese women have a higher chance of miscarriage, gestational diabetes, pregnancy complications, birth defects, and most worryingly, a higher risk of stillbirth and neonatal death. The potential for compounding effects of ART on pregnancy complications and infant morbidities in obese women has not been studied. There is still much debate in the field on whether these poorer outcomes are mainly driven by defects in oocyte quality, abnormal embryo development or an unaccommodating uterine environment, however the clinical evidence to date suggests a combination of all three are responsible. Animal models of maternal obesity shed light on the mechanisms underlaying the effects of obesity on the peri-conception environment, with recent findings pointing to lipotoxicity in the ovarian environment as a key driver of defects in oocytes that have not only reduced developmental competence but long-lasting effects in offspring health.
Collapse
Affiliation(s)
- Macarena B Gonzalez
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Rebecca L Robker
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Ryan D Rose
- Robinson Research Institute, School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia.,Fertility SA, St. Andrews Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
14
|
Wang Y, Wang X, Long Q, Liu Y, Yin T, Sirota I, Ren F, Gu Z, Luo J. Reducing embryonic mtDNA copy number alters epigenetic profile of key hepatic lipolytic genes and causes abnormal lipid accumulation in adult mice. FEBS J 2021; 288:6828-6843. [PMID: 34258867 DOI: 10.1111/febs.16121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/30/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022]
Abstract
Adverse fetal environment, in particular a shortage or excess of nutrients, is associated with increased risks of metabolic diseases later in life. However, the molecular mechanisms underlying this developmental origin of adult diseases remain unclear. Here, we directly tested the role of mitochondrial stress in mediating fetal programming in mice by enzymatically depleting mtDNA in zygotes. mtDNA-targeted plasmid microinjection is used to reduce embryonic mtDNA copy number directly, followed by embryo transfer. Mice with reduced zygote mtDNA copy number were born morphologically normal and showed no accelerated body weight gain. However, at 5 months of age these mice showed markedly increased hepatic lipidosis and became glucose-intolerant. Hepatic mRNA and protein expressions of peroxisome proliferator-activated receptor α (Pparα), a key transcriptional regulator of lipid metabolism, were significantly decreased as a result of increased DNA methylation in its proximal regulatory region. These results indicate that perturbation of mitochondrial function around the periconceptional period causes hypermethylation and thus suppressed expression of PPARα in fetal liver, leading to impaired hepatic lipid metabolism. Our findings provide the first direct evidence that mitochondrial stress mediates epigenetic changes associated with fetal programming of adult diseases in a mammalian system.
Collapse
Affiliation(s)
- Yakun Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Xuan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Qiaoming Long
- Cam-Su Mouse Genomic Resource Center, Soochow University, China
| | - Yuanwu Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Tao Yin
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Inna Sirota
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Junjie Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
15
|
Abstract
The purpose of this American Society for Reproductive Medicine Practice Committee report is to provide clinicians with principles and strategies for the evaluation and treatment of couples with infertility associated with obesity. This revised document replaces the Practice Committee document titled "Obesity and reproduction: an educational bulletin" last published in 2015 (Fertil Steril 2015;104:1116-26).
Collapse
Affiliation(s)
-
- American Society for Reproductive Medicine, Birmingham, Alabama
| | | |
Collapse
|
16
|
Wang Z, Groen H, Van Zomeren KC, Cantineau AEP, Van Oers A, Van Montfoort APA, Kuchenbecker WKH, Pelinck MJ, Broekmans FJM, Klijn NF, Kaaijk EM, Mol BWJ, Hoek A, Van Echten-Arends J. Lifestyle intervention prior to IVF does not improve embryo utilization rate and cumulative live birth rate in women with obesity: a nested cohort study. Hum Reprod Open 2021; 2021:hoab032. [PMID: 34557597 PMCID: PMC8452483 DOI: 10.1093/hropen/hoab032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/13/2021] [Indexed: 12/26/2022] Open
Abstract
STUDY QUESTION Does lifestyle intervention consisting of an energy-restricted diet, enhancement of physical activity and motivational counseling prior to IVF improve embryo utilization rate (EUR) and cumulative live birth rate (CLBR) in women with obesity? SUMMARY ANSWER A 6-month lifestyle intervention preceding IVF improved neither EUR nor CLBR in women with obesity in the first IVF treatment cycle where at least one oocyte was retrieved. WHAT IS KNOWN ALREADY A randomized controlled trial (RCT) evaluating the efficacy of a low caloric liquid formula diet (LCD) preceding IVF in women with obesity was unable to demonstrate an effect of LCD on embryo quality and live birth rate: in this study, only one fresh embryo transfer (ET) or, in case of freeze-all strategy, the first transfer with frozen-thawed embryos was reported. We hypothesized that any effect on embryo quality of a lifestyle intervention in women with obesity undergoing IVF treatment is better revealed by EUR and CLBR after transfer of all fresh and frozen-thawed embryos. STUDY DESIGN, SIZE, DURATION This is a nested cohort study within an RCT, the LIFEstyle study. The original study examined whether a 6-month lifestyle intervention prior to infertility treatment in women with obesity improved live birth rate, compared to prompt infertility treatment within 24 months after randomization. In the original study between 2009 and 2012, 577 (three women withdrew informed consent) women with obesity and infertility were assigned to a lifestyle intervention followed by infertility treatment (n = 289) or to prompt infertility treatment (n = 285). PARTICIPANTS/MATERIALS, SETTING, METHODS Only participants from the LIFEstyle study who received IVF treatment were eligible for the current analysis. In total, 137 participants (n = 58 in the intervention group and n = 79 in the control group) started the first cycle. In 25 participants, the first cycle was cancelled prior to oocyte retrieval mostly due to poor response. Sixteen participants started a second or third consecutive cycle. The first cycle with successful oocyte retrieval was used for this analysis, resulting in analysis of 51 participants in the intervention group and 72 participants in the control group. Considering differences in embryo scoring methods and ET day strategy between IVF centers, we used EUR as a proxy for embryo quality. EUR was defined as the proportion of inseminated/injected oocytes per cycle that was transferred or cryopreserved as an embryo. Analysis was performed per cycle and per oocyte/embryo. CLBR was defined as the percentage of participants with at least one live birth from the first fresh and subsequent frozen-thawed ET(s). In addition, we calculated the Z-score for singleton neonatal birthweight and compared these outcomes between the two groups. MAIN RESULTS AND THE ROLE OF CHANCE The overall mean age was 31.6 years and the mean BMI was 35.4 ± 3.2 kg/m2 in the intervention group, and 34.9 ± 2.9 kg/m2 in the control group. The weight change at 6 months was in favor of the intervention group (mean difference in kg vs the control group: −3.14, 95% CI: −5.73 to −0.56). The median (Q25; Q75) number of oocytes retrieved was 4.00 (2.00; 8.00) in the intervention group versus 6.00 (4.00; 9.75) in the control group, and was not significantly different, as was the number of oocytes inseminated/injected (4.00 [2.00; 8.00] vs 6.00 [3.00; 8.75]), normal fertilized embryos (2.00 [0.50; 5.00] vs 3.00 [1.00; 5.00]) and the number of cryopreserved embryos (2.00 [1.25; 4.75] vs 2.00 [1.00; 4.00]). The median (Q25; Q75) EUR was 33.3% (12.5%; 60.0%) in the intervention group and 33.3% (16.7%; 50.0%) in the control group in the per cycle analysis (adjusted B: 2.7%, 95% CI: −8.6% to 14.0%). In the per oocyte/embryo analysis, in total, 280 oocytes were injected or inseminated in the intervention group, 113 were utilized (transferred or cryopreserved, EUR = 40.4%); in the control group, EUR was 30.8% (142/461). The lifestyle intervention did not significantly improve EUR (adjusted odds ratio [OR]: 1.36, 95% CI: 0.94–1.98) in the per oocyte/embryo analysis, taking into account the interdependency of the oocytes per participant. CLBR was not significantly different between the intervention group and the control group after adjusting for type of infertility (male factor and unexplained) and smoking (27.5% vs 22.2%, adjusted OR: 1.03, 95% CI: 0.43–2.47). Singleton neonatal birthweight and Z-score were not significantly different between the two groups. LIMITATIONS, REASONS FOR CAUTION This study is a nested cohort study within an RCT, and no power calculation was performed. The randomization was not stratified for indicated treatment, and although we corrected our analyses for baseline differences, there may be residual confounding. The limited absolute weight loss and the short duration of the lifestyle intervention might be insufficient to affect EUR and CLBR. WIDER IMPLICATIONS OF THE FINDINGS Our data do not support the hypothesis of a beneficial short-term effect of lifestyle intervention on EUR and CLBR after IVF in women with obesity, although more studies are needed as there may be a potential clinically relevant effect on EUR. STUDY FUNDING/COMPETING INTEREST(S) The study was supported by a grant from ZonMw, the Dutch Organization for Health Research and Development (50-50110-96-518). A.H. has received an unrestricted educational grant from Ferring pharmaceuticals BV, The Netherlands. B.W.J.M. is supported by an NHMRC Investigator grant (GNT1176437). B.W.J.M. reports consultancy for Guerbet, has been a member of the ObsEva advisory board and holds Stock options for ObsEva. B.W.J.M. has received research funding from Guerbet, Ferring and Merck. F.J.M.B. reports personal fees from membership of the external advisory board for Merck Serono and a research support grant from Merck Serono, outside the submitted work. TRIAL REGISTRATION NUMBER The LIFEstyle RCT was registered at the Dutch trial registry (NTR 1530). https://www.trialregister.nl/trialreg/admin/rctview.asp?TC=1530.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Henk Groen
- Department of Epidemiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Koen C Van Zomeren
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Astrid E P Cantineau
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Anne Van Oers
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Aafke P A Van Montfoort
- Department of Obstetrics and Gynecology, GROW School for Oncology and Development Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Marie J Pelinck
- Department of Obstetrics and Gynecology, Treant Zorggroep, Emmen, The Netherlands
| | - Frank J M Broekmans
- Division Women and Baby, Department of Reproductive Medicine and Gynecology, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nicole F Klijn
- Department of Gynecology and Reproductive Medicine, University of Leiden, Leiden University Medical Center, Leiden, The Netherlands
| | - Eugenie M Kaaijk
- Department of Obstetrics and Gynecology, Onze Lieve Vrouwe Gasthuis (OLVG), Amsterdam, The Netherlands
| | - Ben W J Mol
- Department of Obstetrics and Gynecology, Monash University, Clayton, Victoria, Australia
| | - Annemieke Hoek
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Jannie Van Echten-Arends
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
17
|
Furtado RL, Martins JER, Oliveira MAF, Guerreiro DD, de Sá NAR, Ferraz ASM, Ceccatto VM, Rodrigues APR, Araújo VR. Acute effect of high-intensity interval training exercise on redox status in the ovaries of rats fed a high-fat diet. Reprod Fertil Dev 2021; 33:713-724. [PMID: 34437833 DOI: 10.1071/rd20326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 05/05/2021] [Indexed: 11/23/2022] Open
Abstract
This study demonstrates the effect of a single high-intensity interval training (HIIT) session on the redox status of rat ovaries with excess adiposity. Forty Wistar female rats (mean (±s.e.m.) weight 94.40 ± 13.40 g) were divided into two groups and fed either a standard diet (SD) or a high-fat diet (HFD) for 62 days. At the end of this period, the rats were subjected to a single HIIT session and were killed 24 h after exercise. Both groups subjected to exercise (SDex and HFDex) generated a significantly higher antioxidant environment by presenting a higher thiol content, which represents a lower oxidation rate of GSH than their respective controls (SD and HFD). The percentage of morphologically normal primary follicles decreased, whereas that of antral follicles increased, in the SDex group. In addition, the HFD group had a higher percentage of degenerated antral follicles than the SD and SDex groups. Cells immunoreactive for α-smooth muscle actin were seen in the cortical stroma and thecal layer enclosing late secondary and tertiary follicles in all groups. Moreover, heme oxygenase and cytochrome P450 family 19 subfamily A member 1 (Cyp19A1) labelling was seen in all antral follicles. Progesterone concentrations were significantly higher in the HFDex than SDex group. In conclusion, this study indicates that a single session of HIIT may result in an improvement in ovary redox status because of metabolic muscle activity by inducing physiological adaptation after exercise in a paracrine manner.
Collapse
Affiliation(s)
- Rodrigo L Furtado
- Graduate Program in Physiological Sciences, Higher Institute of Biomedical Sciences, State University of Ceará, Fortaleza, CE, 60714-903, Brazil
| | - Jonathan Elias R Martins
- Institute of Physical Education and Sports, Federal University of Ceará, Fortaleza, CE, 60455760, Brazil
| | - Maria Alice F Oliveira
- Microscopy Laboratory of Health Sciences Center, State University of Ceará, Fortaleza, CE, 60714-903, Brazil
| | - Denise D Guerreiro
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Veterinary Faculty, State University of Ceará, Fortaleza, CE, 60714-903, Brazil
| | - Naiza A R de Sá
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Veterinary Faculty, State University of Ceará, Fortaleza, CE, 60714-903, Brazil
| | - Alex S M Ferraz
- Institute of Physical Education and Sports, Federal University of Ceará, Fortaleza, CE, 60455760, Brazil
| | - Vânia M Ceccatto
- Laboratory of Biochemistry and Gene Expression, Higher Institute of Biomedical Sciences, State University of Ceará, Fortaleza, CE, 60714-903, Brazil
| | - Ana Paula R Rodrigues
- Laboratory of Manipulation of Oocytes and Preantral Follicles, Veterinary Faculty, State University of Ceará, Fortaleza, CE, 60714-903, Brazil
| | - Valdevane R Araújo
- Graduate Program in Physiological Sciences, Higher Institute of Biomedical Sciences, State University of Ceará, Fortaleza, CE, 60714-903, Brazil; and Microscopy Laboratory of Health Sciences Center, State University of Ceará, Fortaleza, CE, 60714-903, Brazil; and Corresponding author.
| |
Collapse
|
18
|
Fawcett K, Martinez A, Crimmins M, Sims C, Børsheim E, Andres A. Effect of a dietary and exercise intervention in women with overweight and obesity undergoing fertility treatments: protocol for a randomized controlled trial. BMC Nutr 2021; 7:51. [PMID: 34399856 PMCID: PMC8367391 DOI: 10.1186/s40795-021-00454-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 07/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Distinct molecular, inflammatory, and metabolic signatures are present in oocytes and follicular fluid derived from women with obesity when compared to those derived from normal weight women, which suggest existing signals that may program future offspring for metabolic diseases. This study aims to assess the feasibility and efficacy of a peri-conception nutrition and exercise intervention on mitigating obesity-associated changes in oocyte gene expression profiles and follicular fluid metabolites. METHODS This single blinded randomized control trial will include 120 women with a BMI of 25-45 kg/m2, ≥21 years of age, and undergoing in vitro fertilization (IVF) treatments. Participants will be randomized to standard of care (N = 60) or an intervention group (N = 60) in a block design by polycystic ovary syndrome status. The intervention will combine a dietary component (Mediterranean meal plan) with exercise prescription following the Physical Activity Guidelines for Americans. Participants will be assessed pre- and post-intervention. The standard of care group will be offered to join the intervention group if the IVF treatments are unsuccessful as a cross over design. Recruitment is anticipated to start in July of 2021. Primary outcomes will include single oocyte gene expression profiles and follicular fluid metabolites. Mann-Whitney U nonparametric tests will be used to assess potential differences for each stratum. Follicular fluid and serum metabolites will be analyzed using a one-factor Analysis of Covariance (ANCOVA) at four levels, pair-wise comparisons using Tukey-Kramer post-hoc tests will be used to identify groups whose means differ significantly while retaining the family-wise error rate at 5%. When the design is balanced, two-way Analysis of Variance (ANOVA), or non-parametric Friedman test will be used in data analysis. Additionally, general linear models and ANCOVA may be used to control for covariates. Significance will be set at p < 0.05. Findings will be disseminated via peer-reviewed manuscripts and presentations at scientific conferences. DISCUSSION This study will provide novel data and key information on the impact of a dietary and exercise intervention on oocyte gene expression and follicular fluid content. Results will demonstrate the potential of such intervention in mitigating obesity-induced changes in oocyte gene expression and follicular fluid metabolites. TRIAL REGISTRATION ClinicalTrials.gov ( NCT04273048 ): submitted November 13, 2019; posted February 17, 2020.
Collapse
Affiliation(s)
- Kindann Fawcett
- Arkansas Children's Nutrition Center, 15 Children's Way, Slot 512-20B, Little Rock, AR, 72202, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Audrey Martinez
- Arkansas Children's Nutrition Center, 15 Children's Way, Slot 512-20B, Little Rock, AR, 72202, USA
| | - Meghan Crimmins
- Arkansas Children's Nutrition Center, 15 Children's Way, Slot 512-20B, Little Rock, AR, 72202, USA
| | - Clark Sims
- Arkansas Children's Nutrition Center, 15 Children's Way, Slot 512-20B, Little Rock, AR, 72202, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Elisabet Børsheim
- Arkansas Children's Nutrition Center, 15 Children's Way, Slot 512-20B, Little Rock, AR, 72202, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Aline Andres
- Arkansas Children's Nutrition Center, 15 Children's Way, Slot 512-20B, Little Rock, AR, 72202, USA. .,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA. .,Arkansas Children's Research Institute, Little Rock, AR, USA.
| |
Collapse
|
19
|
The Impact of Unbalanced Maternal Nutritional Intakes on Oocyte Mitochondrial Activity: Implications for Reproductive Function. Antioxidants (Basel) 2021; 10:antiox10010091. [PMID: 33440800 PMCID: PMC7826933 DOI: 10.3390/antiox10010091] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence on the effect of nutrition on reproduction is emerging from both animal and human studies. A healthy dietary pattern and nutrient supplementation, especially during the peri-conceptional period, might be helpful to achieve a live birth, although the mechanisms implicated are not fully understood. The endocrine system and the ooplasmic organelles apparatus, in particular the mitochondria, are clearly key elements during oogenesis and subsequent embryo development, and their proper functioning is associated with nutrition, even beyond maternal aging. Several studies in animal models have reported various adverse effects on mitochondria caused by unbalanced dietary intakes such as high fat diet, high fat high sugar diet, and low protein diet. The alterations produced might include mitochondrial intracellular distribution, content, structure, biogenesis, and functioning. This review summarizes the key role of mitochondria in female reproduction and the effects of different dietary macronutrient compositions on oocyte mitochondrial activity with their possible short-, medium-, and long-term effects.
Collapse
|
20
|
Rao A, Satheesh A, Nayak G, Poojary PS, Kumari S, Kalthur SG, Mutalik S, Adiga SK, Kalthur G. High-fat diet leads to elevated lipid accumulation and endoplasmic reticulum stress in oocytes, causing poor embryo development. Reprod Fertil Dev 2020; 32:1169-1179. [PMID: 32998795 DOI: 10.1071/rd20112] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/28/2020] [Indexed: 11/23/2022] Open
Abstract
The present study was designed to investigate the effect of diet-induced obesity on endoplasmic reticulum (ER) stress in oocytes. Swiss albino mice (3 weeks old) were fed with a high-fat diet (HFD) for 8 weeks. Oocytes were assessed for lipid droplet accumulation, oxidative stress, ER stress and their developmental potential invitro. High lipid accumulation (P<0.01) and elevated intracellular levels of reactive oxygen species were observed in both germinal vesicle and MII oocytes of HFD-fed mice (P<0.05 and P<0.01 respectively compared with control). Further, expression of the ER stress markers X-box binding protein 1 (XBP1), glucose-regulated protein 78 (GRP78), activating transcription factor 4 (ATF4) and activating transcription factor 6 (ATF6) was significantly (P<0.001) higher in oocytes of the HFD than control group. Oocytes from HFD-fed mice exhibited poor fertilisation and blastocyst rates, a decrease in total cell number and high levels of DNA damage (P<0.01) compared with controls. In conclusion, diet-induced obesity resulted in elevated lipid levels and higher oxidative and ER stress in oocytes, which contributed to the compromised developmental potential of embryos.
Collapse
Affiliation(s)
- Arpitha Rao
- Department of Clinical Embryology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576 104, Karnataka State, India
| | - Aparna Satheesh
- Department of Clinical Embryology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576 104, Karnataka State, India
| | - Guruprasad Nayak
- Department of Clinical Embryology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576 104, Karnataka State, India
| | - Pooja Suresh Poojary
- Department of Clinical Embryology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576 104, Karnataka State, India
| | - Sandhya Kumari
- Department of Clinical Embryology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576 104, Karnataka State, India
| | - Sneha Guruprasad Kalthur
- Department of Anatomy, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576 104, Karnataka State, India
| | - Srinivas Mutalik
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576 104, Karnataka State, India
| | - Satish Kumar Adiga
- Department of Clinical Embryology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576 104, Karnataka State, India
| | - Guruprasad Kalthur
- Department of Clinical Embryology, Kasturba Medical College Manipal, Manipal Academy of Higher Education, Manipal 576 104, Karnataka State, India; and Corresponding author.
| |
Collapse
|
21
|
Marei WFA, Smits A, Mohey-Elsaeed O, Pintelon I, Ginneberge D, Bols PEJ, Moerloose K, Leroy JLMR. Differential effects of high fat diet-induced obesity on oocyte mitochondrial functions in inbred and outbred mice. Sci Rep 2020; 10:9806. [PMID: 32555236 PMCID: PMC7299992 DOI: 10.1038/s41598-020-66702-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022] Open
Abstract
Maternal obesity can cause reduced oocyte quality and subfertility. Mitochondrial dysfunction plays a central role here, and most often inbred mouse models are used to study these pathways. We hypothesized that the mouse genetic background can influence the impact of high fat diet (HFD)-induced obesity on oocyte quality. We compared the inbred C57BL/6 (B6) and the outbred Swiss strains after feeding a HFD for 13w. HFD-mice had increased body weight gain, hypercholesterolemia, and increased oocyte lipid droplet (LD) accumulation in both strains. LD distribution was strain-dependent. In Swiss mouse oocytes, HFD significantly increased mitochondrial inner membrane potential (MMP), reactive oxygen species concentrations, mitochondrial ultrastructural abnormalities (by 46.4%), and endoplasmic reticulum (ER) swelling, and decreased mtDNA copy numbers compared with Swiss controls (P < 0.05). Surprisingly, B6-control oocytes exhibited signs of cellular stress compared to the Swiss controls (P < 0.05); upregulated gene expression of ER- and oxidative stress markers, high mitochondrial ultrastructural abnormalities (48.6%) and ER swelling. Consequently, the HFD impact on B6 oocyte quality was less obvious, with 9% higher mitochondrial abnormalities, and no additive effect on MMP and stress marks compared to B6 control (P > 0.1). Interestingly, mtDNA in B6-HFD oocytes was increased suggesting defective mitophagy. In conclusion, we show evidence that the genetic background or inbreeding can affect mitochondrial functions in oocytes and may influence the impact of HFD on oocyte quality. These results should create awareness when choosing and interpreting data obtained from different mouse models before extrapolating to human applications.
Collapse
Affiliation(s)
- Waleed F A Marei
- Gamete Research Centre, University of Antwerp, 2610, Wilrijk, Belgium. .,Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - Anouk Smits
- Gamete Research Centre, University of Antwerp, 2610, Wilrijk, Belgium
| | - Omnia Mohey-Elsaeed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.,Laboratory of Cell Biology & Histology, University of Antwerp, 2610, Wilrijk, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology & Histology, University of Antwerp, 2610, Wilrijk, Belgium
| | - Daisy Ginneberge
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter E J Bols
- Gamete Research Centre, University of Antwerp, 2610, Wilrijk, Belgium
| | - Katrien Moerloose
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jo L M R Leroy
- Gamete Research Centre, University of Antwerp, 2610, Wilrijk, Belgium
| |
Collapse
|
22
|
Metabolomic alternations of follicular fluid of obese women undergoing in-vitro fertilization treatment. Sci Rep 2020; 10:5968. [PMID: 32249791 PMCID: PMC7136245 DOI: 10.1038/s41598-020-62975-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/23/2020] [Indexed: 01/11/2023] Open
Abstract
Obesity exerts negative effects on the metabolic homeostasis of cells in various tissues, but how it influences ovum metabolism is not fully understood. Previous studies demonstrate that oocyte genes that regulate oxidative stress, lipid metabolism, and inflammation are highly expressed in obese women. However, the metabolic effects of these genetic variations are not clear. To address this gap, we conducted an exploratory evaluation of follicular fluid (FF) metabolites in underweight, normal-weight, overweight, and obese women undergoing in vitro fertilization (IVF) treatment. The FF samples from the underweight (Group A, n = 40), normal-weight (Group B, n = 40), overweight (Group C, n = 40), and obese women (Group D, n = 40) were analyzed using ultra-performance liquid chromatography high-resolution mass spectrometry. A novel, high-coverage, semi-targeted metabolomics method (SWATH to MRM) and a targeted metabolomics method were employed to identify and verify the differential metabolites between the four groups. Sixteen differentially expressed FF metabolites were identified. Increase of BMI was associated with upregulation of 5 metabolites, ganoderiol H, LPI (18:3), sedoheptulose 1,7-bisphosphate, austalide L and 2 - {[hydroxyl (3-hydroxy-4-methoxyphenylmethylidene] amino} acetic acid, and downregulation of 5 metabolites, 1-phenyl-1,3-elcosanedione, retinol acetate, p-Cresol sulfate, setariol and arachidonyl carnitine. These metabolites were enriched in different metabolic pathways of retinol metabolism and fatty acid metabolism. These obesity-related differential metabolites provide a pathogenesis mechanism that explains the decline of oocyte development during obesity. These results suggest that obesity affects follicular environment prior to pregnancy, a time-window that may be important for lifestyle interventions to decrease obesity levels.
Collapse
|
23
|
Nicholas LM, Ozanne SE. Early life programming in mice by maternal overnutrition: mechanistic insights and interventional approaches. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180116. [PMID: 30966886 DOI: 10.1098/rstb.2018.0116] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Animal models have been indispensable in elucidating the potential causative mechanisms underlying the effects of maternal diet on offspring health. Of these, the mouse has been widely used to model maternal overnutrition and/or maternal obesity and to study its effects across one or more generations. This review discusses recent findings from mouse models, which resemble the human situation, i.e. overnutrition/obesity across pregnancy and lactation. It also highlights the importance of embryo transfer models in identifying critical developmental period(s) during which specific metabolic changes are programmed in the offspring. The mouse is also an excellent tool for maternal intervention studies aimed at elucidating the longer-term effects on the offspring and for defining possible maternal factors underling the programming of metabolic adversity in offspring. While knowledge of the mouse genome and the molecular tools available have allowed great progress to be made in the field, it is clear that we need to define if the effects on the offspring are mediated by maternal obesity per se or if specific components of the maternal metabolic environment are more important. We can then begin to identify at-risk offspring and to design more effective interventions for the mother and/or her child. This article is part of the theme issue 'Developing differences: early-life effects and evolutionary medicine'.
Collapse
Affiliation(s)
- Lisa M Nicholas
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge , Addenbrooke's Hospital, Cambridge , UK
| | - Susan E Ozanne
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge , Addenbrooke's Hospital, Cambridge , UK
| |
Collapse
|
24
|
Jazwiec PA, Sloboda DM. Nutritional adversity, sex and reproduction: 30 years of DOHaD and what have we learned? J Endocrinol 2019; 242:T51-T68. [PMID: 31013473 DOI: 10.1530/joe-19-0048] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 03/25/2019] [Indexed: 12/12/2022]
Abstract
It is well established that early life environmental signals, including nutrition, set the stage for long-term health and disease risk - effects that span multiple generations. This relationship begins early, in the periconceptional period and extends into embryonic, fetal and early infant phases of life. Now known as the Developmental Origins of Health and Disease (DOHaD), this concept describes the adaptations that a developing organism makes in response to early life cues, resulting in adjustments in homeostatic systems that may prove maladaptive in postnatal life, leading to an increased risk of chronic disease and/or the inheritance of risk factors across generations. Reproductive maturation and function is similarly influenced by early life events. This should not be surprising, since primordial germ cells are established early in life and thus vulnerable to early life adversity. A multitude of 'modifying' cues inducing developmental adaptations have been identified that result in changes in reproductive development and impairments in reproductive function. Many types of nutritional challenges including caloric restriction, macronutrient excess and micronutrient insufficiencies have been shown to induce early life adaptations that produce long-term reproductive dysfunction. Many pathways have been suggested to underpin these associations, including epigenetic reprogramming of germ cells. While the mechanisms still remain to be fully investigated, it is clear that a lifecourse approach to understanding lifetime reproductive function is necessary. Furthermore, investigations of the impacts of early life adversity must be extended to include the paternal environment, especially in epidemiological and clinical studies of offspring reproductive function.
Collapse
Affiliation(s)
- Patrycja A Jazwiec
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- The Farncombe Family Digestive Diseases Research Institute, McMaster University, Hamilton, Canada
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- The Farncombe Family Digestive Diseases Research Institute, McMaster University, Hamilton, Canada
- Department of Pediatrics and Obstetrics and Gynecology, McMaster University, Hamilton, Canada
| |
Collapse
|
25
|
Gilbert RO. Symposium review: Mechanisms of disruption of fertility by infectious diseases of the reproductive tract. J Dairy Sci 2019; 102:3754-3765. [PMID: 30772031 DOI: 10.3168/jds.2018-15602] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 12/15/2018] [Indexed: 12/17/2022]
Abstract
Diseases of postpartum dairy cows impair reproductive processes, resulting in prolonged anestrus, reduced conception, and increased pregnancy attrition, regardless of whether the initial disease precedes insemination (even by many weeks), occurs close to insemination, or follows fertilization. Bacteria and their products activate pattern recognition receptors that respond to pathogen-associated molecular patterns (PAMP). These receptors include toll-like receptors (TLR), nucleotide-binding oligomerization domain (NOD)-like receptors and others, and their activation culminates in upregulation of proinflammatory cytokines such as IL-1β, IL-18, and tumor necrosis factor-α. These may have direct effects on the uterus and conceptus. Importantly, however, these inflammatory mediators, as well as the bacterial products, make their way to the ovary via the general circulation (even from distant sites) or possibly by using the countercurrent vascular mechanism that normally transports endometrial prostaglandin to the ipsilateral ovary. Endotoxin reaches concentrations in follicular fluid that exceed those found in the circulation or even in the infected uterus. Ovarian follicular cells also express TLR and can respond directly to bacterial products including endotoxin, impairing their function. Inflammation is accompanied by increased oxidative stress. The process of oocyte development from activation of primordial oocytes to potential ovulation spans 4 mo. Competence by an oocyte encompasses the ability to undergo not only fertilization but also a complex cytoplasmic maturation that lays the foundation for completion of meiosis at the appropriate time, the transition to mitosis in the zygote, and further development of the conceptus. Oocyte maturation relies on intimate association between cumulus cells and the oocyte, characterized by gap junctions through which molecules of various sizes pass. Signaling also occurs in the oocyte-to-cumulus cell direction. Because both granulosa and theca interna cells are capable of responding to inflammatory mediators, with observed alterations in some functions, it seems likely that disturbed ovarian follicular function may contribute to failure of oocytes to become fully competent, even if the insult occurs well before ovulation. Therefore, interruption of normal fertility by uterine infections may be mediated at the level of the uterine environment but the effect on the ovary and oocyte is likely to be even more important.
Collapse
Affiliation(s)
- Robert O Gilbert
- Ross University School of Veterinary Medicine, PO Box 334, Basseterre, St. Kitts and Nevis, West Indies.
| |
Collapse
|
26
|
La Vignera S, Condorelli RA, Cannarella R, Duca Y, Calogero AE. Sport, doping and female fertility. Reprod Biol Endocrinol 2018; 16:108. [PMID: 30449281 PMCID: PMC6241032 DOI: 10.1186/s12958-018-0437-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 10/24/2018] [Indexed: 01/02/2023] Open
Abstract
This article is a review that addresses the following topics, divided by paragraphs. The first paragraph investigates the effects of physical activity on ovarian function, analyzing in particular the changes concerning the serum concentrations of follicle-stimulating hormone, luteinizing hormone, prolactin, growth hormone, thyroid hormones, leptin, ghrelin, neuropeptide Y. The second paragraph analyzes the effects of doping on the hypothalamic-pituitary-ovarian axis. Finally, the last paragraph analyzes the PCOS category, evaluating the effects of hyperandrogenism in relation to athletic performance.
Collapse
Affiliation(s)
- Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", via S. Sofia 78, 95123, Catania, Italy.
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", via S. Sofia 78, 95123, Catania, Italy
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", via S. Sofia 78, 95123, Catania, Italy
| | - Ylenia Duca
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", via S. Sofia 78, 95123, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", via S. Sofia 78, 95123, Catania, Italy
| |
Collapse
|
27
|
Faraci C, Jin J, Woods DC. Calorie restriction does not influence oocyte quality in oocytes from POLG mitochondrial mutator mice. PLoS One 2018; 13:e0204373. [PMID: 30240410 PMCID: PMC6150528 DOI: 10.1371/journal.pone.0204373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/06/2018] [Indexed: 11/26/2022] Open
Abstract
It has recently been demonstrated that moderate adult onset caloric restriction (e.g. calorie restriction; CR) has a positive impact on female fertility in aged mice, due in large to preventing the age-associated decline in the quality of oocytes available for fertilization. The impact of CR on oocyte quality has been attributed, at least in part, to mitochondrial functions. In mitochondrial DNA (mtDNA) mutator mice (PolgD257A/D257A), which harbor a mutation in the proofreading mtDNA polymerase-gamma (POLG), mitochondrial mutations rapidly accumulate, resulting in a premature aging phenotype and female infertility. As CR has been shown to extend both lifespan and ‘healthspan’ as well as improve oocyte quality in aged mice, we investigated whether adult onset CR could improve oocyte quality in the POLG mouse. Female PolgD257A/D257A mice exhibited infertility based on an inability to produce litters through natural mating. Analysis of oocytes from 8–9-month-old PolgD257A/D257A mice on CR following hormone stimulation revealed no improvement in the number of oocytes ovulated. Furthermore, CR did not result in a greater percentage of metaphase II oocytes, with the majority of the oocytes prematurely arrested at the germinal vesicle stage. Finally, CR did not improve the abnormal mitochondrial distribution or pronounced defects in meiotic spindle assembly and chromosomal distribution observed in the ad libitum fed PolgD257A/D257A. Taken together, these data suggest that although CR benefits oocyte quality and fertility outcomes in naturally aged female mice, it does not sufficiently improve oocyte quality in PolgD257A/D257A.
Collapse
Affiliation(s)
- Christine Faraci
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, United States of America
| | - Joyce Jin
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, United States of America
| | - Dori C. Woods
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
28
|
Gonzalez MB, Lane M, Knight EJ, Robker RL. Inflammatory markers in human follicular fluid correlate with lipid levels and Body Mass Index. J Reprod Immunol 2018; 130:25-29. [PMID: 30174020 DOI: 10.1016/j.jri.2018.08.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/15/2018] [Accepted: 08/20/2018] [Indexed: 12/29/2022]
Abstract
The detrimental consequences of obesity on female fertility are well known, but the functional changes that occur in the ovary in response to elevated BMI are not clear. Obesity induces multiple components of a systemic inflammatory state that is a key pathway by which it initiates tissue dysfunction in adipose, liver and muscle; however whether obesity induces similar inflammatory changes in the ovary has not been fully investigated. This is important to understand because it is increasingly clear that obesity at conception impacts not only pregnancy rates but also influences pre-implantation embryo development. To further understand the characteristics of inflammation in the ovaries of obese women we analysed a panel of cytokines (IL6, IL10 and TNFα), adipokines (adiponectin, leptin and monocyte chemotactic factor 1 (MCP-1)) and acute phase proteins (C-Reactive Protein (CRP) and sICAM-1) in the ovarian follicular fluid obtained at oocyte aspiration from women (n = 48) who were lean, overweight or obese. We hypothesised that adipokines and pro-inflammatory cytokines would be correlated with and/or dysregulated by increasing Body Mass Index (BMI). Surprisingly however, the majority were not related to BMI but instead were positively correlated with lipid levels in follicular fluid, namely triglycerides and free fatty acids. Further, as is typical of metabolic inflammation, the inflammatory markers that were associated with intra-follicular lipids included both pro-inflammatory (CRP, IL6, TNFα) and anti-inflammatory (adiponectin, IL10) mediators. The direct consequences of an ovarian microenvironment containing high levels of lipids and inflammatory mediators are not known but could impact luteinisation, ovulation and/or oocyte developmental competence.
Collapse
Affiliation(s)
- Macarena B Gonzalez
- Robinson Research Institute, School of Medicine, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Michelle Lane
- Robinson Research Institute, School of Medicine, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Emma J Knight
- Robinson Research Institute, School of Public Health, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Rebecca L Robker
- Robinson Research Institute, School of Medicine, The University of Adelaide, Adelaide, SA, 5005, Australia.
| |
Collapse
|
29
|
Faraci C, Annis S, Jin J, Li H, Khrapko K, Woods DC. Impact of exercise on oocyte quality in the POLG mitochondrial DNA mutator mouse. Reproduction 2018; 156:185-194. [PMID: 29875308 PMCID: PMC6074767 DOI: 10.1530/rep-18-0061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/05/2018] [Indexed: 12/12/2022]
Abstract
The mtDNA 'mutator' mouse, also called the 'POLG' mouse, is a well-characterized model frequently used for studies of progeroid aging. Harboring a mutation in the proofreading domain of the mitochondrial polymerase, polymerase-γ (Polg), POLG mice acquire mtDNA mutations at an accelerated rate. This results in premature mitochondrial dysfunction and a systemic aging phenotype. Previous work has demonstrated that the progeroid phenotype in POLG is attenuated following endurance exercise, the only reported intervention to extend health span and lifespan of these mice. Herein, oocyte quality was evaluated in sedentary and exercised POLG mice. In mice homozygous for the Polg mutation, litter size is dramatically reduced as compared to heterozygous Polg mice. Following ovarian hyper-stimulation, oocytes were retrieved until 9 months of age in exercised and sedentary groups, with no oocytes ovulated thereafter. Although ovulated oocyte numbers were not impacted by exercise, we did find a modest improvement in both the ovarian follicle reserve and in oocyte quality based on meiotic spindle assembly, chromosomal segregation and mitochondrial distribution at 7 months of age in exercised POLG mice as compared to sedentary counterparts. Of note, analysis of mtDNA mutational load revealed no differences between exercised and sedentary groups. Collectively, these data indicate that exercise differentially influences somatic tissues of the POLG mouse as compared to oocytes, highlighting important mechanistic differences between mitochondrial regulatory mechanisms in the soma and the germline.
Collapse
Affiliation(s)
- Christine Faraci
- Department of BiologyNortheastern University, Boston, Massachusetts, USA
| | - Sofia Annis
- Department of BiologyNortheastern University, Boston, Massachusetts, USA
| | - Joyce Jin
- Department of BiologyNortheastern University, Boston, Massachusetts, USA
| | - Housaiyin Li
- Department of BiologyNortheastern University, Boston, Massachusetts, USA
| | - Konstantin Khrapko
- Department of BiologyNortheastern University, Boston, Massachusetts, USA
| | - Dori C Woods
- Department of BiologyNortheastern University, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Stuart TJ, O’Neill K, Condon D, Sasson I, Sen P, Xia Y, Simmons RA. Diet-induced obesity alters the maternal metabolome and early placenta transcriptome and decreases placenta vascularity in the mouse. Biol Reprod 2018; 98:795-809. [PMID: 29360948 PMCID: PMC6454478 DOI: 10.1093/biolre/ioy010] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 12/19/2017] [Accepted: 01/17/2018] [Indexed: 12/19/2022] Open
Abstract
Maternal obesity is associated with an increased risk of obesity and metabolic disease in offspring. Increasing evidence suggests that the placenta plays an active role in fetal programming. In this study, we used a mouse model of diet-induced obesity to demonstrate that the abnormal metabolic milieu of maternal obesity sets the stage very early in pregnancy by altering the transcriptome of placenta progenitor cells in the preimplantation (trophectoderm [TE]) and early postimplantation (ectoplacental cone [EPC]) placenta precursors, which is associated with later changes in placenta development and function. Sphingolipid metabolism was markedly altered in the plasma of obese dams very early in pregnancy as was expression of genes related to sphingolipid processing in the early placenta. Upregulation of these pathways inhibits angiogenesis and causes endothelial dysfunction. The expression of many other genes related to angiogenesis and vascular development were disrupted in the TE and EPC. Other key changes in the maternal metabolome in obese dams that are likely to influence placenta and fetal development include a marked decrease in myo and chiro-inositol. These early metabolic and gene expression changes may contribute to phenotypic changes in the placenta, as we found that exposure to a high-fat diet decreased placenta microvessel density at both mid and late gestation. This is the first study to demonstrate that maternal obesity alters the transcriptome at the earliest stages of murine placenta development.
Collapse
Affiliation(s)
- Tami J Stuart
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kathleen O’Neill
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Condon
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Issac Sasson
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Payel Sen
- Epigenetics Center, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yunwei Xia
- College of Arts and Sciences, Cornell University, Ithaca, New York, USA
| | - Rebecca A Simmons
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
31
|
Abstract
The prevalence of obesity is high among reproductive-age women and is associated with impaired reproductive function. Obesity is multifactorial in origin, yet many cases of obesity result from overconsumption of a diet high in fat. Excess dietary fat increases both adipose and nonadipose tissue lipid content and, through lipotoxicity, leads to cell dysfunction and death. High dietary fat intake, with or without the development of obesity, impairs female hypothalamic-pituitary-ovarian (HPO) axis functionality and fertility. Based on the current evidence, it appears the reproductive dysfunction involves increased leptin and insulin signaling at the various levels of the HPO axis, as well as changes in peroxisome proliferator-activated receptor γ actions and increased inflammation, yet other mechanisms may also be involved. This review summarizes the current body of knowledge on impaired female reproductive function after high-fat diet exposure, as well as discusses proposed mechanisms through which this may occur.
Collapse
Affiliation(s)
- Natalie M Hohos
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Malgorzata E Skaznik-Wikiel
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, Colorado 80045
| |
Collapse
|
32
|
Liu HX, Wang YM, Hu JP, Huang LY, Fang NY. Adipocyte differentiation is regulated by mitochondrial trifunctional protein α-subunit via sirtuin 1. Exp Cell Res 2017; 357:271-281. [PMID: 28552586 DOI: 10.1016/j.yexcr.2017.05.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 05/19/2017] [Accepted: 05/23/2017] [Indexed: 12/18/2022]
Abstract
Mitochondrial trifunctional protein α-subunit (MTPα) is involved in the fatty acid β-oxidation (FAO) pathway. Two MTPα activities, 3-hydroxyacyl-CoA dehydrogenase and long-chain hydratase, have been linked with the occurrence and development of obesity and obesity-related disorders. These activities catalyze two steps in the FAO pathway (the second and third reactions). However, the role of MTPα in the pathogenesis of obesity has not been evaluated, and the functional role of MTPα in adipocyte differentiation has not been determined. Here, we analyzed the functional role of MTPα using in vitro and in vivo models of adipogenesis. MTPα expression was upregulated during the differentiation of 3T3-L1 preadipocyte cells into adipocytes. MTPα gene silencing stimulated peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT-enhancer-binding protein alpha(C/EBPα) expression, which promoted adipocyte differentiation. By contrast, MTPα overexpression blocked adipogenesis in 3T3-L1 cells. Further analysis showed that MTPα positively regulated sirtuin 1 (SIRT1). Injection of preadipocytes overexpressing MTPα into athymic mice significantly impaired de novo fat pad formation compared with that of the control, and furthermore MTPα knockdown enhances fat pad formation at a time point earlier than 5-week, such as week-2 and week-3, when the control fat pad is not fully developed. In summary, our data indicate that MTPα is a novel factor that negatively regulates adipocyte differentiation. We propose a pathway in which MTPα inhibits adipogenesis by promoting SIRT1 expression, which represses PPARγ and attenuates adipogenesis.
Collapse
Affiliation(s)
- Hong-Xia Liu
- Department of Geriatrics, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Pudong New Area, Shanghai 200127, China
| | - Yan-Mei Wang
- Department of Geriatrics, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Pudong New Area, Shanghai 200127, China
| | - Jian-Ping Hu
- Department of Geriatrics, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Pudong New Area, Shanghai 200127, China
| | - Li-Ying Huang
- Department of Geriatrics, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Pudong New Area, Shanghai 200127, China
| | - Ning-Yuan Fang
- Department of Geriatrics, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Pudong New Area, Shanghai 200127, China.
| |
Collapse
|
33
|
Okeyo-Owuor T, Benesh E, Bibbey S, Reid M, Halabi J, Sutcliffe S, Moley K. Exposure to maternal obesogenic diet worsens some but not all pre-cancer phenotypes in a murine genetic model of prostate cancer. PLoS One 2017; 12:e0175764. [PMID: 28489892 PMCID: PMC5425180 DOI: 10.1371/journal.pone.0175764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/30/2017] [Indexed: 01/27/2023] Open
Abstract
Prostate cancer research has been predominantly focused on adult exposures and risk factors. However, because the prostate develops during gestation and early life, exposure to external factors, such as obesity, during development could affect the prostate cancer progression in adults. Our previous work demonstrated that exposure to a high fat/high sugar (HF/HS) diet during gestation and until weaning stimulated prostate hyperplasia and altered the Pten/Akt pathway in adult mice fed a normal diet after weaning. Here, we asked whether maternal exposure to HF/HS would worsen prostate phenotypes in mice lacking Pten, a widely accepted driver of prostate cancer. We found that, at six weeks of age, both Chow (control)-and HF/HS-exposed Pten knockout mice showed evidence of murine PIN that included ducts with central comedo necrosis but that the HF/HS exposure did not influence murine PIN progression. The Pten knockout mice exposed to HF/HS in utero had significantly more mitotic cells than Pten knockouts exposed to Chow diet. In the Pten null background, the maternal HF/HS diet enhanced proliferation but did not have an additive effect on Akt activation. We observed neuroendocrine differentiation in Pten knockout mice, a phenotype that had not been previously described in this model.
Collapse
Affiliation(s)
- Theresa Okeyo-Owuor
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Emily Benesh
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Scott Bibbey
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Michaela Reid
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jacques Halabi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Siobhan Sutcliffe
- Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Kelle Moley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
34
|
Boudoures AL, Saben J, Drury A, Scheaffer S, Modi Z, Zhang W, Moley KH. Obesity-exposed oocytes accumulate and transmit damaged mitochondria due to an inability to activate mitophagy. Dev Biol 2017; 426:126-138. [PMID: 28438607 DOI: 10.1016/j.ydbio.2017.04.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/22/2017] [Accepted: 04/17/2017] [Indexed: 11/26/2022]
Abstract
Mitochondria are the most prominent organelle in the oocyte. Somatic cells maintain a healthy population of mitochondria by degrading damaged mitochondria via mitophagy, a specialized autophagy pathway. However, evidence from previous work investigating the more general macroautophagy pathway in oocytes suggests that mitophagy may not be active in the oocyte. This would leave the vast numbers of mitochondria - poised to be inherited by the offspring - vulnerable to damage. Here we test the hypothesis that inactive mitophagy in the oocyte underlies maternal transmission of dysfunctional mitochondria. To determine whether oocytes can complete mitophagy, we used either CCCP or AntimycinA to depolarize mitochondria and trigger mitophagy. After depolarization, we did not detect co-localization of mitochondria with autophagosomes and mitochondrial DNA copy number remained unchanged, indicating the non-functional mitochondrial population was not removed. To investigate the impact of an absence of mitophagy in oocytes with damaged mitochondria on offspring mitochondrial function, we utilized in vitro fertilization of high fat high sugar (HF/HS)-exposed oocytes, which have lower mitochondrial membrane potential and damaged mitochondria. Here, we demonstrate that blastocysts generated from HF/HS oocytes have decreased mitochondrial membrane potential, lower metabolites involved in ATP generation, and accumulation of PINK1, a mitophagy marker protein. This mitochondrial phenotype in the blastocyst mirrors the phenotype we show in HF/HS exposed oocytes. Taken together, these data suggest that the mechanisms governing oocyte mitophagy are fundamentally distinct from those governing somatic cell mitophagy and that the absence of mitophagy in the setting of HF/HS exposure contributes to the oocyte-to-blastocyst transmission of dysfunctional mitochondria.
Collapse
Affiliation(s)
- Anna L Boudoures
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Jessica Saben
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Andrea Drury
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Suzanne Scheaffer
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Zeel Modi
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Wendy Zhang
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Kelle H Moley
- Center for Reproductive and Health Sciences, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
35
|
Abstract
Recent human and animal studies investigating the roles of the genome, epigenome, and environmental cues have identified associations between offspring predisposition to life-long obesity/metabolic disease and epigenetic modifications such as DNA methylation. This review explores the mechanisms by which maternal exposures impair the health of not only the next generation but also potentially future generations of offspring.
Collapse
Affiliation(s)
- Kathleen Jaeger
- Washington University School of Medicine, St. Louis, Missouri
| | - Jessica L Saben
- Washington University School of Medicine, St. Louis, Missouri
| | - Kelle H Moley
- Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
36
|
Saben JL, Boudoures AL, Asghar Z, Thompson A, Drury A, Zhang W, Chi M, Cusumano A, Scheaffer S, Moley KH. Maternal Metabolic Syndrome Programs Mitochondrial Dysfunction via Germline Changes across Three Generations. Cell Rep 2016; 16:1-8. [PMID: 27320925 DOI: 10.1016/j.celrep.2016.05.065] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 04/04/2016] [Accepted: 05/16/2016] [Indexed: 01/13/2023] Open
Abstract
Maternal obesity impairs offspring health, but the responsible mechanisms are not fully established. To address this question, we fed female mice a high-fat/high-sugar diet from before conception until weaning and then followed the outcomes in the next three generations of offspring, all fed a control diet. We observed that female offspring born to obese mothers had impaired peripheral insulin signaling that was associated with mitochondrial dysfunction and altered mitochondrial dynamic and complex proteins in skeletal muscle. This mitochondrial phenotype persisted through the female germline and was passed down to the second and third generations. Our results indicate that maternal programming of metabolic disease can be passed through the female germline and that the transfer of aberrant oocyte mitochondria to subsequent generations may contribute to the increased risk for developing insulin resistance.
Collapse
Affiliation(s)
- Jessica L Saben
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Anna L Boudoures
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Zeenat Asghar
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Alysha Thompson
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Andrea Drury
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Wendy Zhang
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Maggie Chi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Andrew Cusumano
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Suzanne Scheaffer
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Kelle H Moley
- Department of Obstetrics and Gynecology, Washington University School of Medicine, 425 South Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
37
|
Affiliation(s)
- John S Davis
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center and Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska 68198-3255
| |
Collapse
|