1
|
Kantapan J, Katsube T, Wang B. High-Fat Diet and Altered Radiation Response. BIOLOGY 2025; 14:324. [PMID: 40282189 PMCID: PMC12024794 DOI: 10.3390/biology14040324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025]
Abstract
High-fat diets (HFDs) have become increasingly prevalent in modern societies, driving rising rates of obesity and metabolic syndrome. Concurrently, radiation exposure from medical treatments and environmental sources poses health risks shaped by both biological and environmental factors. This review explores the intersection between HFDs and radiation sensitivity/susceptibility, focusing on how diet-induced metabolic alterations influence the body's response to radiation. Evidence from preclinical and clinical studies indicates that HFDs significantly alter metabolism, leading to increased oxidative stress and immune system dysregulation. These metabolic changes can exacerbate radiation-induced oxidative stress, inflammation, and DNA damage, potentially increasing radiation sensitivity in normal tissues. Conversely, obesity and HFD-induced metabolic disruptions may activate cellular pathways involved in DNA repair, cell survival, and inflammatory responses, fostering tumor resistance and modifying the tumor microenvironment, which may impair the efficacy of radiation therapy in cancer treatment. Understanding the interplay between diet and radiation exposure is critical for optimizing public health guidelines and improving therapeutic outcomes. These findings underscore the need for further research into dietary interventions that may mitigate radiation-associated risks.
Collapse
Affiliation(s)
- Jiraporn Kantapan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Takanori Katsube
- Institute for Radiological Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan;
| | - Bing Wang
- Institute for Radiological Science, National Institutes for Quantum Science and Technology (QST), Chiba 263-8555, Japan;
| |
Collapse
|
2
|
Lai T, Mao A, Yang L, Ren Y, Yang X, Song W, Luo Y. Association of maternal metabolic risk factors with offspring body mass index (BMI) trajectories in early childhood: a retrospective cohort study. BMJ Open 2025; 15:e088641. [PMID: 40082003 PMCID: PMC11907022 DOI: 10.1136/bmjopen-2024-088641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 02/14/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVE This study aimed to identify body mass index (BMI) growth trajectories from birth to 24 months of age and examine the independent and additive effects of four maternal metabolic risk factors, namely prepregnancy BMI, the rate of gestational weight gain, gestational diabetes mellitus (GDM) and gestational hypertension, on offspring growth trajectories in childhood in China. DESIGN A retrospective cohort study was conducted. SETTING The study used Maternal and Child Health Management Database in Chengdu, China, including the mothers' antenatal care data, birth certificate records and 0-3-year-old children's healthcare data. PARTICIPANTS The study included mothers who gave birth between January 2014 and December 2014, and followed their offspring through 31 December 2016. The final analysis included 4492 mother-child pairs. PRIMARY OUTCOME MEASURES The primary outcomes were children's BMI measurements from birth to 24 months of age. We performed group-based trajectories modelling to identify children's BMI growth trajectories. Then, we applied logistic regression to examine the associations between maternal metabolic risk factors and offspring BMI trajectories in childhood. RESULTS Four distinct trajectories were identified: stable low (16.83%), stable average (40.69%), stable high (32.06%) and early increase (10.42%) trajectories. Relative to the stable average trajectory, maternal prepregnancy overweight (adjusted OR (aOR)=2.001, 95% CI 1.482-2.702, p<0.001), an excessive rate of gestational weight gain (aOR=1.496, 95% CI 1.138-1.966, p=0.004) and GDM (aOR=1.470, 95% CI 1.097-1.970, p=0.010) were positively associated with their offspring being in the early increase trajectory. In addition, the children's risk of being included in the early increase trajectory showed an increasing trend with an increasing number of adverse maternal metabolic risk factors. CONCLUSION Exposure to maternal prepregnancy overweight, excessive rate of weight gain and GDM resulted in a greater risk of offspring exhibiting an early increase trajectory for BMI. Decreasing maternal metabolic risk before and during pregnancy and monitoring childhood growth trajectories may prevent or delay the onset of childhood obesity.
Collapse
Affiliation(s)
- Ting Lai
- Department of Healthcare, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| | - Ang Mao
- Department of Medical Administration, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| | - Liu Yang
- Department of Healthcare, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| | - Yan Ren
- Department of Healthcare, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| | - Xiao Yang
- Department of Obstetrics, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| | - Wei Song
- Department of Women and Children Health, Chengdu Municipal Health Commission, Chengdu, Sichuan, China
| | - Yingjuan Luo
- Department of Healthcare, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Zhang JK, Li XP, Tang Y, Zeng LP, Liu X, Zhang JL, Chen CY, Zheng S, Liu ZZ, Gong X, Jose PA, Guo L, Zeng CY. Semaglutide ameliorates metabolic disorders in offspring via regulation of oocyte ROS of pre-pregnancy obesity mice. Acta Pharmacol Sin 2025:10.1038/s41401-025-01501-1. [PMID: 39984623 DOI: 10.1038/s41401-025-01501-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025]
Abstract
Pre-pregnancy obesity (PPO) seriously threatens the health of both mother and offspring. Pre-pregnancy weight management is particularly important for the prevention of metabolic diseases in offspring. Semaglutide is one of the most effective glucagon-like peptide-1 agonizts for the management of obesity and metabolic diseases, but little is known about its effect on the long-term health of offspring. In this study we investigated the effects of semaglutide administered before pregnancy on the offspring health from PPO mice. PPO mice model was established by feeding with high-fat diet for 16 weeks, and then injected with semaglutide (30 nmol/kg-1·d-1, sc.) for 22 days before pregnancy. After the treatment, the mice were mated with normal males or underwent in vitro fertilization (IVF) for offspring reproduction. We showed that the semaglutide treatment not only improved the lipid and glucose metabolic disorders and fertility of PPO mice, but also significantly reversed the overweight, impaired energy balance, adipose inflammatory state, lipid and glucose metabolic disorders and insulin resistance of their IVF offspring. By conducting RNA-seq analysis, SOD activity and malondialdehyde assays in ovaries, as well as ROS staining in oocytes, we revealed that the semaglutide treatment reduced the elevated oxidative stress in ovaries and high ROS levels in oocytes from PPO mice, possibly through activating the PI3K/AKT pathway and improving the state of SOD. Interestingly, incubation of oocytes from semaglutide-treated dams with H2O2 (100 μM) in vitro during IVF blocked the protective effects of semaglultide against the metabolic disorders in the offspring. In conclusion, semaglutide treatment before pregnancy effectively alleviates obesity-related metabolic disorders in offspring. The regulation of ROS in oocytes plays a crucial role in the protective effects of semaglutide.
Collapse
Affiliation(s)
- Jun-Kai Zhang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Department of Cardiology, Sichuan Provincial Corps Hospital of Chinese People's Armed Police Force (Sichuan Corps Hospital of PAP), Leshan, 614000, China
| | - Xiao-Ping Li
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Department of Cardiology, 921 Hospital of Joint Logistics Support Force People's Liberation Army of China (the Second Affiliated Hospital of Hunan Normal University), Changsha, 410081, China
| | - Yang Tang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Li-Ping Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xuan Liu
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Jian-Li Zhang
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Cai-Yu Chen
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Zhi-Zhao Liu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Xue Gong
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, The George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Li Guo
- Endocrinology Department, The First Affiliated Hospital of The Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Chun-Yu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China.
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China, Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, 400042, China.
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
4
|
Lee M, Son S, Oh S, Shin E, Shin H, Kwon O, Hwang S, Song H, Lim HJ. Diet-Induced Obesity Alters Granulosa Cell Transcriptome and Ovarian Immune Environment in Mice. Life (Basel) 2025; 15:330. [PMID: 40141675 PMCID: PMC11943477 DOI: 10.3390/life15030330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 03/28/2025] Open
Abstract
Obesity affects female reproductive performance by impairing the ovarian and uterine environments. Using a diet-induced obesity mouse model, we examined whether a high-fat diet (HFD) regimen affects the gene expression profile in ovarian granulosa cells (GCs) and whether short-term HFD has similar effects on gene expression as long-term HFD. C57BL/6J mice were fed a HFD or normal diet (ND) for 16-18 weeks (long-term group) or 4 weeks (short-term group). GCs were collected from each group of mice for RNA-sequencing. RT-PCR and immunofluorescence staining were performed to validate the results. RNA-sequencing analyses of the GCs revealed that several immediate early genes, including early growth response 1 (Egr1), an important mediator of ovulation, were significantly downregulated in HFD GCs. Protein tyrosine phosphatase receptor type C (Ptprc) and hematopoietic type prostaglandin D synthase (Hpgds), both of which are associated with increased inflammation, were significantly upregulated in HFD GCs. Downregulation of Egr1 was also confirmed in the GCs of short-term HFD mice, suggesting that it constitutes an early change in response to a HFD. Increased expression of several transcription factors in HFD GCs suggests that a HFD may affect the overall transcriptional landscape. The results may indicate possible modulation of the immune environment in HFD ovaries. These results provide novel insights into the molecular changes in GCs in obese environments.
Collapse
Affiliation(s)
- Minseo Lee
- Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Sujin Son
- Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Surim Oh
- Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Eunbin Shin
- Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyejin Shin
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Ohrim Kwon
- Department of Life Science, Graduate School, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Sohyun Hwang
- Department of Life Science, Graduate School, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- Department of Pathology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si 13520, Gyeonggi-do, Republic of Korea
| | - Haengseok Song
- Department of Life Science, Graduate School, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Hyunjung Jade Lim
- Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
- Department of Biomedical Science and Technology, Institute of Biomedical Science & Technology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
5
|
Kuyoro O, Shalamov MM, Brogan C, Goldman R. A retrospective cohort study examining the outcomes of patients who present for fertility care and exceed a set body mass index threshold for treatment. F S Rep 2024; 5:417-421. [PMID: 39781074 PMCID: PMC11705578 DOI: 10.1016/j.xfre.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 01/12/2025] Open
Abstract
Objective To evaluate the characteristics of patients who exceeded the body mass index (BMI) threshold for fertility treatment at their initial visit and identify those for whom treatment would be constrained. Design Retrospective cohort study. Setting Academic medical center. Patients All new patients who presented for infertility treatment at an academic center between January 2020 and December 2022 and had BMI measured and recorded. Main Outcome Measures Likelihood of weight loss and treatment initiation for patients who exceed a set BMI threshold of 40 kg/m2. Results Of the 1,268 patients who had their BMI recorded at initial visit, 48% identified as non-Hispanic White, 15% as non-Hispanic Black, 13% as Asian, 11% as Hispanic, 0.2% as Native American, 4% as other; 9% were of unknown race/ethnicity. Overall, 6% of women exceeded the 40 kg/m2 cutoff. Among Latino women, 7.5% exceeded the cutoff; among non-Hispanic Black women, 12% exceeded the cutoff. These percentages were greater than the percentage of non-Hispanic White women who exceeded the BMI cutoff (4.8%). Conclusions Body mass index thresholds disproportionately affect the ability of ethnic minorities to use fertility treatment and could potentially be worsening barriers to care these population of patients already face.
Collapse
Affiliation(s)
- Olutunmike Kuyoro
- Northwell, New Hyde Park, New York
- Department of Obstetrics and Gynecology, Northwell Health Fertility, New York, New York
- Zucker School of Medicine, Uniondale, New York
| | | | - Cailey Brogan
- Northwell, New Hyde Park, New York
- Department of Obstetrics and Gynecology, Northwell Health Fertility, New York, New York
- Zucker School of Medicine, Uniondale, New York
| | - Randi Goldman
- Northwell, New Hyde Park, New York
- Department of Obstetrics and Gynecology, Northwell Health Fertility, New York, New York
- Zucker School of Medicine, Uniondale, New York
| |
Collapse
|
6
|
Gong J, Xu W, Chen Y, Chen S, Wu Y, Chen Y, Li Y, He Y, Yu H, Xie L. Maternal Gestational Diabetes Mellitus and High-Fat Diet Influenced Hepatic Polyunsaturated Fatty Acids Profile in the Offspring of C57BL/6J Mice. Mol Nutr Food Res 2024; 68:e2400386. [PMID: 39246092 DOI: 10.1002/mnfr.202400386] [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: 05/23/2024] [Revised: 08/04/2024] [Indexed: 09/10/2024]
Abstract
SCOPE This research examines the effects of maternal high-fat (HF) diet and gestational diabetes mellitus (GDM) on offspring lipid metabolism and polyunsaturated fatty acids (PUFA) profile. METHODS AND RESULTS GDM is induced using the insulin receptor antagonist S961. Weaning offspring are categorized into HF-GDM, HF-CON, NC-GDM, and NC-CON groups based on maternal diet or GDM. Adult offspring are then grouped into NC-CON-NC, NC-CON-HF, NC-GDM-NC, NC-GDM-HF, HF-CON-NC, HF-CON-HF, HF-GDM-NC, and HF-GDM-HF according to dietary patterns. Gas chromatography determines PUFA composition. Western blot assesses PI3K/Akt signaling pathway-related protein expression. Feeding a normal chow diet until adulthood improves the distribution of hepatic PUFA during weaning across the four groups. PI3K expression is upregulated during weaning in HF-CON and HF-GDM, particularly in HF-CON-NC and HF-GDM-NC, compared to NC-CON-NC during adulthood. Akt expression increases in NC-GDM-NC after weaning with a normal diet. The hepatic PUFA profile in HF-CON-HF significantly distinguishes among the maternal generation health groups. Maternal HF diet exacerbates the combined impact of maternal GDM and offspring HF diet on hepatic PUFA and PI3K/Akt signaling pathway-related proteins during adulthood. CONCLUSIONS Early exposure to HF diets and GDM affects hepatic PUFA profiles and PI3K/Akt signaling pathway protein expression in male offspring during weaning and adulthood.
Collapse
Affiliation(s)
- JiaYu Gong
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun City, Jilin Province, 130021, China
| | - WenHui Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun City, Jilin Province, 130021, China
| | - YiFei Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun City, Jilin Province, 130021, China
| | - ShuTong Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun City, Jilin Province, 130021, China
| | - YanYan Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun City, Jilin Province, 130021, China
| | - YiRu Chen
- Clinical Nutrition Department, Third Hospital of Jilin University, Changchun City, Jilin Province, 130032, China
| | - YueTing Li
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun City, Jilin Province, 130021, China
| | - Yuan He
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun City, Jilin Province, 130021, China
| | - HaiTao Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun City, Jilin Province, 130021, China
| | - Lin Xie
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun City, Jilin Province, 130021, China
| |
Collapse
|
7
|
Meneghini MA, Flores Quiroga JP, Heinecke F, Galarza RA, White V, Faletti AG. Impact of diet-induced maternal obesity on the reproductive capacity of F1 female offspring and the early development of the second generation. J Nutr Biochem 2024; 132:109700. [PMID: 39019120 DOI: 10.1016/j.jnutbio.2024.109700] [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: 12/01/2023] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
The aim of this study was to examine the impact of maternal obesity on the reproductive capacity of the female offspring (F1) and on the early development of the second generation (F2). To this end, rats were fed either standard (SD) or cafeteria (CD) diet. CD rats and their offspring were divided into 2 groups: rats with 18% and ≥25% overweight (CD18 and CD25, respectively) and offspring from CD18 and CD25 rats (OCD18 and OCD25, respectively). Both OCD groups achieved greater weight gain than controls, without changes in the serum levels of glucose, cholesterol or triglycerides. However, they showed increased gonadal cholesterol concentration and fat content compared to controls. Female OCD groups showed a slight prolongation of the estrous cycle and different pattern of changes in the weight gain during pregnancy. The OCD25 group displayed an increased fertility index and preimplantation losses, and changes in some fetal measurements. Some OCD25 dams gave birth to a larger litter of pups and displayed a lower viability index and lactation rate than controls. OCD25 dams also showed an increase in estradiol and a decrease in testosterone and anti-Müllerian hormone. OCD25 rats showed increased mRNA levels of steroidogenenic enzymes. The offspring from OCD25 females (F2OCD25 offspring) showed early vaginal opening and higher ovulation rate in females, and lower ano-genital distances in males, compared to controls. In conclusion, these results reflect that maternal obesity impacts on the reproductive health of successive generations, probably as a result of epigenetic changes in different systems, including germ cells.
Collapse
Affiliation(s)
- María Agustina Meneghini
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Facultad de Medicina, Buenos Aires, Argentina
| | - Jeremías Pablo Flores Quiroga
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Facultad de Medicina, Buenos Aires, Argentina
| | - Florencia Heinecke
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Facultad de Medicina, Buenos Aires, Argentina
| | - Rocío Alejandra Galarza
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Facultad de Medicina, Buenos Aires, Argentina
| | - Verónica White
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Facultad de Medicina, Buenos Aires, Argentina
| | - Alicia Graciela Faletti
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Facultad de Medicina, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Shan L, Fan H, Guo J, Zhou H, Li F, Jiang Z, Wu D, Feng X, Mo R, Liu Y, Zhang T, Zhou Y. Impairment of oocyte quality caused by gut microbiota dysbiosis in obesity. Genomics 2024; 116:110941. [PMID: 39306049 DOI: 10.1016/j.ygeno.2024.110941] [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: 06/18/2024] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 10/08/2024]
Abstract
Obesity poses risks to oocyte maturation and embryonic development in mice and humans, linked to gut microbiota dysbiosis and altered host metabolomes. However, it is unclear whether symbiotic gut microbes have a pivotal role in oocyte quality. In mouse models of fecal microbiota transplantation, we demonstrated aberrant meiotic apparatus and impaired maternal mRNA in oocytes, which is coincident with the poor developmental competence of embryos. Using metabolomics profiling, we discovered that the cytosine and cytidine metabolism was disturbed, which could account for the fertility defects observed in the high-fat diet (HFD) recipient mice. Additionally, cytosine and cytidine are closely related with gut microbiota dysbiosis, which is accompanied by a notable reduction of abundance of Christensenellaceae R-7 group in the HFD mice. In summary, our findings provided evidence that modifying the gut microbiota may be of value in the treatment of infertile female individuals with obesity.
Collapse
Affiliation(s)
- Liying Shan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R(2)BGL), College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Haitao Fan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R(2)BGL), College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Jing Guo
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, 410008 Changsha, China
| | - Heyang Zhou
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R(2)BGL), College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Fengguo Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R(2)BGL), College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Zhimin Jiang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R(2)BGL), College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Duo Wu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R(2)BGL), College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Xinlei Feng
- Shandong Animal Products Quality and Safety Center, Jinan 250100, China
| | - Ren Mo
- Department of Urology, Inner Mongolia people's Hospital, Inner Mongolia Urological Institute, Hohhot, Inner Mongolia 010017, China.
| | - Yongbin Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R(2)BGL), College of Life Sciences, Inner Mongolia University, Hohhot 010070, China.
| | - Teng Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R(2)BGL), College of Life Sciences, Inner Mongolia University, Hohhot 010070, China.
| | - Yang Zhou
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock (R(2)BGL), College of Life Sciences, Inner Mongolia University, Hohhot 010070, China.
| |
Collapse
|
9
|
Tang S, Wu H, Chen Q, Tang T, Li J, An H, Zhu S, Han L, Sun H, Ge J, Qian X, Wang X, Wang Q. Maternal Obesity Induces the Meiotic Defects and Epigenetic Alterations During Fetal Oocyte Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309184. [PMID: 38868907 PMCID: PMC11321662 DOI: 10.1002/advs.202309184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/23/2024] [Indexed: 06/14/2024]
Abstract
It has been widely reported that obesity adversely impacts reproductive performance of females. However, the effects of maternal obesity on fetal germ cells remain poorly understood. In the present study, by employing a high-fat diet (HFD)-based mouse model, it is discovered that maternal obesity disrupts the chromosomal synapsis and homologous recombination during fetal oogenesis. Moreover, transcriptomic profiling reveales the potential molecular network controlling this process. Of note, the global hypermethylation of genomic DNA in fetal oocytes from obese mouse is detected. Importantly, time-restricted feeding (TRF) of obese mice not only ameliorate the meiotic defects, but also partly restore the epigenetic remodeling in fetal oocytes. In sum, the evidence are provided showing the deficit fetal oogenesis in obese mother, implicating a mechanism underlying the intergenerational effects of environmental insults. TRF may represent a potentially effective approach for mitigating fertility issues in obese patients.
Collapse
Affiliation(s)
- Shoubin Tang
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
- Department of Nutrition and Food HygieneSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Huihua Wu
- Suzhou Municipal HospitalNanjing Medical UniversityNanjing211166China
| | - Qiuzhen Chen
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Tao Tang
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Jiashuo Li
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Huiqing An
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Longsen Han
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Hongzheng Sun
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Xu Qian
- Department of Nutrition and Food HygieneSchool of Public HealthNanjing Medical UniversityNanjing211166China
| | - Xi Wang
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine and Offspring HealthChangzhou Maternity and Child Health Care HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166China
| |
Collapse
|
10
|
Bozdemir N, Kablan T, Altintas MO, Sukur G, Cinar O, Uysal F. Altered DNA methylation and Dnmt expression in obese uterus may cause implantation failure. J Mol Histol 2024; 55:427-436. [PMID: 38850446 DOI: 10.1007/s10735-024-10212-6] [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: 10/28/2023] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Obesity is defined by increased adipose tissue volume and has become a major risk factor for reproduction. Recent studies have revealed a substantial link between obesity and epigenetics. The epigenome is dynamically regulated mainly by DNA methylation. DNA methylation, which is controlled by DNA methyltransferases (Dnmts), has been widely studied because it is essential for imprinting and regulation of gene expression. In our previous study, we showed that the levels of Dnmt1, Dnmt3a and global DNA methylation was dramatically altered in the testis and ovary of high-fat diet (HFD)-induced obese mice. However, the effect of HFD on Dnmts and global DNA methylation in mouse uterus has not yet been demonstrated. Therefore, in the present study, we aimed to evaluate the effect of HFD on the level of Dnmt1, Dnmt3a, Dnmt3b, Dnmt3l and global DNA methylation in uterus. Our results showed that HFD significantly altered the levels of Dnmts and global DNA methylation in the uterus. The total expression of Dnmt1, Dnmt3a and Dnmt3b was significantly upregulated, while level of Dnmt3l and global DNA methylation were dramatically decreased (p < 0.05). Furthermore, we observed that the expression of Dnmt3b and Dnmt3l was significantly increased in endometrium including gland and epithelium (p < 0.05). Although Dnmt3b was the only protein whose expression significantly increased, the level of global DNA methylation and Dnmt3l significantly decreased in stroma and myometrium (p < 0.05). In conclusion, our results show for the first time that obesity dramatically alters global DNA methylation and expression of Dnmts, and decreased DNA methylation and Dnmt expression may cause abnormal gene expression, especially in the endometrium.
Collapse
Affiliation(s)
- Nazlican Bozdemir
- Department of Histology and Embryology, Ankara Medipol University School of Medicine, Ankara, 06050, Turkey
| | - Tuba Kablan
- Department of Histology and Embryology, Ankara Medipol University School of Medicine, Ankara, 06050, Turkey
| | - Mehmet Ozgen Altintas
- Department of Physiology, Ankara Medipol University School of Medicine, Ankara, Turkey
- Department of Physiology, Istanbul Medipol University Institute of Health Sciences, Istanbul, Turkey
| | - Gozde Sukur
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
| | - Ozgur Cinar
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
| | - Fatma Uysal
- Department of Histology and Embryology, Ankara Medipol University School of Medicine, Ankara, 06050, Turkey.
| |
Collapse
|
11
|
Timme K, González-Alvarez ME, Keating AF. Pre-pubertal obesity compromises ovarian oxidative stress, DNA repair and chemical biotransformation. Toxicol Appl Pharmacol 2024; 489:116981. [PMID: 38838792 DOI: 10.1016/j.taap.2024.116981] [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: 03/11/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/07/2024]
Abstract
Obesity in adult females impairs fertility by altering oxidative stress, DNA repair and chemical biotransformation. Whether prepubertal obesity results in similar ovarian impacts is under-explored. The objective of this study was to induce obesity in prepubertal female mice and assess puberty onset, follicle number, and abundance of oxidative stress, DNA repair and chemical biotransformation proteins basally and in response to 7,12-dimethylbenz(a)anthracene (DMBA) exposure. DMBA is a polycyclic aromatic hydrocarbon that has been shown to be ovotoxic. Lactating dams (C57BL6J) were fed either a normal rodent containing 3.5% kCal from fat (lean), or a high fat diet comprised of 60% kCal from fat, and 9% kCal from sucrose. The offspring were weaned onto the diet of their dam and exposed at postnatal day 35 to either corn oil or DMBA (1 mg/kg) for 7 d via intraperitoneal injection. Mice on the HFD had reduced (P < 0.05) age at puberty onset as measured by vaginal opening but DMBA did not impact puberty onset. Heart, spleen, kidney, uterus and ovary weight were increased (P < 0.05) by obesity and liver weight was increased (P < 0.05) by DMBA exposure in obese mice. Follicle number was largely unaffected by obesity or DMBA exposure, with the exception of primary follicle number, which were higher (P < 0.05) in lean DMBA exposed and obese control relative to lean control mice. There were also greater numbers (P < 0.05) of corpora lutea in obese relative to lean mice. In lean mice, DMBA exposure reduced (P < 0.05) the level of CYP2E1, EPHX1, GSTP1, BRCA1, and CAT but this DMBA-induced reduction was absent in obese mice. Basally, obesity reduced (P < 0.05) the abundance of CYP2E1, EPHX1, GSTP1, BRCA1, SOD1 and CAT. There was greater (P < 0.05) fibrotic staining in obese DMBA-exposed ovaries and PPP2CA was decreased (P < 0.05) in growing follicles by both obesity and DMBA exposure. Thus, prepubertal obesity alters the capacity of the ovary to respond to DNA damage, ovotoxicant exposure and oxidative stress.
Collapse
Affiliation(s)
- Kelsey Timme
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | | | - Aileen F Keating
- Department of Animal Science, Iowa State University, Ames, IA, USA.
| |
Collapse
|
12
|
Schon SB, Cabre HE, Redman LM. The impact of obesity on reproductive health and metabolism in reproductive-age females. Fertil Steril 2024; 122:194-203. [PMID: 38704081 PMCID: PMC11527540 DOI: 10.1016/j.fertnstert.2024.04.036] [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: 03/14/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
Obesity is a highly prevalent chronic disease that impacts >40% of reproductive-aged females. The pathophysiology of obesity is complex and can be understood simply as a chronic energy imbalance whereby caloric intake exceeds caloric expenditure with an energy surplus stored in adipose tissue. Obesity may be categorized into degrees of severity as well as different phenotypes on the basis of metabolic health and underlying pathophysiology. Obesity and excess adiposity have a significant impact on fertility and reproductive health, with direct effects on the hypothalamic-pituitary-ovarian axis, the ovary and oocyte, and the endometrium. There are significant adverse pregnancy outcomes related to obesity, and excess weight gain before, during, and after pregnancy that can alter the lifelong risk for metabolically unhealthy obesity. Given the high prevalence and pervasive impact of obesity on reproductive health, there is a need for better and individualized care for reproductive-aged females that considers obesity phenotype, underlying pathophysiology, and effective and sustainable interventions to treat obesity and manage weight gain before, during, and after pregnancy.
Collapse
Affiliation(s)
- Samantha B Schon
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan.
| | - Hannah E Cabre
- Reproductive Endocrinology and Women's Health Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Leanne M Redman
- Reproductive Endocrinology and Women's Health Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| |
Collapse
|
13
|
Rosales-Nieto CA, Daigneault BW, Roberts JN, Sánchez-López R, Makela B, Pu Y, Ehrhardt R, Jabur Bittar JH, Veiga-Lopez A. Birth weight, growth indices, and seminal parameters in male offspring are resilient features to maternal pre-conceptional dietary manipulation in sheep. Domest Anim Endocrinol 2024; 88:106849. [PMID: 38608396 PMCID: PMC11156533 DOI: 10.1016/j.domaniend.2024.106849] [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: 10/24/2023] [Revised: 02/17/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024]
Abstract
Gestational diet manipulation can lead to inadequate fetal nutrient supply resulting in low birth weight, limited postnatal growth, and consequently, reduced reproductive performance in the progeny. However, effects of short-term maternal pre-conceptional dietary manipulation on postnatal growth and reproductive parameters of male offspring in large animals remains unexplored. To determine these consequences, female crossbred (Polypay x Dorset) sheep were allocated to three groups (n = 33/group) of dietary manipulation for 21 days prior to mating under the following conditions: (1) control at 100 % of maintenance energy requirements (40 Kcal of metabolizable energy/kg body weight [BW]), (2) undernutrition (UN) at 50 % of Control intake, and (3) overnutrition (ON) at 200 % of maintenance energy. Singleton ram lambs (UN:9; C:12; ON:6) were monitored from birth until 8 months of age, including birth weight, weekly weights, weight gain, body mass index (BMI), and circulating testosterone. After weaning, monthly scrotal circumference and subcutaneous fat depth were measured. Semen morphology and motility were evaluated at 7 and 8 months of age. Birth weight, weight gain, and BMI at birth and weaning were not significantly different among nutritional treatments. None of the pre-conceptional diets affected body weight change from weaning until 36 weeks of age, BMI, fat depth, or scrotal circumference across the experiment. A sustained rise in plasma testosterone concentrations was detected when ram lambs were, on average, 82 days old and 37 kg. Both testosterone concentrations and scrotal circumference were positively correlated to body weight regardless of treatment group. In addition, seminal parameters did not differ among treatments, but a transient increase in plasma testosterone at 18 weeks of age was observed in ON ram lambs compared to control rams. In conclusion, birth weight, growth indices, and seminal parameters in singleton rams are resilient features in the progeny upon maternal pre-conceptional dietary manipulation in sheep.
Collapse
Affiliation(s)
- Cesar A Rosales-Nieto
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA; Facultad de Agronomía y Veterinaria, Universidad Autónoma de San Luis Potosí, San Luis Potosí, 78321, México
| | - Bradford W Daigneault
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA; Department of Animal Sciences, University of Florida, Gainesville, FL, 32611, USA
| | - Jennifer N Roberts
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48824, USA
| | - Rodrigo Sánchez-López
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Barbara Makela
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Yong Pu
- Department of Pathology, University of Illinois at Chicago, IL, 60612, USA
| | - Richard Ehrhardt
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA; Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48824, USA
| | - João H Jabur Bittar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Almudena Veiga-Lopez
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA; Department of Pathology, University of Illinois at Chicago, IL, 60612, USA.
| |
Collapse
|
14
|
Di Berardino C, Barceviciute U, Camerano Spelta Rapini C, Peserico A, Capacchietti G, Bernabò N, Russo V, Gatta V, Konstantinidou F, Donato M, Barboni B. High-fat diet-negative impact on female fertility: from mechanisms to protective actions of antioxidant matrices. Front Nutr 2024; 11:1415455. [PMID: 38915855 PMCID: PMC11194403 DOI: 10.3389/fnut.2024.1415455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/27/2024] [Indexed: 06/26/2024] Open
Abstract
Introduction Excessive calorie intake poses a significant threat to female fertility, leading to hormonal imbalances and reproductive challenges. Overconsumption of unhealthy fats exacerbates ovarian dysfunction, with an overproduction of reactive oxygen species causing oxidative stress, impairing ovarian follicle development and leading to irregular ovulation and premature ovarian failure. Interest in biological matrices with high antioxidant properties to combat diet-related oxidative stress has grown, as they contain various bioactive factors crucial for neutralizing free radicals potentially preventing female reproductive health. This systematic review evaluates the female reproductive impact of biological matrices in mitigating oxidative damages induced by over calory habits and, in particular, high fat diets. Methods A comparative approach among mammalian models was utilized to interpret literature available data. This approach specifically investigates the antioxidant mechanisms of biological matrices on early and late ovarian folliculogenesis, under physiological and hormone-induced female reproductive cycle. Adhering to the PRISMA 2020 guidelines, only English-language publications from peer-reviewed international indexes were considered. Results The analysis of 121 publications meeting the inclusion criteria facilitated the identification of crucial components of biological matrices. These components, including carbocyclic sugars, phytonutrients, organosulfur compounds, and vitamins, were evaluated for their impact on ovarian follicle resilience, oocyte quality, and reproductive lifespan. The detrimental effects of oxidative stress on female fertility, particularly exacerbated by high saturated fat diets, are well-documented. In vivo studies across mammalian preclinical models have underscored the potential of antioxidants derived from biological matrices to mitigate diet-induced conditions. These antioxidants enhance steroidogenesis and ovarian follicle development, thereby improving oocyte quality. Additionally, discussions within these publications emphasized the clinical significance of these biological matrices, translating research findings into practical applications for female health. Conclusion Further research is essential to fully exploit the potential of these matrices in enhancing female reproduction and mitigating the effects of diets rich in fatty acids. This requires intensified in vitro studies and comprehensive collection of in vivo data before clinical trials. The promotion of ovarian resilience offers promising avenues for enhancing understanding and advancing female reproductive health world-wide.
Collapse
Affiliation(s)
- Chiara Di Berardino
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Urte Barceviciute
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | | | - Alessia Peserico
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Giulia Capacchietti
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Nicola Bernabò
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Rome, Italy
| | - Valentina Russo
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Fani Konstantinidou
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Marisa Donato
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Barbara Barboni
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
15
|
Abdoon ASS, Hegazy AM, Abdel-Azeem AS, Al-Atrash AM, Mohammed DM. The protective effects of some herbs on mitigating HFD-induced obesity via enhancing biochemical indicators and fertility in female rats. Heliyon 2024; 10:e30249. [PMID: 38726161 PMCID: PMC11078881 DOI: 10.1016/j.heliyon.2024.e30249] [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: 01/04/2024] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
The potential of plant-based diets and drugs to prevent and control obesity has been attributed to the presence of several biologically active phytochemicals. The study aimed to assess herb consumption's impact on alleviating the risks and hazards associated with obesity induced by a high-fat diet (HFD) and the promotion of fertility. Eighty rats were allocated into four distinct groups. Group 1 (G1) was provided with a basal diet and acted as the control group. Group 2 (G2) was provided with an HFD. Group 3 (G3) was provided with HFD supplemented with chia seeds and Hibiscus sabdariffa L. The fourth group of subjects was provided with HFD supplemented with Foeniculum vulgare (fennel) and Coriandrum sativum L. (coriander). The feeding session was sustained for 10 weeks, and the biochemical parameters were evaluated. The administration of Foeniculum vulgare (fennel) and Coriandrum sativum L. (coriander) (G4) resulted in a more significant reduction in all biochemical parameters compared to G3, which received a diet consisting of chia seeds and Hibiscus sabdariffa L. Additionally, the average number of embryonic lobes and the average number of offspring after birth were found to be considerably more significant in the normal control group (G1) and group (G4) compared to the HFD group (G2) and group (G3) (P < 0.01). Group 4 (G4) was administered a diet enriched with Foeniculum vulgare (fennel) and Coriandrum sativum L. (coriander), which demonstrated superior outcomes in many biochemical indicators and the promotion of fertility in obese female rats.
Collapse
Affiliation(s)
- Ahmed Sabry S. Abdoon
- Animal Reproduction, Veterinary Research division, National Research Centre, Dokki, Giza, Egypt
| | - Amany M. Hegazy
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, Egypt
| | - Amal S. Abdel-Azeem
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, Egypt
| | - Ahmed M. Al-Atrash
- Medical Administration, Nuclear Materials Authority, Kattmya, Cairo, Egypt
| | - Dina Mostafa Mohammed
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
16
|
González-Alvarez ME, Inyang I, Keating AF. Exposure to 7,12-dimethylbenz[a]anthracene impacts ovarian DNA damage sensing and repair proteins differently in lean and obese female mice and weight loss may mitigate obesity-induced ovarian dysfunction. Toxicol Appl Pharmacol 2024; 486:116930. [PMID: 38626870 DOI: 10.1016/j.taap.2024.116930] [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: 02/29/2024] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 05/21/2024]
Abstract
Obesity impairs oocyte quality, fertility, pregnancy maintenance, and is associated with offspring birth defects. The model ovotoxicant, 7,12-dimethylbenz[a]anthracene (DMBA), causes ovarian DNA damage and follicle loss. Both DMBA-induced chemical biotransformation and the DNA damage response are partially attenuated in obese relative to lean female mice but whether weight loss could improve the DNA damage response to DMBA exposure has not been explored. Thus, at six weeks of age, C57BL/6 J female mice were divided in three groups: 1) Lean (L; n = 20) fed a chow diet for 12 weeks, 2) obese (O; n = 20) fed a high fat high sugar (HFHS) diet for 12 weeks and, 3) slim-down (S; n = 20). The S group was fed with HFHS diet for 7 weeks until attaining a higher body relative to L mice on week 7.5 and switched to a chow diet for 5 weeks to achieve weight loss. Mice then received either corn oil (CT) or DMBA (D; 1 mg/kg) for 7 d via intraperitoneal injection (n = 10/treatment). Obesity increased (P < 0.05) kidney and spleen weight, and DMBA decreased uterine weight (P < 0.05). Ovarian weight was reduced (P < 0.05) in S mice, but DMBA exposure increased ovary weight in the S mice. LC-MS/MS identified 18, 64, and 7 ovarian proteins as altered (P < 0.05) by DMBA in the L, S and O groups, respectively. In S and O mice, 24 and 8 proteins differed, respectively, from L mice. These findings support weight loss as a strategy to modulate the ovarian genotoxicant response.
Collapse
Affiliation(s)
| | - Imaobong Inyang
- Department of Animal Science, Iowa State University, United States of America
| | - Aileen F Keating
- Department of Animal Science, Iowa State University, United States of America.
| |
Collapse
|
17
|
Adamowski M, Sharma Y, Molcan T, Wołodko K, Kelsey G, Galvão AM. Leptin signalling regulates transcriptional differences in granulosa cells from genetically obese mice but not the activation of NLRP3 inflammasome. Sci Rep 2024; 14:8070. [PMID: 38580672 PMCID: PMC10997671 DOI: 10.1038/s41598-024-58181-w] [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: 09/04/2023] [Accepted: 03/26/2024] [Indexed: 04/07/2024] Open
Abstract
Obesity is associated with increased ovarian inflammation and the establishment of leptin resistance. We presently investigated the role of impaired leptin signalling on transcriptional regulation in granulosa cells (GCs) collected from genetically obese mice. Furthermore, we characterised the association between ovarian leptin signalling, the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome and macrophage infiltration in obese mice. After phenotype characterisation, ovaries were collected from distinct group of animals for protein and mRNA expression analysis: (i) mice subjected to a diet-induced obesity (DIO) protocol, where one group was fed a high-fat diet (HFD) and another a standard chow diet (CD) for durations of 4 or 16 weeks; (ii) mice genetically deficient in the long isoform of the leptin receptor (ObRb; db/db); (iii) mice genetically deficient in leptin (ob/ob); and (iv) mice rendered pharmacologically hyperleptinemic (LEPT). Next, GCs from antral follicles isolated from db/db and ob/ob mice were subjected to transcriptome analysis. Transcriptional analysis revealed opposing profiles in genes associated with steroidogenesis and prostaglandin action between the genetic models, despite the similarities in body weight. Furthermore, we observed no changes in the mRNA and protein levels of NLRP3 inflammasome components in the ovaries of db/db mice or in markers of M1 and M2 macrophage infiltration. This contrasted with the downregulation of NLRP3 inflammasome components and M1 markers in ob/ob and 16-wk HFD-fed mice. We concluded that leptin signalling regulates NLRP3 inflammasome activation and the expression of M1 markers in the ovaries of obese mice in an ObRb-dependent and ObRb-independent manner. Furthermore, we found no changes in the expression of leptin signalling and NLRP3 inflammasome genes in GCs from db/db and ob/ob mice, which was associated with no effects on macrophage infiltration genes, despite the dysregulation of genes associated with steroidogenesis in homozygous obese db/db. Our results suggest that: (i) the crosstalk between leptin signalling, NLRP3 inflammasome and macrophage infiltration takes place in ovarian components other than the GC compartment; and (ii) transcriptional changes in GCs from homozygous obese ob/ob mice suggest structural rearrangement and organisation, whereas in db/db mice the impairment in steroidogenesis and secretory activity.
Collapse
Affiliation(s)
- Marek Adamowski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Yashaswi Sharma
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Tomasz Molcan
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Karolina Wołodko
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
| | - António M Galvão
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland.
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
- Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
18
|
Ma Y, Zhang W, Gao M, Li J, Wang Q, Chen M, Gu L. Combined analysis of temporal metabolomics and transcriptomics reveals the metabolic patterns in goat oocytes during maturation. Theriogenology 2024; 218:69-78. [PMID: 38301509 DOI: 10.1016/j.theriogenology.2024.01.036] [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: 11/06/2023] [Revised: 12/26/2023] [Accepted: 01/25/2024] [Indexed: 02/03/2024]
Abstract
Well-balanced and orderly metabolism is a crucial prerequisite for promoting oogenesis. Involvement of single metabolites in oocyte development has been widely reported; however, the comprehensive metabolic framework controlling oocyte maturation is still lacking. In the present study, we employed an integrated temporal metabolomic and transcriptomic method to analyze metabolism in goat oocytes at GV, GVBD, and MII stages (GV, fully-grown immature oocyte; GVBD, stage of meiotic resumption; MII, mature oocyte) during in vitro maturation, revealing the global picture of the metabolic patterns during maturation. In particular, several significantly altered metabolic pathways during goat oocyte meiosis have been identified, including active serine metabolism, increased utilization of tryptophan, and marked accumulation of purine nucleotide. In summary, the current study provides transcriptomic and metabolomic datasets for goat oocyte development that can be applied in cross-species comparative studies.
Collapse
Affiliation(s)
- Yixin Ma
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, China
| | - Wei Zhang
- College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Ming Gao
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, China
| | - Jiashuo Li
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Minjian Chen
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Ling Gu
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
19
|
Fabozzi G, Cimadomo D, Maggiulli R, Vaiarelli A, Badajoz V, Aura M, Canosa S, Bongioanni F, Benini F, Livi C, Zacà C, Borini A, Alviggi E, Iussig B, Hebles M, Sànchez P, Cimadomo V, Rienzi L, Llàcer J. Association between oocyte donors' or recipients' body mass index and clinical outcomes after first single blastocyst transfers-the uterus is the most affected. Fertil Steril 2024; 121:281-290. [PMID: 37549838 DOI: 10.1016/j.fertnstert.2023.07.029] [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: 02/28/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/09/2023]
Abstract
OBJECTIVE To assess whether high body mass index (BMI) in either oocyte donors or recipients is associated with poorer outcomes after the first single blastocyst transfer. DESIGN Retrospective study including 1,394 first blastocyst single embryo transfers (SETs) conducted by 1,394 recipients during oocyte donation cycles with the gametes retrieved from 1,394 women (January 2019-July 2021). Four BMI clusters were defined for both donors and recipients (underweight: <18.5 kg; normal weight: 18.5-24.9 kg; overweight: 25-29.9 kg; and obese: ≥30 kg). SETTING Network of private IVF centers. PATIENTS A total of 1,394 recipients aged 42.4 ± 4.0 and with a BMI of 23.2 ± 3.8 kg/m2, and 1,394 donors aged 26.1 ± 4.2 and with a BMI of 21.9 ± 2.5 kg/m2. INTERVENTION All oocytes were vitrified at 2 egg banks and warmed at 8 in vitro fertilization clinics that were part of the same network. Intracytoplasmic sperm injection, blastocyst culture, and either fresh or vitrified-warmed SETs were conducted. Putative confounders were investigated, and the data were adjusted through regression analyses. MAIN OUTCOME MEASURES The primary outcome was the live birth rate (LBR) per SET according to donors' and/or recipients' BMI. The main secondary outcome was the miscarriage rate (<22 gestational weeks) per clinical pregnancy. RESULTS The LBR per blastocyst SET showed no significant association with donors' BMI. Regarding recipients' BMI, instead, the multivariate odds ratio was significant in obese vs. normal-weight recipients (0.58, 95% confidence interval, 0.37-0.91). The miscarriage rate per clinical pregnancy was also significantly associated with recipients' obesity, with a multivariate odds ratio of 2.31 (95% confidence interval, 1.18-4.51) vs. normal-weight patients. A generalized additive model method was used to represent the relationship between predicted LBR or miscarriage rates and donors' or recipients' BMI; it pictured a scenario where the former outcome moderately but continuously decreases with increasing recipients' BMI to then sharply decline in the BMI range of 25-35 kg/m2. The miscarriage rate, instead, increases almost linearly with respect to both donors' and recipients' increasing BMI. CONCLUSION Obesity mostly affects the uterus, especially because of higher miscarriage rates. Yet, poorer outcomes can be appreciated already with a BMI of 25 kg/m2 in both oocyte donors and recipients. Finer markers of nutritional homeostasis are therefore desirable; recipients should be counseled about poorer expected outcomes in cases of overweight and obesity; and oocyte banks should avoid assigning oocytes from overweight donors to overweight and obese recipients.
Collapse
Affiliation(s)
- Gemma Fabozzi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy; IVIRMA Global Research Alliance, B-WOMAN, Rome, Italy; Department of Biomedicine and Prevention, University of Rome, Tor Vergata, Rome, Italy
| | - Danilo Cimadomo
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy.
| | - Roberta Maggiulli
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | - Alberto Vaiarelli
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy
| | | | - Monica Aura
- IVIRMA Global Research Alliance, GINEFIV, Barcelona, Spain
| | | | | | | | - Claudia Livi
- IVIRMA Global Research Alliance, DEMETRA, Florence, Italy
| | - Carlotta Zacà
- IVIRMA Global Research Alliance, 9.baby, Bologna, Italy
| | - Andrea Borini
- IVIRMA Global Research Alliance, 9.baby, Bologna, Italy
| | - Erminia Alviggi
- IVIRMA Global Research Alliance, GENERA, Clinica Ruesch, Naples, Italy
| | - Benedetta Iussig
- IVIRMA Global Research Alliance, GENERA, GENERA Veneto, Marostica, Italy
| | - Maria Hebles
- IVIRMA Global Research Alliance, GINEMED, Sevilla, Spain
| | | | | | - Laura Rienzi
- IVIRMA Global Research Alliance, GENERA, Clinica Valle Giulia, Rome, Italy; Department of Biomolecular Sciences, the University of Urbino "Carlo Bo", Urbino, Italy
| | | |
Collapse
|
20
|
Bakkensen JB, Strom D, Boots CE. Frozen embryo transfer outcomes decline with increasing female body mass index in female but not male factor infertility: analysis of 56,564 euploid blastocyst transfers. Fertil Steril 2024; 121:271-280. [PMID: 37549839 DOI: 10.1016/j.fertnstert.2023.07.027] [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: 01/20/2023] [Revised: 04/11/2023] [Accepted: 07/28/2023] [Indexed: 08/09/2023]
Abstract
OBJECTIVE To evaluate the association of body mass index (BMI) with cycle outcomes after euploid frozen blastocyst transfer. DESIGN Retrospective cohort study. SETTING Not applicable. PATIENT(S) A total of 56,564 first single autologous euploid frozen embryo transfers from the 2016-2019 Society for Assisted Reproductive Technology database were analyzed using BMI and using World Health Organization BMI cohorts. Subanalyses were performed on cycles among patients with a sole diagnosis of polycystic ovary syndrome (PCOS) (n = 4,626) and among patients with only a male factor (n = 10,854). INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Clinical pregnancy, pregnancy loss, and live birth (LB). RESULT(S) Success rates and adjusted odds ratios (aORs) with 95% confidence intervals (CIs) for all outcomes were most favorable among those with normal BMI and progressively worsened with increasing BMI. These trends persisted among patients with PCOS for clinical pregnancy (aOR, 0.99; 95% CI, 0.98-0.997), pregnancy loss (aOR, 1.02; 95% CI, 1.01-1.04), and LB (aOR, 0.98; 95% CI, 0.97-0.99), but not among patients with a male factor only for clinical pregnancy (aOR, 1.00; 95% CI, 0.99-1.01), pregnancy loss (aOR, 1.01; 95% CI, 0.99-1.03), or LB (aOR, 0.99; 95% CI, 0.98-1.00). CONCLUSION(S) In the largest cohort to date, increasing BMI was associated with decreased pregnancy and LB and increased pregnancy loss after euploid frozen embryo transfers among the entire cohort and among patients with a sole diagnosis of PCOS; however, these results were attenuated among patients with a sole diagnosis of male factor infertility, suggesting that associated female infertility diagnoses and not BMI alone may underlie this trend.
Collapse
Affiliation(s)
- Jennifer B Bakkensen
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Danielle Strom
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Christina E Boots
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
21
|
Inoue Y, Fukushima M, Hirasawa G, Furukawa F, Takeda H, Umatani C. Maternal High-Fat Diet Affects the Contents of Eggs and Causes Abnormal Development in the Medaka Fish. Endocrinology 2024; 165:bqae006. [PMID: 38279936 DOI: 10.1210/endocr/bqae006] [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: 10/17/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 01/29/2024]
Abstract
Maternal nutritional status can affect development and metabolic phenotypes of progeny in animals. The effects of maternal diet are thought to be mediated mainly by changes inside oocytes such as organelles, maternal RNAs, and metabolites. However, to what extent each factor contributes to offspring phenotypes remains uncertain, especially in viviparous mammalian systems, where factors other than oocytes, such as placenta and milk, need to be considered. Here, using the medaka fish as an oviparous vertebrate model, we examined whether maternal high-fat diet (mHFD) feeding affects offspring development and what kind of changes occur in the contents of mature eggs. We found that mHFD caused the high frequency of embryonic deformities of offspring, accompanied by downregulation of transcription- and translation-related genes and zygotic transcripts at the blastula stage. Transcriptomic and metabolomic analyses of mature eggs suggested decreased catabolism of amino acids and glycogen, moderate upregulation of endoplasmic reticulum stress-related genes, and elevated lipid levels in mHFD eggs. Furthermore, high-fat diet females showed a higher incidence of oocyte atresia and downregulation of egg protein genes in the liver. These data suggest that attenuated amino acid catabolism triggered by decreased yolk protein load/processing, as well as elevated lipid levels inside eggs, are the prime candidates that account for the higher incidence of embryonic deformities in mHFD offspring. Our study presents a comprehensive data on the changes inside eggs in a mHFD model of nonmammalian vertebrates and provides insights into the mechanisms of parental nutritional effects on offspring.
Collapse
Affiliation(s)
- Yusuke Inoue
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Manatsu Fukushima
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Go Hirasawa
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo 113-8657, Japan
| | - Fumiya Furukawa
- School of Marine Biosciences, Kitasato University, Kanagawa 252-0373, Japan
| | - Hiroyuki Takeda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Chie Umatani
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Division of Applied Biological Chemistry, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| |
Collapse
|
22
|
Meulders B, Marei WFA, Xhonneux I, Bols PEJ, Leroy JLMR. Effect of lipotoxicity on mitochondrial function and epigenetic programming during bovine in vitro embryo production. Sci Rep 2023; 13:21664. [PMID: 38066095 PMCID: PMC10709407 DOI: 10.1038/s41598-023-49184-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
Maternal metabolic disorders may cause lipotoxic effects on the developing oocyte. Understanding the timing at which this might disrupt embryo epigenetic programming and how this is linked with mitochondrial dysfunction is crucial for improving assisted reproductive treatments, but has not been investigated before. Therefore, we used a bovine in vitro model to investigate if pathophysiological palmitic acid (PA) concentrations during in vitro oocyte maturation and in vitro embryo culture alter embryo epigenetic patterns (DNA methylation (5mC) and histone acetylation/methylation (H3K9ac/H3K9me2)) compared to control (CONT) and solvent control (SCONT), at the zygote and morula stage. Secondly, we investigated if these epigenetic alterations are associated with mitochondrial dysfunction and changes in ATP production rate, or altered expression of epigenetic regulatory genes. Compared to SCONT, H3K9ac and H3K9me2 levels were increased in PA-derived zygotes. Also, 5mC and H3K9me2 levels were increased in PA-exposed morulae compared to SCONT. This was associated with complete inhibition of glycolytic ATP production in oocytes, increased mitochondrial membrane potential and complete inhibition of glycolytic ATP production in 4-cell embryos and reduced SOD2 expression in PA-exposed zygotes and morulae. For the first time, epigenetic alterations in metabolically compromised zygotes and morulae have been observed in parallel with mitochondrial dysfunction in the same study.
Collapse
Affiliation(s)
- Ben Meulders
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium.
| | - Waleed F A Marei
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Inne Xhonneux
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Peter E J Bols
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Jo L M R Leroy
- Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
23
|
Zhao H, Wong RJ, Stevenson DK. The placental vasculature is affected by changes in gene expression and glycogen-rich cells in a diet-induced obesity mouse model. PLoS One 2023; 18:e0294185. [PMID: 37948457 PMCID: PMC10637699 DOI: 10.1371/journal.pone.0294185] [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: 07/22/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Maternal obesity is a risk factor for pregnancy complications. Obesity caused by a high-fat diet (HFD) may alter maternal glucose/glycogen metabolism. Here, our objective was to investigate whether the placental vasculature is altered via changes in gene expression and glycogen-rich cells using a preclinical mouse model of diet-induced obesity. We subjected female FVB/N mice to one of three feeding regimens: regular chow (RC) given at preconception and during pregnancy (Control); RC given at preconception and then a HFD during pregnancy (HFD-P); or HFD initiated 4 weeks preconception and during pregnancy (HFD-PreCP). Daily food consumption and weekly maternal weights were recorded. Maternal blood glucose levels were measured at preconception and 4 gestational epochs (E6.5-E9.5, E10.5-E12.5, E13.5-E15.5, E16.5-E19.5). At E8.5-E16.5, total RNA in placentas were isolated for gene expression analyses. Placentas were also collected for HE and periodic acid Schiff's (PAS) staining and glycogen content assays. Dams in the HFD-P and HFD-PreCP groups gained significantly more weight than controls. Pre- and antenatal glucose levels were also significantly higher (15%-30%) in HFD-PreCP dams. Expression of several placental genes were also altered in HFD dams compared with controls. Consumption of the HFD also led to phenotypic and morphologic changes in glycogen trophoblasts (GlyTs) and uterine natural killer (uNK) cells. Alterations in vascularity were also observed in the labyrinth of HFD-PreCP placentas, which correlated with decreased placental efficiency. Overall, we observed that a HFD induces gestational obesity in mice, alters expression of placental genes, affects glucose homeostasis, and alters glycogen-positive GlyTs and uNK cells. All these changes may lead to impaired placental vascular development, and thus heighten the risk for pregnancy complications.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Ronald J. Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - David K. Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| |
Collapse
|
24
|
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: 0.5] [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
|
25
|
Paula VG, Sinzato YK, Gallego FQ, Cruz LL, Aquino AMD, Scarano WR, Corrente JE, Volpato GT, Damasceno DC. Intergenerational Hyperglycemia Impairs Mitochondrial Function and Follicular Development and Causes Oxidative Stress in Rat Ovaries Independent of the Consumption of a High-Fat Diet. Nutrients 2023; 15:4407. [PMID: 37892483 PMCID: PMC10609718 DOI: 10.3390/nu15204407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
We analyzed the influence of maternal hyperglycemia and the post-weaning consumption of a high-fat diet on the mitochondrial function and ovarian development of the adult pups of diabetic rats. Female rats received citrate buffer (Control-C) or Streptozotocin (for diabetes induction-D) on postnatal day 5. These adult rats were mated to obtain female pups (O) from control dams (OC) or from diabetic dams (OD), and they received a standard diet (SD) or high-fat diet (HFD) from weaning to adulthood and were distributed into OC/SD, OC/HFD, OD/SD, and OD/HFD. In adulthood, the OGTT and AUC were performed. These rats were anesthetized and euthanized for sample collection. A high percentage of diabetic rats were found to be in the OD/HFD group (OD/HFD 40% vs. OC/SD 0% p < 0.05). Progesterone concentrations were lower in the experimental groups (OC/HFD 0.40 ± 0.04; OD/SD 0.30 ± 0.03; OD/HFD 0.24 ± 0.04 vs. OC/SD 0.45 ± 0.03 p < 0.0001). There was a lower expression of MFF (OD/SD 0.34 ± 0.33; OD/HFD 0.29 ± 0.2 vs. OC/SD 1.0 ± 0.41 p = 0.0015) and MFN2 in the OD/SD and OD/HFD groups (OD/SD 0.41 ± 0.21; OD/HFD 0.77 ± 0.18 vs. OC/SD 1.0 ± 0.45 p = 0.0037). The number of follicles was lower in the OD/SD and OD/HFD groups. A lower staining intensity for SOD and Catalase and higher staining intensity for MDA were found in ovarian cells in the OC/HFD, OD/SD, and OD/HFD groups. Fetal programming was responsible for mitochondrial dysfunction, ovarian reserve loss, and oxidative stress; the association of maternal diabetes with an HFD was responsible for the higher occurrence of diabetes in female adult pups.
Collapse
Affiliation(s)
- Verônyca Gonçalves Paula
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Yuri Karen Sinzato
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Franciane Quintanilha Gallego
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Larissa Lopes Cruz
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Ariana Musa de Aquino
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| | - Wellerson Rodrigo Scarano
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| | - José Eduardo Corrente
- Research Support Office, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças 78600-000, MG, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| |
Collapse
|
26
|
Giedt MS, Thomalla JM, White RP, Johnson MR, Lai ZW, Tootle TL, Welte MA. Adipose triglyceride lipase promotes prostaglandin-dependent actin remodeling by regulating substrate release from lipid droplets. Development 2023; 150:dev201516. [PMID: 37306387 PMCID: PMC10281261 DOI: 10.1242/dev.201516] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
Lipid droplets (LDs), crucial regulators of lipid metabolism, accumulate during oocyte development. However, their roles in fertility remain largely unknown. During Drosophila oogenesis, LD accumulation coincides with the actin remodeling necessary for follicle development. Loss of the LD-associated Adipose Triglyceride Lipase (ATGL) disrupts both actin bundle formation and cortical actin integrity, an unusual phenotype also seen when the prostaglandin (PG) synthase Pxt is missing. Dominant genetic interactions and PG treatment of follicles indicate that ATGL acts upstream of Pxt to regulate actin remodeling. Our data suggest that ATGL releases arachidonic acid (AA) from LDs to serve as the substrate for PG synthesis. Lipidomic analysis detects AA-containing triglycerides in ovaries, and these are increased when ATGL is lost. High levels of exogenous AA block follicle development; this is enhanced by impairing LD formation and suppressed by reducing ATGL. Together, these data support the model that AA stored in LD triglycerides is released by ATGL to drive the production of PGs, which promote the actin remodeling necessary for follicle development. We speculate that this pathway is conserved across organisms to regulate oocyte development and promote fertility.
Collapse
Affiliation(s)
- Michelle S. Giedt
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | - Roger P. White
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Matthew R. Johnson
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Zon Weng Lai
- Harvard T.H. Chan Advanced Multi-omics Platform, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tina L. Tootle
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Michael A. Welte
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
27
|
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; 1869:166802. [PMID: 37414229 DOI: 10.1016/j.bbadis.2023.166802] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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 the oocytes of obese mothers 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", México.
| | | | - A N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, UK.
| |
Collapse
|
28
|
Muhammad T, Wan Y, Lv Y, Li H, Naushad W, Chan WY, Lu G, Chen ZJ, Liu H. Maternal obesity: A potential disruptor of female fertility and current interventions to reduce associated risks. Obes Rev 2023; 24:e13603. [PMID: 37452501 DOI: 10.1111/obr.13603] [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: 10/27/2022] [Revised: 05/17/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
Currently, obesity has achieved epidemic levels in reproductive-aged women with a myriad of consequences. Obesity is susceptible to several reproductive complications that eventually affect fertility rates. These complications originate from the deteriorated quality of oocytes from mothers with obesity, which increases the probability of chromosomal aneuploidy, elevated reactive oxygen species production, compromised embryonic developmental competency, and eventually reduced fertility. Maternal obesity is linked to pregnancy complications such as implantation error, abortion, miscarriage, and early pregnancy loss. This review highlights the adverse effects of maternal obesity on female fertility, with a focus on the mechanistic link between maternal obesity and oocyte quality and discusses possible measures to reduce its associated risks.
Collapse
Affiliation(s)
- Tahir Muhammad
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Pakistan
| | - Yanling Wan
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
| | - Yue Lv
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Hanzhen Li
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
| | - Wasifa Naushad
- Department of Pathology, Microbiology and Immunology, New York Medical College, 15 Dana Road, Valhalla, NY, 10595, USA
| | - Wai-Yee Chan
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Gang Lu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, 999077, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200000, China
| | - Hongbin Liu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, China
- Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Jinan, Shandong, 250012, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jinan, Shandong, 250012, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, 999077, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, China
| |
Collapse
|
29
|
Wang Y, Wang C, Hong W, Tian S, Lu Q, Wang BJ, Jin H, Zhuoma N, Lu X, Zhao H. Genomic analysis of Tibetan ground tits identifies molecular adaptations associated with cooperative breeding. Curr Zool 2023; 69:620-630. [PMID: 37637321 PMCID: PMC10449426 DOI: 10.1093/cz/zoac067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/16/2022] [Indexed: 08/29/2023] Open
Abstract
Cooperative breeding is a sophisticated altruistic social behavior that helps social animals to adapt to harsh environments. The Tibetan ground tit, Pseudopodoces humilis, is a high-altitude bird endemic to the Tibetan plateau. Recently, it has become an exciting system for studying the evolution of facultative cooperative breeding. To test for molecular adaptations associated with cooperative breeding, we resequenced the whole genome of ground tits from 6 wild populations that display remarkable variation in the frequency of cooperative breeding. Population structure analyses showed that the 6 populations were divided into 4 lineages, which is congruent with the major geographical distribution of the sampling sites. Using genome-wide selective sweep analysis, we identified putative positively selected genes (PSGs) in groups of tits that displayed high and low cooperative breeding rates. The total number of PSGs varied from 146 to 722 in high cooperative breeding rate populations, and from 272 to 752 in low cooperative breeding rate populations. Functional enrichment analysis of these PSGs identified several significantly enriched ontologies related to oxytocin signaling, estrogen signaling, and insulin secretion. PSGs involved in these functional ontologies suggest that molecular adaptations in hormonal regulation may have played important roles in shaping the evolution of cooperative breeding in the ground tit. Taken together, our study provides candidate genes and functional ontologies involved in molecular adaptations associated with cooperative breeding in Tibetan ground tits, and calls for a better understanding of the genetic roles in the evolution of cooperative breeding.
Collapse
Affiliation(s)
- Yi Wang
- Department of Ecology, Tibetan Centre for Ecology and Conservation at Wuhan University—Tibet University, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chen Wang
- Department of Ecology, Tibetan Centre for Ecology and Conservation at Wuhan University—Tibet University, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Wei Hong
- Department of Ecology, Tibetan Centre for Ecology and Conservation at Wuhan University—Tibet University, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Shilin Tian
- Department of Ecology, Tibetan Centre for Ecology and Conservation at Wuhan University—Tibet University, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Qin Lu
- Department of Ecology, Tibetan Centre for Ecology and Conservation at Wuhan University—Tibet University, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Bing-Jun Wang
- Department of Ecology, Tibetan Centre for Ecology and Conservation at Wuhan University—Tibet University, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Huaiming Jin
- Research Center for Ecology, College of Science, Tibet University, Lhasa 850000, China
| | - Nima Zhuoma
- Research Center for Ecology, College of Science, Tibet University, Lhasa 850000, China
| | - Xin Lu
- Department of Ecology, Tibetan Centre for Ecology and Conservation at Wuhan University—Tibet University, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Huabin Zhao
- Department of Ecology, Tibetan Centre for Ecology and Conservation at Wuhan University—Tibet University, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
- Research Center for Ecology, College of Science, Tibet University, Lhasa 850000, China
| |
Collapse
|
30
|
Nguyen LT, Pollock CA, Saad S. Nutrition and Developmental Origins of Kidney Disease. Nutrients 2023; 15:4207. [PMID: 37836490 PMCID: PMC10574202 DOI: 10.3390/nu15194207] [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: 09/13/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
The developmental programming hypothesis proposes that adverse environmental insults during critical developmental periods increase the risk of diseases later in life. The kidneys are deemed susceptible to such a process, although the exact mechanisms remain elusive. Many factors have been reported to contribute to the developmental origin of chronic kidney diseases (CKD), among which peri-gestational nutrition has a central role, affecting kidney development and metabolism. Physiologically, the link between malnutrition, reduced glomerular numbers, and increased blood pressure is key in the developmental programming of CKD. However, recent studies regarding oxidative stress, mitochondrial dysfunction, epigenetic modifications, and metabolic changes have revealed potential novel pathways for therapeutic intervention. This review will discuss the role of imbalanced nutrition in the development of CKD.
Collapse
Affiliation(s)
- Long T. Nguyen
- Renal Research Group, Kolling Institute, St. Leonards, NSW 2065, Australia; (C.A.P.); (S.S.)
| | | | | |
Collapse
|
31
|
González-Alvarez ME, Keating AF. Hepatic and ovarian effects of perfluorooctanoic acid exposure differ in lean and obese adult female mice. Toxicol Appl Pharmacol 2023; 474:116614. [PMID: 37422089 DOI: 10.1016/j.taap.2023.116614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/29/2023] [Accepted: 07/01/2023] [Indexed: 07/10/2023]
Abstract
Obesity and overweight cause poor oocyte quality, miscarriage, infertility, polycystic ovarian syndrome, and offspring birth defects and affects 40% and 20% of US women and girls, respectively. Perfluorooctanoic acid (PFOA), a per- and poly-fluoroalkyl substance (PFAS), is environmentally persistent and has negative female reproductive effects including endocrine disruption, oxidative stress, altered menstrual cyclicity, and decreased fertility in humans and animal models. PFAS exposure is associated with non-alcoholic fatty liver disease which affects ∼24-26% of the US population. This study investigated the hypothesis that PFOA exposure impacts hepatic and ovarian chemical biotransformation and alters the serum metabolome. At 7 weeks of age, female lean, wild type (KK.Cg-a/a) or obese (KK.Cg-Ay/J) mice received saline (C) or PFOA (2.5 mg/Kg) per os for 15 d. Hepatic weight was increased by PFOA exposure in both lean and obese mice (P < 0.05) and obesity also increased liver weight (P < 0.05) compared to lean mice. The serum metabolome was also altered (P < 0.05) by PFOA exposure and differed between lean and obese mice. Exposure to PFOA altered (P < 0.05) the abundance of ovarian proteins with roles in xenobiotic biotransformation (lean - 6; obese - 17), metabolism of fatty acids (lean - 3; obese - 9), cholesterol (lean - 8; obese - 11), amino acids (lean - 18; obese - 19), glucose (lean - 7; obese - 10), apoptosis (lean - 18; obese - 13), and oxidative stress (lean - 3; obese - 2). Use of qRT-PCR determined that exposure to PFOA increased (P < 0.05) hepatic Ces1 and Chst1 in lean but Ephx1 and Gstm3 in obese mice. Also, obesity basally increased (P < 0.05) Nat2, Gpi and Hsd17b2 mRNA levels. These data identify molecular changes resultant from PFOA exposure that may cause liver injury and ovotoxicity in females. In addition, differences in toxicity induced by PFOA exposure occurs in lean and obese mice.
Collapse
Affiliation(s)
- M Estefanía González-Alvarez
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames, IA 50011, United States of America
| | - Aileen F Keating
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames, IA 50011, United States of America.
| |
Collapse
|
32
|
Shelling AN, Ahmed Nasef N. The Role of Lifestyle and Dietary Factors in the Development of Premature Ovarian Insufficiency. Antioxidants (Basel) 2023; 12:1601. [PMID: 37627595 PMCID: PMC10451748 DOI: 10.3390/antiox12081601] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Premature ovarian insufficiency (POI) is a condition that arises from dysfunction or early depletion of the ovarian follicle pool accompanied by an earlier-than-normal loss of fertility in young women. Oxidative stress has been suggested as an important factor in the decline of fertility in women and POI. In this review, we discuss the mechanisms of oxidative stress implicated in ovarian ageing and dysfunction in relation to POI, in particular mitochondrial dysfunction, apoptosis and inflammation. Genetic defects, autoimmunity and chemotherapy, are some of the reviewed hallmarks of POI that can lead to increased oxidative stress. Additionally, we highlight lifestyle factors, including diet, low energy availability and BMI, that can increase the risk of POI. The final section of this review discusses dietary factors associated with POI, including consumption of oily fish, mitochondria nutrient therapy, melatonin, dairy and vitamins that can be targeted as potential interventions, especially for at-risk women and in combination with personalised nutrition. Understanding the impact of lifestyle and its implications for POI and oxidative stress holds great promise in reducing the burden of this condition.
Collapse
Affiliation(s)
- Andrew N. Shelling
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand;
- Centre for Cancer Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
| | - Noha Ahmed Nasef
- Riddet Research Institute, Massey University, Palmerston North 4474, New Zealand
- School of Food and Advanced Technology, College of Science, Massey University, Palmerston North 4474, New Zealand
| |
Collapse
|
33
|
Moorkens K, Leroy JLMR, Quanico J, Baggerman G, Marei WFA. How the Oviduct Lipidomic Profile Changes over Time after the Start of an Obesogenic Diet in an Outbred Mouse Model. BIOLOGY 2023; 12:1016. [PMID: 37508445 PMCID: PMC10376370 DOI: 10.3390/biology12071016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/03/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023]
Abstract
We investigated whether a high-fat/high-sugar (HF/HS) diet alters the lipidomic profile of the oviductal epithelium (OE) and studied the patterns of these changes over time. Female outbred Swiss mice were fed either a control (10% fat) or HF/HS (60% fat, 20% fructose) diet. Mice (n = 3 per treatment per time point) were sacrificed and oviducts were collected at 3 days and 1, 4, 8, 12 and 16 weeks on the diet. Lipids in the OE were imaged using matrix-assisted laser desorption ionisation mass spectrometry imaging. Discriminative m/z values and differentially regulated lipids were determined in the HF/HS versus control OEs at each time point. Feeding the obesogenic diet resulted in acute changes in the lipid profile in the OE already after 3 days, and thus even before the development of an obese phenotype. The changes in the lipid profile of the OE progressively increased and became more persistent after long-term HF/HS diet feeding. Functional annotation revealed a differential abundance of phospholipids, sphingomyelins and lysophospholipids in particular. These alterations appear to be not only caused by the direct accumulation of the excess circulating dietary fat but also a reduction in the de novo synthesis of several lipid classes, due to oxidative stress and endoplasmic reticulum dysfunction. The described diet-induced lipidomic changes suggest alterations in the OE functions and the oviductal microenvironment which may impact crucial reproductive events that take place in the oviduct, such as fertilization and early embryo development.
Collapse
Affiliation(s)
- Kerlijne Moorkens
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Jo L M R Leroy
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Jusal Quanico
- Centre for Proteomics, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Geert Baggerman
- Centre for Proteomics, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
- Health Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400 Mol, Belgium
| | - Waleed F A Marei
- Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| |
Collapse
|
34
|
Ermisch AF, Bidne KL, Kurz SG, Bochantin KA, Wood JR. Ovarian inflammation mediated by Toll-like receptor 4 increased transcripts of maternal effect genes and decreased embryo development†. Biol Reprod 2023; 108:423-436. [PMID: 36461933 DOI: 10.1093/biolre/ioac212] [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: 05/23/2022] [Revised: 11/03/2022] [Accepted: 11/25/2022] [Indexed: 12/07/2022] Open
Abstract
Obese women are subfertile and have reduced assisted reproduction success, which may be due to reduced oocyte competence. We hypothesize that consumption of a high-fat/high-sugar diet induces ovarian inflammation, which is a primary contributor to decreased oocyte quality and pre-implantation embryo development. To test this hypothesis, C57BL/6 (B6) mice with a normal inflammatory response and C3H/HeJ (C3H) mice with a dampened inflammatory response due to dysfunctional Toll-like receptor 4 were fed either normal chow or high-fat/high-sugar diet. In both B6 and C3H females, high-fat/high-sugar diet induced excessive adiposity and hyperglycemia compared to normal chow-fed counterparts. Conversely, ovarian CD68 levels and oocyte expression of oxidative stress markers were increased when collected from B6 high-fat/high-sugar but not C3H high-fat/high-sugar mice. Following in vitro fertilization of in vivo matured oocytes, blastocyst development was decreased in B6-high-fat/high-sugar but not C3H high-fat/high-sugar mice. Expression of cumulus cell markers of oocyte quality were altered in both B6 high-fat/high-sugar and C3H high-fat/high-sugar. However, there were no diet-dependent differences in spindle abnormalities in either B6 or C3H mice, suggesting potential defects in cytoplasmic maturation. Indeed, there were significant increases in the abundance of maternal effect gene mRNAs in oocytes from only B6 high-fat/high-sugar mice. These differentially expressed genes encode proteins of the subcortical maternal complex and associated with mRNA metabolism and epigenetic modifications. These genes regulate maternal mRNA degradation at oocyte maturation, mRNA clearance at the zygotic genome activation, and methylation of imprinted genes suggesting a mechanism by which inflammation induced oxidative stress impairs embryo development.
Collapse
Affiliation(s)
- Alison F Ermisch
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Katie L Bidne
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Scott G Kurz
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Kerri A Bochantin
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jennifer R Wood
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
35
|
Abstract
In this chapter, we highlight examples of the diverse array of developmental, cellular, and biochemical insights that can be gained by using Drosophila melanogaster oogenesis as a model tissue. We begin with an overview of ovary development and adult oogenesis. Then we summarize how the adult Drosophila ovary continues to advance our understanding of stem cells, cell cycle, cell migration, cytoplasmic streaming, nurse cell dumping, and cell death. We also review emerging areas of study, including the roles of lipid droplets, ribosomes, and nuclear actin in egg development. Finally, we conclude by discussing the growing conservation of processes and signaling pathways that regulate oogenesis and female reproduction from flies to humans.
Collapse
|
36
|
A Multi-Ingredient Supplement Protects against Obesity and Infertility in Western Diet-Fed Mice. Nutrients 2023; 15:nu15030611. [PMID: 36771318 PMCID: PMC9921271 DOI: 10.3390/nu15030611] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
The Western diet (WD) predisposes to bodyweight gain and obesity and is linked to mitochondrial dysfunction, oxidative damage, inflammation, and multisystem disease, even affecting the reproductive organs, fertility, and pregnancy outcomes. In this study, we investigated the effects of multi-ingredient supplementation (MIS) with antioxidants, phytonutrients, and vitamins ('Fertility Enhancer'; FE) on white adipose tissue (WAT) expansion, nonalcoholic fatty liver disease (NAFLD), and infertility in WD-fed C57BL/6J mice. Five-month-old male (M) and female (F) mice were fed a low-fat diet (LF) or a high fat/sucrose WD (HF) for six weeks, followed by six weeks of LF (3.64 kcal/g), HF (4.56 kcal/g), or HF combined with FE (4.50 kcal/g). A sub-set of animals were sacrificed at 12 weeks, while the remainder were harem-mated in a 1:2 male-to-female ratio, and singly housed during the gestational period. Two-way, factorial ANOVA analysis revealed a main effect of diet on bodyweight (BW), total body fat, % body fat, white adipose tissue mass, and liver lipid content (all p < 0.001), driven by the anti-obesogenic effects of the 'Fertility Enhancer'. Similarly, a main effect of diet was found on PGC1-α mRNA levels (p < 0.05) and mitochondrial protein content (p < 0.001) in perigonadal WAT, with PGC1-α induction and higher complex II and complex III expression in FE vs. HF animals. Copulatory plug counts were higher in FE vs. HE couples (30% vs. 6%), resulting in more litters (4 vs. 0) and higher copulatory success (67% vs. 0%). Although the trends of all histology outcomes were suggestive of a benefit from the FE diet, only the number of atretic follicles and testicular mass were significant. Ovarian IL-1β mRNA induction was significantly attenuated in the FE group (p < 0.05 vs. HF) with CASP1 attenuation trending lower (p = 0.09 vs. HF), which is indicative of anti-inflammatory benefits of the 'Fertility Enhancer.' We conclude that supplementation with specific phytonutrients, antioxidants, and vitamins may have utility as an adjunctive therapy for weight management, fatty liver disease, and infertility in overweight and obese couples.
Collapse
|
37
|
Gao M, Chen M, Chen Q, Zhu S, Wang H, Yang W, Wang X, Wang Q, Gu L. Integration of parallel metabolomics and transcriptomics reveals metabolic patterns in porcine oocytes during maturation. Front Endocrinol (Lausanne) 2023; 14:1131256. [PMID: 36817597 PMCID: PMC9929430 DOI: 10.3389/fendo.2023.1131256] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Well-controlled metabolism is the prerequisite for optimal oocyte development. To date, numerous studies have focused mainly on the utilization of exogenous substrates by oocytes, whereas the underlying mechanism of intrinsic regulation during meiotic maturation is less characterized. Herein, we performed an integrated analysis of parallel metabolomics and transcriptomics by isolating porcine oocytes at three time points, cooperatively depicting the global picture of the metabolic patterns during maturation. In particular, we identified the novel metabolic features during porcine oocyte meiosis, such as the fall in bile acids, the active one-carbon metabolism and a progressive decline in nucleotide metabolism. Collectively, the current study not only provides a comprehensive multiple omics data resource, but also may facilitate the discovery of molecular biomarkers that could be used to predict and improve oocyte quality.
Collapse
Affiliation(s)
- Ming Gao
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, China
| | - Minjian Chen
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiuzhen Chen
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Hengjie Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Weizheng Yang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Xi Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Ling Gu, ; Qiang Wang, ; Xi Wang,
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Ling Gu, ; Qiang Wang, ; Xi Wang,
| | - Ling Gu
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Ling Gu, ; Qiang Wang, ; Xi Wang,
| |
Collapse
|
38
|
Deluao JC, Winstanley Y, Robker RL, Pacella-Ince L, Gonzalez MB, McPherson NO. OXIDATIVE STRESS AND REPRODUCTIVE FUNCTION: Reactive oxygen species in the mammalian pre-implantation embryo. Reproduction 2022; 164:F95-F108. [PMID: 36111646 DOI: 10.1530/rep-22-0121] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 09/15/2022] [Indexed: 11/08/2022]
Abstract
In brief Reactive oxygen species are generated throughout the pre-implantation period and are necessary for normal embryo formation. However, at pathological levels, they result in reduced embryo viability which can be mediated through factors delivered by sperm and eggs at conception or from the external environment. Abstract Reactive oxygen species (ROS) occur naturally in pre-implantation embryos as a by-product of ATP generation through oxidative phosphorylation and enzymes such as NADPH oxidase and xanthine oxidase. Biological concentrations of ROS are required for crucial embryonic events such as pronuclear formation, first cleavage and cell proliferation. However, high concentrations of ROS are detrimental to embryo development, resulting in embryo arrest, increased DNA damage and modification of gene expression leading to aberrant fetal growth and health. In vivo embryos are protected against oxidative stress by oxygen scavengers present in follicular and oviductal fluids, while in vitro, embryos rely on their own antioxidant defence mechanisms to protect against oxidative damage, including superoxide dismutase, catalase, glutathione and glutamylcysteine synthestase. Pre-implantation embryonic ROS originate from eggs, sperm and embryos themselves or from the external environment (i.e. in vitro culture system, obesity and ageing). This review examines the biological and pathological roles of ROS in the pre-implantation embryo, maternal and paternal origins of embryonic ROS, and from a clinical perspective, we comment on the growing interest in combating increased oxidative damage in the pre-implantation embryo through the addition of antioxidants.
Collapse
Affiliation(s)
- Joshua C Deluao
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, Australia.,Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia
| | - Yasmyn Winstanley
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia
| | - Rebecca L Robker
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia.,Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Leanne Pacella-Ince
- Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia.,Repromed, Dulwich, Australia
| | - Macarena B Gonzalez
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia
| | - Nicole O McPherson
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia.,Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, Australia.,Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, Australia.,Repromed, Dulwich, Australia
| |
Collapse
|
39
|
Estefanía González-Alvarez M, Severin A, Sayadi M, Keating AF. PFOA-Induced Ovotoxicity Differs Between Lean and Obese Mice With Impacts on Ovarian Reproductive and DNA Damage Sensing and Repair Proteins. Toxicol Sci 2022; 190:173-188. [PMID: 36214631 PMCID: PMC9789752 DOI: 10.1093/toxsci/kfac104] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Perfluorooctanoic acid (PFOA) is an environmentally persistent perfluoroalkyl substance that is widely used in consumer products. Exposure to PFOA is associated with reproductive and developmental effects including endocrine disruption, delayed puberty in girls, and decreased fetal growth. In the United States, obesity affects 40% of women and 20% of girls, with higher rates in minority females. Obesity causes infertility, poor oocyte quality, miscarriage, and offspring defects. This study proposed that PFOA exposure would impact estrous cyclicity, ovarian steroid hormones, and the ovarian proteome and further hypothesized that obesity would impact PFOA-induced ovotoxicity. Female wild type (KK.Cg-a/a; lean) or KK.Cg-Ay/J mice (obese) received saline (CT) or PFOA (2.5 mg/kg) per os for 15 days beginning at 7 weeks of age. There were no effects on food intake, body weight, estrous cyclicity, serum progesterone, and heart, spleen, kidney, or uterus weight (p > .05). Ovary weight was decreased (p < .05) by PFOA exposure relative to vehicle control-treated mice in lean but not obese mice. Liquid chromatography-tandem mass spectrometry was performed on isolated ovarian protein and PFOA exposure altered the ovarian abundance of proteins involved in DNA damage sensing and repair pathways and reproduction pathways (p < .05) differentially in lean and obese mice. The data suggest that PFOA exposure alters ovary weight and differentially targets ovarian proteins in lean and obese females in ways that might reduce female fecundity.
Collapse
Affiliation(s)
| | - Andrew Severin
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames, Iowa 50011, USA
| | - Maryam Sayadi
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames, Iowa 50011, USA
| | - Aileen F Keating
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames, Iowa 50011, USA
| |
Collapse
|
40
|
Cinzori ME, Strakovsky RS. Effects of Elevated Maternal Adiposity on Offspring Reproductive Health: A Perspective From Epidemiologic Studies. J Endocr Soc 2022; 7:bvac163. [PMID: 36438545 PMCID: PMC9683496 DOI: 10.1210/jendso/bvac163] [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: 08/22/2022] [Indexed: 11/07/2022] Open
Abstract
One in seven couples in developed countries suffers from infertility. Maternal overweight or obesity have detrimental and lasting effects on offspring cardiometabolic health, and although substantially more data are needed, hormonal imbalances in utero resulting from excessive maternal adiposity could also disrupt reproductive programming and affect the future reproductive health of offspring. Therefore, this mini-review evaluates the human epidemiologic evidence that maternal overweight/obesity could be associated with poor reproductive health outcomes in offspring. We searched PubMed for relevant studies using terms such as "maternal obesity" and "reproductive development." While the human epidemiologic literature is limited, studies have thus far observed that maternal obesity is associated with disrupted external genital development and several other markers of reproductive health across the lifespan. Specifically, maternal obesity is associated with higher risks of hypospadias and cryptorchidism in males and disrupted anogenital distance both in males and females. Maternal obesity has also been linked to earlier age at menarche in daughters, and precocious puberty in both sons and daughters. Finally, daughters of women with overweight or obesity have higher risks of developing polycystic ovarian syndrome, which has implications for fertility. This body of research suggests that in utero exposure to maternal obesity could disrupt reproductive system development, but substantially more evidence is needed, as almost no human epidemiologic studies have evaluated the long-term consequences of maternal obesity with regard to offspring fertility/fecundity.
Collapse
Affiliation(s)
- Maria E Cinzori
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan 48824, USA
| | - Rita S Strakovsky
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
41
|
Hu D, Huang B, Xiong M, Yao J, Yang S, Wu R, Zhang H, Zhao Y. Impact of elevated body mass index on cumulative live birth rate and obstetric safety in women undergoing assisted reproductive technology. Sci Rep 2022; 12:18858. [PMID: 36344718 PMCID: PMC9640544 DOI: 10.1038/s41598-022-23576-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
This study evaluated the impact of elevated body mass index (BMI) on short- and long-term outcomes of in-vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) treatments. A total of 7229 patients undergoing IVF/ICSI fresh cycles and subsequent frozen embryo transfer cycles from 2014 to 2020 were divided into normal (18.5-24.9 kg/m2) and high BMI (≥ 25 kg/m2) groups. Ovarian response, pregnancy outcomes, and safety of both mother and fetus were the main outcome measures. Furthermore, multivariate analysis was used to determine whether BMI was associated with cumulative live birth rate (CLBR). Results showed that for younger women (< 38 year), CLBR was significantly reduced in the high BMI group compared with the normal BMI control and was accompanied by fewer retrieved oocytes and available embryos. Additionally, the incidence of hypertensive disorders of pregnancy, fetal macrosomia, and cleft lip and palate birth defects resulting from cumulative live births was significantly higher compared with the normal BMI group. No differences were observed among older women (≥ 38 year). Multivariate analysis revealed that high BMI was a risk factor for CLBR. Our study suggested that elevated BMI has a greater adverse impact on younger women.
Collapse
Affiliation(s)
- Dan Hu
- grid.33199.310000 0004 0368 7223Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030 People’s Republic of China
| | - Bo Huang
- grid.33199.310000 0004 0368 7223Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030 People’s Republic of China
| | - Min Xiong
- grid.33199.310000 0004 0368 7223Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030 People’s Republic of China
| | - Junning Yao
- grid.33199.310000 0004 0368 7223Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030 People’s Republic of China
| | - Shulin Yang
- grid.33199.310000 0004 0368 7223Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030 People’s Republic of China
| | - Ruxing Wu
- grid.33199.310000 0004 0368 7223Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030 People’s Republic of China
| | - Hanwang Zhang
- grid.33199.310000 0004 0368 7223Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030 People’s Republic of China
| | - Yiqing Zhao
- grid.33199.310000 0004 0368 7223Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030 People’s Republic of China
| |
Collapse
|
42
|
Schon SB, Yang K, Schindler R, Jiang L, Neff LM, Seeley RJ, Marsh EE. Obesity-related alterations in protein expression in human follicular fluid from women undergoing in vitro fertilization. F&S SCIENCE 2022; 3:331-339. [PMID: 36096447 DOI: 10.1016/j.xfss.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To compare the proteomic composition of follicular fluid from women with normal weight vs. women with obesity but without a history of polycystic ovary syndrome or known ovarian dysfunction undergoing in vitro fertilization. DESIGN Cross-sectional. SETTING Academic medical center. PATIENT(S) Eight women with normal weight and 8 women with obesity undergoing in vitro fertilization and without a history of polycystic ovary syndrome, ovulatory dysfunction, diminished ovarian reserve, or known endometriosis were included in the analysis. INTERVENTION(S) Not applicable. MAIN OUTCOME MEASURE(S) Proteomic assessment using liquid chromatography-mass spectrometry analysis. RESULT(S) The mean age of women with normal weight was similar to that of women with obesity (32.9 vs. 32.6 years, not significant). The mean body mass index of women with normal weight was 21.2 kg/m2 compared with a body mass index of 37.1 kg/m2 in women with obesity. A total of 1,174 proteins were identified with ≥2 peptides present. Twenty-five proteins were found to be significantly altered in the follicular fluid from women with obesity. Of these 25 proteins, 19 were up-regulated and 6 were down-regulated. Notably, C-reactive protein was 11-fold higher in the follicular fluid from women with obesity than in the follicular fluid from women with normal weight. CONCLUSION(S) Obesity is associated with dysregulation at the level of the follicle, including alterations in proteins related to inflammation and metabolism. These include proteins with emerging roles in energy homeostasis and follicular regulation.
Collapse
Affiliation(s)
- Samantha B Schon
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Kun Yang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Ronald Schindler
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | - Li Jiang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan
| | | | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Erica E Marsh
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
43
|
Thaller M, Mytton J, Wakerley BR, Mollan SP, Sinclair AJ. Idiopathic intracranial hypertension: Evaluation of births and fertility through the Hospital Episode Statistics dataset. BJOG 2022; 129:2019-2027. [PMID: 35620863 PMCID: PMC9796176 DOI: 10.1111/1471-0528.17241] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 05/05/2022] [Accepted: 05/19/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Idiopathic intracranial hypertension (IIH) predominantly affects women of reproductive age with obesity, and these women have a distinct profile of hyperandrogenism and insulin resistance. Polycystic ovary syndrome (PCOS) has an established adverse fertility phenotype that typically affects obese women. As IIH may impact reproductive health, we sought to evaluate fertility, gestational complications and pregnancy outcome in IIH. DESIGN Prospective cohort study from English Hospital Episode Statistics dataset. SETTING English hospitals, UK. POPULATION Women aged 18-45 years seen in English hospitals between 1 April 2002 and 31 March 2019. Patients were required to have an IIH diagnosis and were compared with those with PCOS and general population female controls. MAIN OUTCOME MEASURES Pregnancies resulting in live births, complications of gestational diabetes and pre-eclampsia, and method of delivery. RESULTS Data was collected from 17 587 IIH, 199633 PCOS and 10 947 012 women in the general population. The live birth rate, adjusted for age, was significantly lower among women with IIH (54.1%) than PCOS (67.9%), p < 0.0001 and the general population (57.7%), p < 0.0001. Pre-eclampsia and gestational diabetes risks were higher following a diagnosis of IIH (5.3-fold and 2.7-fold, respectively, p < 0.0001) compared with the general population controls. Following a diagnosis of IIH, elective caesarean section rates were more than twice that of general population (odds ratio [OR] 2.4) and prior to a diagnosis of IIH (OR 2.2). CONCLUSIONS These data indicate there are lower age-adjusted total pregnancy rates, increased risk of pre-eclampsia and gestational diabetes, and a doubling of elective caesarean section rates in those with a diagnosis of IIH.
Collapse
Affiliation(s)
- Mark Thaller
- Metabolic NeurologyInstitute of Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK,Department of NeurologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Jemma Mytton
- InformaticsUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Benjamin R. Wakerley
- Metabolic NeurologyInstitute of Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK,Department of NeurologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Susan P. Mollan
- Birmingham Neuro‐OphthalmologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK
| | - Alexandra J. Sinclair
- Metabolic NeurologyInstitute of Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK,Department of NeurologyUniversity Hospitals Birmingham NHS Foundation TrustBirminghamUK,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health PartnersBirminghamUK
| |
Collapse
|
44
|
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: 16] [Impact Index Per Article: 5.3] [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
|
45
|
Kelly AC, J Rosario F, Chan J, Cox LA, Powell TL, Jansson T. Transcriptomic responses are sex-dependent in the skeletal muscle and liver in offspring of obese mice. Am J Physiol Endocrinol Metab 2022; 323:E336-E353. [PMID: 35858246 PMCID: PMC9529275 DOI: 10.1152/ajpendo.00263.2021] [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: 07/22/2021] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022]
Abstract
Infants born to obese mothers are more likely to develop metabolic disease, including glucose intolerance and hepatic steatosis, in adult life. We examined the effects of maternal obesity on the transcriptome of skeletal muscle and liver tissues of the near-term fetus and 3-mo-old offspring in mice born to dams fed a high-fat and -sugar diet. Previously, we have shown that male, but not female, offspring develop glucose intolerance, insulin resistance, and liver steatosis at 3 mo old. Female C57BL6/J mice were fed normal chow or an obesogenic high-calorie diet before mating and throughout pregnancy. RNAseq was performed on the liver and gastrocnemius muscle following collection from fetuses on embryonic day 18.5 (E18.5) as well as from 3-mo-old offspring from obese dams and control dams. Significant genes were generated for each sex, queried for enrichment, and modeled to canonical pathways. RNAseq was corroborated by protein quantification in offspring. The transcriptomic response to maternal obesity in the liver was more marked in males than females. However, in both male and female offspring of obese dams, we found significant enrichment for fatty acid metabolism, mitochondrial transport, and oxidative stress in the liver transcriptomes as well as decreased protein concentrations of electron transport chain members. In skeletal muscle, pathway analysis of gene expression revealed sexual dimorphic patterns, including metabolic processes of fatty acids and glucose, as well as PPAR, AMPK, and PI3K-Akt signaling pathways. Transcriptomic responses to maternal obesity in skeletal muscle were more marked in female offspring than males. Female offspring had greater expression of genes associated with glucose uptake, and protein abundance reflected greater activation of mTOR signaling. Skeletal muscle and livers in mice born to obese dams had sexually dimorphic transcriptomic responses that changed from the fetus to the adult offspring. These data provide insights into mechanisms underpinning metabolic programming in maternal obesity.NEW & NOTEWORTHY Transcriptomic data support that fetuses of obese mothers modulate metabolism in both muscle and liver. These changes were strikingly sexually dimorphic in agreement with published findings that male offspring of obese dams exhibit pronounced metabolic disease earlier. In both males and females, the transcriptomic responses in the fetus were different than those at 3 mo, implicating adaptive mechanisms throughout adulthood.
Collapse
Affiliation(s)
- Amy C Kelly
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Fredrick J Rosario
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jeannie Chan
- Section of Molecular Medicine, Department of Internal Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Laura A Cox
- Section of Molecular Medicine, Department of Internal Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Theresa L Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Pediatrics, Section of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
46
|
Moorkens K, Leroy JLMR, Verheyen S, Marei WFA. Effects of an obesogenic diet on the oviduct depend on the duration of feeding. PLoS One 2022; 17:e0275379. [PMID: 36174086 PMCID: PMC9522283 DOI: 10.1371/journal.pone.0275379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/15/2022] [Indexed: 12/04/2022] Open
Abstract
Research question How long does it take for an obesogenic (high-fat/high-sugar, HF/HS) diet to influence the oviductal microenvironment? What are the affected cellular pathways and are they dependent on the genetic background of the mouse model? Design Female Swiss (outbred) and C57BL/6N (B6, inbred) mice were fed either a control (10% fat) or HF/HS (60% fat, 20% fructose) diet. Body weight was measured weekly. Mice were sacrificed at 3 days (3d), 1 week (1w), 4w, 8w, 12w and 16w on the diet (n = 5 per treatment per time point). Total cholesterol concentrations and inflammatory cytokines were measured in serum. Oviductal epithelial cells (OECs) were used to study the expression of genes involved in (mitochondrial) oxidative stress (OS), endoplasmic reticulum (ER) stress and inflammation using qPCR. Results Body weight and blood cholesterol increased significantly in the HF/HS mice in both strains compared to controls. In Swiss mice, HF/HS diet acutely increased ER-stress and OS-related genes in the OECs already after 3d. Subsequently, mitochondrial and cytoplasmic antioxidants were upregulated and ER-stress was alleviated at 1w. After 4-8w (mid-phase), the expression of ER-stress and OS-related genes was increased again and persisted throughout the late-phase (12-16w). Serum inflammatory cytokines and inflammatory marker-gene expression in the OECs were increased only in the late-phase. Some of the OEC stress responses were stronger or earlier in the B6. Conclusions OECs are sensitive to an obesogenic diet and may exhibit acute stress responses already after a few days of feeding. This may impact the oviductal microenvironment and contribute to diet-induced subfertility.
Collapse
Affiliation(s)
- Kerlijne Moorkens
- Department of Veterinary Sciences, Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Antwerp, Belgium
- * E-mail:
| | - Jo L. M. R. Leroy
- Department of Veterinary Sciences, Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Sara Verheyen
- Department of Veterinary Sciences, Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Waleed F. A. Marei
- Department of Veterinary Sciences, Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, University of Antwerp, Wilrijk, Antwerp, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
47
|
Wan Y, Muhammad T, Huang T, Lv Y, Sha Q, Yang S, Lu G, Chan WY, Ma J, Liu H. IGF2 reduces meiotic defects in oocytes from obese mice and improves embryonic developmental competency. Reprod Biol Endocrinol 2022; 20:101. [PMID: 35836183 PMCID: PMC9281013 DOI: 10.1186/s12958-022-00972-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Maternal obesity is a global issue that has devastating effects across the reproductive spectrum such as meiotic defects in oocytes, consequently worsening pregnancy outcomes. Different studies have shown that such types of meiotic defects originated from the oocytes of obese mothers. Thus, there is an urgent need to develop strategies to reduce the incidence of obesity-related oocyte defects that adversely affect pregnancy outcomes. Multiple growth factors have been identified as directly associated with female reproduction; however, the impact of various growth factors on female fertility in response to obesity remains poorly understood. METHODS The immature GV-stage oocytes from HFD female mice were collected and cultured in vitro in two different groups (HFD oocytes with and without 50 nM IGF2), however; the oocytes from ND mice were used as a positive control. HFD oocytes treated with or without IGF2 were further used to observe the meiotic structure using different analysis including, the spindle and chromosomal analysis, reactive oxygen species levels, mitochondrial functional activities, and early apoptotic index using immunofluorescence. Additionally, the embryonic developmental competency and embryos quality of IGF2-treated zygotes were also determined. RESULTS In our findings, we observed significantly reduced contents of insulin-like growth factor 2 (IGF2) in the serum and oocytes of obese mice. Our data indicated supplementation of IGF2 in a culture medium improves the blastocyst formation: from 46% in the HFD group to 61% in the HFD + IGF2-treatment group (50 nM IGF2). Moreover, adding IGF2 to the culture medium reduces the reactive oxygen species index and alleviates the frequency of spindle/chromosome defects. We found increased mitochondrial functional activity in oocytes from obese mice after treating the oocytes with IGF2: observed elevated level of adenosine triphosphate, increased mitochondrial distribution, higher mitochondrial membrane potentials, and reduced mitochondrial ultrastructure defects. Furthermore, IGF2 administration also increases the overall protein synthesis and decreases the apoptotic index in oocytes from obese mice. CONCLUSIONS Collectively, our findings are strongly in favor of adding IGF2 in culture medium to overcome obesity-related meiotic structural-developmental defects by helping ameliorate the known sub-optimal culturing conditions that are currently standard with assisted reproduction technologies.
Collapse
Affiliation(s)
- Yanling Wan
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Tahir Muhammad
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Tao Huang
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
| | - Yue Lv
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, Beijing, China
| | - Qianqian Sha
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Shuang Yang
- Department of Physiology School of Basic Medical Sciences Cheeloo College of Medicine Shandong University, Jinan, Shandong, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai-Yee Chan
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jinlong Ma
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hongbin Liu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China.
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, Beijing, China.
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
48
|
Horn EJ, Read CC, Edwards JL, Schrick FN, Rhinehart JD, Payton RR, Campagna SR, Klabnik JL, Clark HM, Myer PR, McLean KJ, Moorey SE. Preovulatory follicular fluid and serum metabolome profiles in lactating beef cows with thin, moderate, and obese body condition. J Anim Sci 2022; 100:skac152. [PMID: 35772755 PMCID: PMC9246665 DOI: 10.1093/jas/skac152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/25/2022] [Indexed: 12/20/2022] Open
Abstract
Extremes in body condition reduce fertility and overall productivity in beef cattle herds, due in part to altered systemic metabolic conditions that influence the intrafollicular and uterine environment. Follicular fluid and serum metabolome profiles are influenced by body composition in women and dairy cattle; however, such information is lacking in beef cattle. We hypothesized that body condition score (BCS)-related alterations in the metabolome of preovulatory follicular fluid and serum may influence oocyte maturation while impacting the oviductal or uterine environment. Therefore, we performed a study with the objective to determine the relationship between BCS and the metabolome of follicular fluid and serum in lactating beef cattle. We synchronized the development of a preovulatory follicle in 130 cows of varying BCS. We collected blood and performed transvaginal follicle aspirations to collect follicular fluid from the preovulatory follicle ~18 h after gonadotropin-releasing hormone administration to stimulate the preovulatory gonadotropin surge. We then selected follicular fluid and serum samples from cows with BCS 4 (Thin; n = 14), BCS 6 (Moderate; n = 18), or BCS >8 (Obese; n = 14) for ultra-high performance liquid chromatography-high resolution mass spectrometry. We identified differences in the follicular fluid or serum of thin, moderate, and obese animals based on multiple linear regression. MetaboAnalyst 5.0 was used for enrichment analysis of significant metabolites. We identified 38 metabolites in follicular fluid and 49 metabolites in serum. There were no significant differences in follicular fluid metabolite content among BCS classifications. There were 5, 22, and 1 serum metabolites differentially abundant between thin-obese, moderate-thin, and moderate-obese classifications, respectively (false discovery rate [FDR] < 0.10). These metabolites were enriched in multiple processes including "arginine biosynthesis," "arginine/proline metabolism," and "D-glutamine/D-glutamate metabolism" (FDR < 0.04). Pathways enriched with serum metabolites associated with BCS indicate potentially increased reactive oxygen species (ROS) in serum of thin cows. ROS crossing the blood follicular barrier may negatively impact the oocyte during oocyte maturation and contribute to the reduced pregnancy rates observed in thin beef cows.
Collapse
Affiliation(s)
- Emma J Horn
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Casey C Read
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - J Lannett Edwards
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - F Neal Schrick
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Justin D Rhinehart
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Rebecca R Payton
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Shawn R Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, USA
| | - Jessica L Klabnik
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Hannah M Clark
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Phillip R Myer
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Kyle J McLean
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| | - Sarah E Moorey
- Department of Animal Science, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
49
|
Fabian D, Kubandová-Babeľová J, Kšiňanová M, Waczulíková I, Fabianová K, Koppel J. Overweight and Fertility: What We Can Learn from an Intergenerational Mouse Obesity Model. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19137918. [PMID: 35805577 PMCID: PMC9266121 DOI: 10.3390/ijerph19137918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 12/02/2022]
Abstract
The aim of this study was to evaluate the effects of being overweight on the ability to conceive, fertilization rate, and in vivo development of embryos in regularly cycling, spontaneously ovulating, and naturally mated female mice. The study was based on statistical analysis of data collected during 14 experiments with identical design, performed on 319 control and 327 obese mice, developed in an intergenerational model of obesity induction which eliminates the impact of aging and high-fat feeding. Six-week-old mice with a vaginal sperm plug were slaughtered on embryonic days 2, 3, or 4, and the flushed contents of the oviducts and uteri were assessed by stereomicroscopy. The results showed no association between being overweight and the proportion of ovulating or fertilized females. On the other hand, a strong association was found between being overweight and ovulation yield. On embryonic day 2, significantly higher numbers of eggs were recovered from the oviducts of fertilized obese mice. Maternal overweight status was also associated with higher developmental capacities of preimplantation embryos. In conclusion, contrary to studies based on the high-fat-diet model, in female mice fed regular chow, being overweight was associated with an increased ovulation quota and higher developmental rate of fertilized oocytes. Being overweight did not impact ability to conceive. On the other hand, as documented in our previous studies, the quality of oocytes and blastocysts recovered from overweight mice developed in an intergenerational model of obesity was low.
Collapse
Affiliation(s)
- Dušan Fabian
- Centre of Biosciences, Institute of Animal Physiology, Slovak Academy of Sciences, Šoltésovej 4/6, 040 01 Košice, Slovakia; (J.K.-B.); (J.K.)
- Correspondence:
| | - Janka Kubandová-Babeľová
- Centre of Biosciences, Institute of Animal Physiology, Slovak Academy of Sciences, Šoltésovej 4/6, 040 01 Košice, Slovakia; (J.K.-B.); (J.K.)
| | - Martina Kšiňanová
- Centre of Biosciences, Institute of Molecular Physiology and Genetics, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia;
| | - Iveta Waczulíková
- Faculty of Mathematics, Physics and Informatics, Comenius University, Mlynská dolina F1, 842 48 Bratislava, Slovakia;
| | - Kamila Fabianová
- Biomedical Research Center, Institute of Neurobiology, Slovak Academy of Sciences, Šoltésovej 4/6, 040 01 Košice, Slovakia;
| | - Juraj Koppel
- Centre of Biosciences, Institute of Animal Physiology, Slovak Academy of Sciences, Šoltésovej 4/6, 040 01 Košice, Slovakia; (J.K.-B.); (J.K.)
| |
Collapse
|
50
|
Zhu S, Wang Q. Metabolic control of oocyte development. Biol Reprod 2022; 107:54-61. [PMID: 35470861 DOI: 10.1093/biolre/ioac082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/19/2022] [Accepted: 04/18/2022] [Indexed: 11/12/2022] Open
Abstract
Well balanced and timed metabolism is essential for oocyte development. The effects of extrinsic nutrients on oocyte maturation have been widely reported. In contrast, intrinsic control of oogenesis by intracellular metabolites and metabolic enzymes has received little attention. The comprehensive characterization of metabolic patterns could lead to more complete understanding of regulatory mechanisms underlying oocyte development. A cell's metabolic state is integrated with epigenetic regulation. Epigenetic modifications in germ cells are therefore sensitive to parental environmental exposures. Nevertheless, direct genetic evidence for metabolites involvement in epigenetic establishment during oocyte development is still lacking. Moreover, metabolic disorder-induced epigenetic perturbations during oogenesis might mediate the inter/transgenerational effects of environmental insults. The molecular mechanisms responsible for this deserve further investigation. Here, we summarize the findings on metabolic regulation in oocyte maturation, and how it contributes to oocyte epigenetic modification. Finally, we propose a mouse model that metabolic disorder in oocyte serves as a potential factor mediating the maternal environment effects on offspring health.
Collapse
Affiliation(s)
- Shuai Zhu
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing 211166, China.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|