1
|
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
|
2
|
Herup-Wheeler T, Shi M, Harvey ME, Talwar C, Kommagani R, MacLean JA, Hayashi K. High-fat diets promote peritoneal inflammation and augment endometriosis-associated abdominal hyperalgesia. Front Endocrinol (Lausanne) 2024; 15:1336496. [PMID: 38559689 PMCID: PMC10978581 DOI: 10.3389/fendo.2024.1336496] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Immune dysfunction is one of the central components in the development and progression of endometriosis by establishing a chronic inflammatory environment. Western-style high-fat diets (HFD) have been linked to greater systemic inflammation to cause metabolic and chronic inflammatory diseases, and are also considered an environmental risk factor for gynecologic diseases. Here, we aimed to examine how HFD cause an inflammatory environment in endometriosis and discern their contribution to endometriotic-associated hyperalgesia. Our results showed that HFD-induced obesity enhanced abdominal hyperalgesia that was induced by endometriotic lesions. Peritoneal inflammatory macrophages and cytokine levels increased by lesion induction were elevated by chronic exposure to HFD. Increased expression of pain-related mediators in the dorsal root ganglia was observed after lesion induction under the HFD condition. Although HFD did not affect inflammatory macrophages in the peritoneal cavity without lesion induction, the diversity and composition of the gut microbiota were clearly altered by HFD as a sign of low-grade systemic inflammation. Thus, HFD alone might not establish a local inflammatory environment in the pelvic cavity, but it can contribute to further enhancing chronic inflammation, leading to the exacerbation of endometriosis-associated abdominal hyperalgesia following the establishment and progression of the disease.
Collapse
Affiliation(s)
- Tristin Herup-Wheeler
- School of Molecular Bioscience, Center for Reproductive Biology, Washington State University, Pullman, WA, United States
| | - Mingxin Shi
- School of Molecular Bioscience, Center for Reproductive Biology, Washington State University, Pullman, WA, United States
| | - Madeleine E. Harvey
- School of Molecular Bioscience, Center for Reproductive Biology, Washington State University, Pullman, WA, United States
| | - Chandni Talwar
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Ramakrishna Kommagani
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - James A. MacLean
- School of Molecular Bioscience, Center for Reproductive Biology, Washington State University, Pullman, WA, United States
| | - Kanako Hayashi
- School of Molecular Bioscience, Center for Reproductive Biology, Washington State University, Pullman, WA, United States
| |
Collapse
|
3
|
Pinto S, Guerra-Carvalho B, Crisóstomo L, Rocha A, Barros A, Alves MG, Oliveira PF. Metabolomics Integration in Assisted Reproductive Technologies for Enhanced Embryo Selection beyond Morphokinetic Analysis. Int J Mol Sci 2023; 25:491. [PMID: 38203668 PMCID: PMC10778973 DOI: 10.3390/ijms25010491] [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: 11/02/2023] [Revised: 12/12/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Embryo quality evaluation during in vitro development is a crucial factor for the success of assisted reproductive technologies (ARTs). However, the subjectivity inherent in the morphological evaluation by embryologists can introduce inconsistencies that impact the optimal embryo choice for transfer. To provide a more comprehensive evaluation of embryo quality, we undertook the integration of embryo metabolomics alongside standardized morphokinetic classification. The culture medium of 55 embryos (derived from 21 couples undergoing ICSI) was collected at two timepoints (days 3 and 5). Samples were split into Good (n = 29), Lagging (n = 19), and Bad (n = 10) according to embryo morphokinetic evaluation. Embryo metabolic performance was assessed by monitoring the variation in specific metabolites (pyruvate, lactate, alanine, glutamine, acetate, formate) using 1H-NMR. Adjusted metabolite differentials were observed during the first 3 days of culture and found to be discriminative of embryo quality at the end of day 5. Pyruvate, alanine, glutamine, and acetate were major contributors to this discrimination. Good and Lagging embryos were found to export and accumulate pyruvate and glutamine in the first 3 days of culture, while Bad embryos consumed them. This suggests that Bad embryos have less active metabolic activity than Good and Lagging embryos, and these two metabolites are putative biomarkers for embryo quality. This study provides a more comprehensive evaluation of embryo quality and can lead to improvements in ARTs by enabling the selection of the best embryos. By combining morphological assessment and metabolomics, the selection of high-quality embryos with the potential to result in successful pregnancies may become more accurate and consistent.
Collapse
Affiliation(s)
- Soraia Pinto
- Centre for Reproductive Genetics Alberto Barros, 4100-012 Porto, Portugal; (S.P.); (A.B.)
| | | | - Luís Crisóstomo
- Institute of Biomedicine, University of Turku, 20014 Turku, Finland;
| | - António Rocha
- CECA/ICETA–Centro de Estudos de Ciência Animal, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4200-135 Porto, Portugal;
| | - Alberto Barros
- Centre for Reproductive Genetics Alberto Barros, 4100-012 Porto, Portugal; (S.P.); (A.B.)
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Marco G. Alves
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Pedro F. Oliveira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| |
Collapse
|
4
|
Herup-Wheeler T, Shi M, Harvey ME, Talwar C, Kommagani R, MacLean JA, Hayashi K. High-fat diets promote peritoneal inflammation and augment endometriosis-associated abdominal hyperalgesia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566474. [PMID: 38014254 PMCID: PMC10680790 DOI: 10.1101/2023.11.09.566474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Immune dysfunction is one of the central components in the development and progression of endometriosis by establishing a chronic inflammatory environment. Western-style high-fat diets (HFD) have been linked to greater systemic inflammation to cause metabolic and chronic inflammatory diseases, and are also considered an environmental risk factor for gynecologic diseases. Here, we aimed to examine how HFD alter an inflammatory environment in endometriosis and discern their contribution to endometriotic-associated hyperalgesia. Our results showed that HFD-induced obesity enhanced abdominal mechanical allodynia that was induced by endometriotic lesions. Peritoneal inflammatory macrophages and cytokine levels increased by lesion induction were elevated by chronic exposure to HFD. Pain-related mediators in the dorsal root ganglia were further stimulated after lesion induction under the HFD condition. Although HFD did not affect inflammatory macrophages in the peritoneal cavity without lesion induction, the diversity and composition of the gut microbiota were clearly altered by HFD as a sign of low-grade systemic inflammation. Thus, HFD alone might not establish a local inflammatory environment in the pelvic cavity, but it can contribute to further enhancing chronic inflammation, leading to the exacerbation of endometriosis-associated abdominal hyperalgesia following the establishment and progression of the disease.
Collapse
Affiliation(s)
- Tristin Herup-Wheeler
- School of Molecular Bioscience, Center for Reproductive Biology, Washington State University, Pullman, WA 99614, USA
| | - Mingxin Shi
- School of Molecular Bioscience, Center for Reproductive Biology, Washington State University, Pullman, WA 99614, USA
| | - Madeleine E Harvey
- School of Molecular Bioscience, Center for Reproductive Biology, Washington State University, Pullman, WA 99614, USA
| | - Chandni Talwar
- Department of Pathology & Immunology, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ramakrishna Kommagani
- Department of Pathology & Immunology, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - James A MacLean
- School of Molecular Bioscience, Center for Reproductive Biology, Washington State University, Pullman, WA 99614, USA
| | - Kanako Hayashi
- School of Molecular Bioscience, Center for Reproductive Biology, Washington State University, Pullman, WA 99614, USA
| |
Collapse
|
5
|
Peel A, Saini A, Deluao JC, McPherson NO. Sperm DNA damage: The possible link between obesity and male infertility, an update of the current literature. Andrology 2023; 11:1635-1652. [PMID: 36789664 DOI: 10.1111/andr.13409] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/13/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
Obesity prevalence worldwide is increasing significantly. Whilst maternal obesity has clear detrimental impacts on fertility, pregnancy and foetal outcomes, more recently there has been an increasing focus on the role of paternal obesity in human fertility. Recent meta-analyses have indicated that obesity in men negatively affects basic sperm parameters such as sperm count, concentration and motility, increases the incidence of infertility and reduces the chances of conception. Sperm DNA damage, typically characterised by DNA strand breaks and oxidation of DNA nucleotides, is a specialised marker of sperm quality that has been independently associated with recurrent miscarriage, reduced assisted reproduction success and increased mutational loads in subsequent offspring. Whilst, there are still conflicting data in humans as to the association of obesity in men with sperm DNA damage, evidence from rodent models is clear, indicating that male obesity increases sperm DNA damage. Human data are often conflicting because of the large heterogeneity amongst studies, the use of body mass index as the indicator of obesity and the methods used for detection of sperm DNA damage. Furthermore, comorbidities of obesity (i.e., heat stress, adipokines, insulin resistance, changes in lipids, hypogonadism and obstructive sleep apnoea) are also independently associated with increased sperm DNA damage that is not always modified in men with obesity, and as such may provide a causative link to the discrepancies amongst human studies. In this review, we provide an update on the literature regarding the associations between obesity in men and fertility, basic sperm parameters and sperm DNA damage. We further discuss potential reasons for the discrepancies in the literature and outline possible direct and indirect mechanisms of increased sperm DNA damage resulting from obesity. Finally, we summarise intergenerational obesity through the paternal linage and how sperm DNA damage may contribute to the transmission.
Collapse
Affiliation(s)
- Andrew Peel
- Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anmol Saini
- Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Joshua C Deluao
- Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, South Australia, Australia
| | - Nicole O McPherson
- Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
- Freemasons Centre for Male Health and Wellbeing, The University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Health and Medical School, School of Biomedicine, Discipline of Reproduction and Development, The University of Adelaide, Adelaide, South Australia, Australia
- Repromed IVF Adelaide, Dulwich, South Australia, Australia
| |
Collapse
|
6
|
Velazquez MA, Idriss A, Chavatte-Palmer P, Fleming TP. The mammalian preimplantation embryo: Its role in the environmental programming of postnatal health and performance. Anim Reprod Sci 2023; 256:107321. [PMID: 37647800 DOI: 10.1016/j.anireprosci.2023.107321] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 09/01/2023]
Abstract
During formation of the preimplantation embryo several cellular and molecular milestones take place, making the few cells forming the early embryo vulnerable to environmental stressors than can impair epigenetic reprogramming and controls of gene expression. Although these molecular alterations can result in embryonic death, a significant developmental plasticity is present in the preimplantation embryo that promotes full-term pregnancy. Prenatal epigenetic modifications are inherited during mitosis and can perpetuate specific phenotypes during early postnatal development and adulthood. As such, the preimplantation phase is a developmental window where developmental programming can take place in response to the embryonic microenvironment present in vivo or in vitro. In this review, the relevance of the preimplantation embryo as a developmental stage where offspring health and performance can be programmed is discussed, with emphasis on malnutrition and assisted reproductive technologies; two major environmental insults with important implications for livestock production and human reproductive medicine.
Collapse
Affiliation(s)
- Miguel A Velazquez
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK.
| | - Abdullah Idriss
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; Pathology and laboratory medicine, King Faisal Specialist Hospital and Research Centre, P.O. Box 40047, MBC J-10, Jeddah 21499, Kingdom of Saudi Arabia
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; Ecole Nationale Vétérinaire d'Alfort, BREED, 94700 Maisons-Alfort, France
| | - Tom P Fleming
- Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
7
|
Lin Y, Chen Z, Qian Q, Wang Y, Xiu X, Ou P, Fang J, Li G. Effects of paternal obesity on maternal-neonatal outcomes and long-term prognosis in adolescents. Front Endocrinol (Lausanne) 2023; 14:1114250. [PMID: 37082119 PMCID: PMC10111374 DOI: 10.3389/fendo.2023.1114250] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/16/2023] [Indexed: 04/07/2023] Open
Abstract
ObjectiveThis study evaluated whether paternal body mass index (BMI) before pregnancy was a risk factor for maternal-neonatal outcomes and long-term prognosis in offspring.MethodsThis study included 29,518 participants from eight cities in Fujian, China using a stratified cluster random sampling method from May to September 2019. They were divided into four groups based on paternal BMI. Univariate and multivariate logistic regression were used to explore the relationship between paternal BMI groups, maternal-neonatal outcomes, and long-term prognosis in offspring. Further subgroup analysis was conducted to examine the stability of the risk. ResultsThe incidences of hypertensive disorder complicating pregnancy (HDCP), cesarean delivery, gestational weight gain (GWG) over guideline, and macrosomia were significantly higher in the paternal overweight and obesity group. Importantly, this study demonstrated that the incidence of asthma, hand-foot-and-mouth disease (HFMD), anemia, dental caries, and obesity of adolescents in paternal obesity increased. Furthermore, logistic regression and subgroup analysis confirm paternal obesity is a risk factor for HDCP, cesarean delivery, and macrosomia. It caused poor long-term prognosis in adolescents, including asthma, dental caries, and HFMD.ConclusionsPaternal obesity is a risk factor for adverse maternal-neonatal outcomes and poor long-term prognosis in adolescents. In addition to focusing on maternal weight, expectant fathers should pay more attention to weight management since BMI is a modifiable risk factor. Preventing paternal obesity can lead to better maternal and child outcomes. It would provide new opportunities for chronic diseases.
Collapse
Affiliation(s)
- Yingying Lin
- Department of Healthcare, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Zhiwei Chen
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Qinfang Qian
- Department of Child Healthcare Centre, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Yanxia Wang
- Department of Child Healthcare Centre, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Xiaoyan Xiu
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Ping Ou
- Department of Healthcare, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Guobo Li, ; Jiaoning Fang, ; Ping Ou,
| | - Jiaoning Fang
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Guobo Li, ; Jiaoning Fang, ; Ping Ou,
| | - Guobo Li
- Department of Child Healthcare Centre, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- *Correspondence: Guobo Li, ; Jiaoning Fang, ; Ping Ou,
| |
Collapse
|
8
|
Sharma Y, Galvão AM. Maternal obesity and ovarian failure: is leptin the culprit? Anim Reprod 2023; 19:e20230007. [PMID: 36855701 PMCID: PMC9968511 DOI: 10.1590/1984-3143-ar2023-0007] [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: 01/10/2023] [Accepted: 01/24/2023] [Indexed: 02/22/2023] Open
Abstract
At the time of its discovery and characterization in 1994, leptin was mostly considered a metabolic hormone able to regulate body weight and energy homeostasis. However, in recent years, a great deal of literature has revealed leptin's pleiotropic nature, through its involvement in numerous physiological contexts including the regulation of the female reproductive tract and ovarian function. Obesity has been largely associated with infertility, and leptin signalling is known to be dysregulated in the ovaries of obese females. Hence, the disruption of ovarian leptin signalling was shown to contribute to the pathophysiology of ovarian failure in obese females, affecting transcriptional programmes in the gamete and somatic cells. This review attempts to uncover the underlying mechanisms contributing to female infertility associated with obesity, as well as to shed light on the role of leptin in the metabolic dysregulation within the follicle, the effects on the oocyte epigenome, and the potential long-term consequence to embryo programming.
Collapse
Affiliation(s)
- Yashaswi Sharma
- Institute of Animal Reproduction and Food Research of PAS, Department of Reproductive Immunology and Pathology, Olsztyn, Poland
| | - António Miguel Galvão
- Institute of Animal Reproduction and Food Research of PAS, Department of Reproductive Immunology and Pathology, Olsztyn, Poland,Babraham Institute, Epigenetics Programme, Cambridge, United Kingdom UK,Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom UK,Corresponding author: ;
| |
Collapse
|
9
|
Sferruzzi‐Perri AN, Lopez‐Tello J, Salazar‐Petres E. Placental adaptations supporting fetal growth during normal and adverse gestational environments. Exp Physiol 2023; 108:371-397. [PMID: 36484327 PMCID: PMC10103877 DOI: 10.1113/ep090442] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
NEW FINDINGS What is the topic of this review? How the placenta, which transports nutrients and oxygen to the fetus, may alter its support of fetal growth developmentally and with adverse gestational conditions. What advances does it highlight? Placental formation and function alter with the needs of the fetus for substrates for growth during normal gestation and when there is enhanced competition for substrates in species with multiple gestations or adverse gestational environments, and this is mediated by imprinted genes, signalling pathways, mitochondria and fetal sexomes. ABSTRACT The placenta is vital for mammalian development and a key determinant of life-long health. It is the interface between the mother and fetus and is responsible for transporting the nutrients and oxygen a fetus needs to develop and grow. Alterations in placental formation and function, therefore, have consequences for fetal growth and birthweight, which in turn determine perinatal survival and risk of non-communicable diseases for the offspring in later postnatal life. However, the placenta is not a static organ. As this review summarizes, research from multiple species has demonstrated that placental formation and function alter developmentally to the needs of the fetus for substrates for growth during normal gestation, as well as when there is greater competition for substrates in polytocous species and monotocous species with multiple gestations. The placenta also adapts in response to the gestational environment, integrating information about the ability of the mother to provide nutrients and oxygen with the needs of the fetus in that prevailing environment. In particular, placental structure (e.g. vascularity, surface area, blood flow, diffusion distance) and transport capacity (e.g. nutrient transporter levels and activity) respond to suboptimal gestational environments, namely malnutrition, obesity, hypoxia and maternal ageing. Mechanisms mediating developmentally and environmentally induced homeostatic responses of the placenta that help support normal fetal growth include imprinted genes, signalling pathways, subcellular constituents and fetal sexomes. Identification of these placental strategies may inform the development of therapies for complicated human pregnancies and advance understanding of the pathways underlying poor fetal outcomes and their consequences for health and disease risk.
Collapse
Affiliation(s)
- Amanda Nancy Sferruzzi‐Perri
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Jorge Lopez‐Tello
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Esteban Salazar‐Petres
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Facultad de CienciasDepartamento de Ciencias Básicas, Universidad Santo TomásValdiviaChile
| |
Collapse
|
10
|
Bhadsavle SS, Golding MC. Paternal epigenetic influences on placental health and their impacts on offspring development and disease. Front Genet 2022; 13:1068408. [PMID: 36468017 PMCID: PMC9716072 DOI: 10.3389/fgene.2022.1068408] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 07/25/2023] Open
Abstract
Our efforts to understand the developmental origins of birth defects and disease have primarily focused on maternal exposures and intrauterine stressors. Recently, research into non-genomic mechanisms of inheritance has led to the recognition that epigenetic factors carried in sperm also significantly impact the health of future generations. However, although researchers have described a range of potential epigenetic signals transmitted through sperm, we have yet to obtain a mechanistic understanding of how these paternally-inherited factors influence offspring development and modify life-long health. In this endeavor, the emerging influence of the paternal epigenetic program on placental development, patterning, and function may help explain how a diverse range of male exposures induce comparable intergenerational effects on offspring health. During pregnancy, the placenta serves as the dynamic interface between mother and fetus, regulating nutrient, oxygen, and waste exchange and coordinating fetal growth and maturation. Studies examining intrauterine maternal stressors routinely describe alterations in placental growth, histological organization, and glycogen content, which correlate with well-described influences on infant health and adult onset of disease. Significantly, the emergence of similar phenotypes in models examining preconception male exposures indicates that paternal stressors transmit an epigenetic memory to their offspring that also negatively impacts placental function. Like maternal models, paternally programmed placental dysfunction exerts life-long consequences on offspring health, particularly metabolic function. Here, focusing primarily on rodent models, we review the literature and discuss the influences of preconception male health and exposure history on placental growth and patterning. We emphasize the emergence of common placental phenotypes shared between models examining preconception male and intrauterine stressors but note that the direction of change frequently differs between maternal and paternal exposures. We posit that alterations in placental growth, histological organization, and glycogen content broadly serve as reliable markers of altered paternal developmental programming, predicting the emergence of structural and metabolic defects in the offspring. Finally, we suggest the existence of an unrecognized developmental axis between the male germline and the extraembryonic lineages that may have evolved to enhance fetal adaptation.
Collapse
Affiliation(s)
| | - Michael C. Golding
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
11
|
Anwer H, Mason D, Zajitschek S, Hesselson D, Noble DWA, Morris MJ, Lagisz M, Nakagawa S. Intergenerational effects of overfeeding on aversive learning in zebrafish ( Danio rerio). Ecol Evol 2022; 12:e9423. [PMID: 36311397 PMCID: PMC9596326 DOI: 10.1002/ece3.9423] [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: 04/28/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
The obesity epidemic is concerning as obesity appears to negatively impact cognition and behavior. Furthermore, some studies suggest that this negative effect could be carried across generations from both mothers and fathers although evidence is not consistent. Here, we attempt to address how obesogenic diets in the parental generation (F0) can impact offspring's cognition and anxiety intergenerationally (F1) in a zebrafish model. We compare both mean trait values and their variances. Using a multifactorial design, we created a total of four groups: F1T (treatment mothers × treatment fathers); F1M (treatment mothers × control fathers); F1P (treatment fathers × control mothers); and F1C (control mothers × control fathers, F1C); and subjected them to anxiety tank tests and aversive learning assays. When both parents were exposed, offspring (F1T) displayed the poorest aversive learning, while offspring that only had one parent exposed (F1P and F1M) learnt the aversive learning task the best. Zebrafish in all groups displayed no statistically significant differences in anxiety-associated behaviors. Males and females also performed similarly in both anxiety and aversive learning assays. While all F1 groups had similar levels of fasting blood glucose, variance in glucose levels were reduced in F1P and F1T indicating the importance of investigating heteroskedasticity between groups. Furthermore, anxiety behaviors of these two groups appeared to be less repeatable. To our knowledge, this is the first study to test the intergenerational effects of an obesogenic diet on zebrafish cognition. Our multifactorial design as well as repeated tests also allowed us to disentangle maternal and paternal effects (as well as combined effects) and accurately detect subtle information such as between-individual variation.
Collapse
Affiliation(s)
- Hamza Anwer
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
| | - Dominic Mason
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
| | - Susanne Zajitschek
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
- Liverpool John Moores UniversitySchool of Biological and Environmental SciencesLiverpoolUK
| | - Daniel Hesselson
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
- Centenary Institute and Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| | - Daniel W. A. Noble
- Division of Ecology and Evolution, Research School of BiologyThe Australian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Margaret J. Morris
- Department of Pharmacology, School of Medical SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - Malgorzata Lagisz
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
| | - Shinichi Nakagawa
- Evolution & Ecology Research Centre and School of Biological, Earth and Environmental SciencesUniversity of New South WalesSydneyNew South WalesAustralia
- Diabetes and Metabolism DivisionGarvan Institute of Medical ResearchDarlinghurst, SydneyNew South WalesAustralia
| |
Collapse
|
12
|
Batra V, Norman E, Morgan HL, Watkins AJ. Parental Programming of Offspring Health: The Intricate Interplay between Diet, Environment, Reproduction and Development. Biomolecules 2022; 12:biom12091289. [PMID: 36139133 PMCID: PMC9496505 DOI: 10.3390/biom12091289] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
As adults, our health can be influenced by a range of lifestyle and environmental factors, increasing the risk for developing a series of non-communicable diseases such as type 2 diabetes, heart disease and obesity. Over the past few decades, our understanding of how our adult health can be shaped by events occurring before birth has developed into a well-supported concept, the Developmental Origins of Health and Disease (DOHaD). Supported by epidemiological data and experimental studies, specific mechanisms have been defined linking environmental perturbations, disrupted fetal and neonatal development and adult ill-health. Originally, such studies focused on the significance of poor maternal health during pregnancy. However, the role of the father in directing the development and well-being of his offspring has come into recent focus. Whereas these studies identify the individual role of each parent in shaping the long-term health of their offspring, few studies have explored the combined influences of both parents on offspring well-being. Such understanding is necessary as parental influences on offspring development extend beyond the direct genetic contributions from the sperm and oocyte. This article reviews our current understanding of the parental contribution to offspring health, exploring some of the mechanisms linking parental well-being with gamete quality, embryo development and offspring health.
Collapse
|
13
|
Jazwiec PA, Patterson VS, Ribeiro TA, Yeo E, Kennedy KM, Mathias PCF, Petrik JJ, Sloboda DM. Paternal obesity induces placental hypoxia and sex-specific impairments in placental vascularization and offspring metabolism. Biol Reprod 2022; 107:574-589. [PMID: 35377412 PMCID: PMC9382389 DOI: 10.1093/biolre/ioac066] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/31/2022] [Indexed: 12/03/2022] Open
Abstract
Paternal obesity predisposes offspring to metabolic dysfunction, but the underlying mechanisms remain unclear. We investigated whether this metabolic dysfunction is associated with changes in placental vascular development and is fueled by endoplasmic reticulum (ER) stress-mediated changes in fetal hepatic development. We also determined whether paternal obesity indirectly affects the in utero environment by disrupting maternal metabolic adaptations to pregnancy. Male mice fed a standard chow or high fat diet (60%kcal fat) for 8–10 weeks were time-mated with female mice to generate pregnancies and offspring. Glucose tolerance was evaluated in dams at mid-gestation (embryonic day (E) 14.5) and late gestation (E18.5). Hypoxia, angiogenesis, endocrine function, macronutrient transport, and ER stress markers were evaluated in E14.5 and E18.5 placentae and/or fetal livers. Maternal glucose tolerance was assessed at E14.5 and E18.5. Metabolic parameters were assessed in offspring at ~60 days of age. Paternal obesity did not alter maternal glucose tolerance but induced placental hypoxia and altered placental angiogenic markers, with the most pronounced effects in female placentae. Paternal obesity increased ER stress-related protein levels (ATF6 and PERK) in the fetal liver and altered hepatic expression of gluconeogenic factors at E18.5. Offspring of obese fathers were glucose intolerant and had impaired whole-body energy metabolism, with more pronounced effects in female offspring. Metabolic deficits in offspring due to paternal obesity may be mediated by sex-specific changes in placental vessel structure and integrity that contribute to placental hypoxia and may lead to poor fetal oxygenation and impairments in fetal metabolic signaling pathways in the liver.
Collapse
Affiliation(s)
- Patrycja A Jazwiec
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8S 4L8, Canada
| | - Violet S Patterson
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8S 4L8, Canada
| | - Tatiane A Ribeiro
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8S 4L8, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton L8S 4L8, Canada.,Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Paraná 87020-900, Brazil
| | - Erica Yeo
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8S 4L8, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton L8S 4L8, Canada
| | - Katherine M Kennedy
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8S 4L8, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton L8S 4L8, Canada
| | - Paulo C F Mathias
- Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Paraná 87020-900, Brazil
| | - Jim J Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph N1G 2W1, Canada
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8S 4L8, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton L8S 4L8, Canada.,Department of Pediatrics, McMaster University, Hamilton L8S 4L8, Canada.,Department of Obstetrics and Gynecology, McMaster University, Hamilton L8S 4L8, Canada
| |
Collapse
|
14
|
Han Y, Zhang M, Duan J, Li L, Du J, Cheng H, Zhang S, Zhai Y, An X, Li Q, Zhang X, Li Z, Tang B. Maternal Prepregnancy 5-Hydroxytryptamine Exposure Affects the Early Development of the Fetus. Front Physiol 2022; 13:761357. [PMID: 35370795 PMCID: PMC8969228 DOI: 10.3389/fphys.2022.761357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/31/2022] [Indexed: 11/29/2022] Open
Abstract
In recent decades, the increasing incidence of depression has contributed to an increase in the use of serotonergic drugs, such as antidepressants, which predisposes humans to serotonin syndrome. Serotonin syndrome is caused by elevated serotonin levels in the central and peripheral nervous systems. It has been well documented that the development of offspring can be affected by maternal exposure to environmental challenges, such as stress, diseases, or an unhealthy diet during pregnancy. Serotonin, also called 5-hydroxytryptamine (5-HT), is widely expressed in the female reproductive system and plays an important role in the development of follicles and embryos. However, whether the suffering of the mother from serotonin syndrome before pregnancy affects fetal development is still uncertain. In the present study, to explore the effect of maternal prepregnancy 5-HT exposure on the fetus, intraperitoneal injection of 5-HT was used to change maternal prepregnancy 5-HT levels. It was found that maternal prepregnancy 5-HT exposure significantly reduced the body weight and liver weight and the levels of estrogen and progesterone in female mice. Although there was no significant difference in the cleavage rate and blastocyst rate between the 5-HT and control groups, maternal prepregnancy 5-HT exposure increased the percentage of embryo resorption, decreased placental weight, and led to placental inflammation at E13.5. Notably, 5-HT exposure caused weight loss in the offspring at 2 weeks. These results suggested that maternal prepregnancy 5-HT exposure could affect the development of the offspring, which was partly caused by reduced hormonal secretion and placental inflammation.
Collapse
Affiliation(s)
- Yu Han
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Meng Zhang
- Academy of Translational Medicine, First Hospital, Jilin University, Changchun, China
| | - Jiahui Duan
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Leyi Li
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jinge Du
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hui Cheng
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Sheng Zhang
- Academy of Translational Medicine, First Hospital, Jilin University, Changchun, China
| | - Yanhui Zhai
- Academy of Translational Medicine, First Hospital, Jilin University, Changchun, China
| | - Xinglan An
- Academy of Translational Medicine, First Hospital, Jilin University, Changchun, China
| | - Qi Li
- Academy of Translational Medicine, First Hospital, Jilin University, Changchun, China
| | - Xueming Zhang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ziyi Li
- Academy of Translational Medicine, First Hospital, Jilin University, Changchun, China
| | - Bo Tang
- College of Veterinary Medicine, Jilin University, Changchun, China
- *Correspondence: Bo Tang,
| |
Collapse
|
15
|
Gauvrit T, Benderradji H, Buée L, Blum D, Vieau D. Early-Life Environment Influence on Late-Onset Alzheimer's Disease. Front Cell Dev Biol 2022; 10:834661. [PMID: 35252195 PMCID: PMC8891536 DOI: 10.3389/fcell.2022.834661] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/27/2022] [Indexed: 12/30/2022] Open
Abstract
With the expand of the population's average age, the incidence of neurodegenerative disorders has dramatically increased over the last decades. Alzheimer disease (AD) which is the most prevalent neurodegenerative disease is mostly sporadic and primarily characterized by cognitive deficits and neuropathological lesions such as amyloid -β (Aβ) plaques and neurofibrillary tangles composed of hyper- and/or abnormally phosphorylated Tau protein. AD is considered a complex disease that arises from the interaction between environmental and genetic factors, modulated by epigenetic mechanisms. Besides the well-described cognitive decline, AD patients also exhibit metabolic impairments. Metabolic and cognitive perturbations are indeed frequently observed in the Developmental Origin of Health and Diseases (DOHaD) field of research which proposes that environmental perturbations during the perinatal period determine the susceptibility to pathological conditions later in life. In this review, we explored the potential influence of early environmental exposure to risk factors (maternal stress, malnutrition, xenobiotics, chemical factors … ) and the involvement of epigenetic mechanisms on the programming of late-onset AD. Animal models indicate that offspring exposed to early-life stress during gestation and/or lactation increase both AD lesions, lead to defects in synaptic plasticity and finally to cognitive impairments. This long-lasting epigenetic programming could be modulated by factors such as nutriceuticals, epigenetic modifiers or psychosocial behaviour, offering thus future therapeutic opportunity to protect from AD development.
Collapse
Affiliation(s)
- Thibaut Gauvrit
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Hamza Benderradji
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Luc Buée
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Didier Vieau
- Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience and Cognition, Université de Lille, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| |
Collapse
|
16
|
McPherson NO, Vincent AD, Zander-Fox D, Grieger JA. Birthweight associations with parental obesity: retrospective analysis of 1,778 singleton term births following assisted reproductive treatment. F S Rep 2021; 2:405-412. [PMID: 34934980 PMCID: PMC8655430 DOI: 10.1016/j.xfre.2021.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/24/2021] [Accepted: 04/24/2021] [Indexed: 11/06/2022] Open
Abstract
Objective To determine the association of combined parental preconception overweight and obesity on infant birthweight. Design Retrospective study of fresh in vitro fertilization or intracytoplasmic sperm injection cycles (2009–2017). Setting Repromed, South Australia, assisted reproductive technology clinic. Patients Couples undergoing in vitro fertilization/intracytoplasmic sperm injection insemination with their own gametes and transfer of a fresh single blastocyst (N = 1,778). Intervention(s) None. Main Outcome Measures Parental body mass index (BMI) was recorded prior to cycle initiation. Infant birthweight was recorded at delivery. The impact of parental obesity and their interaction on first singleton term (≥37 weeks’ gestation) birthweight was assessed using linear regressions assessing nonlinearity and a pairwise linear interactions. Results In the base model where parental BMI is assumed linear, there was strong evidence for higher birthweight with increasing maternal BMI (11.2 g per maternal kg/m2; 95% confidence interval, 7.2, 15.1) but not paternal BMI. The inclusion of a pairwise linear interaction indicated that paternal BMI attenuates the positive association between maternal BMI and infant birthweight (interaction −0.88; 95% confidence interval, −1.49, −0.27). The inclusion of nonlinear maternal BMI terms did not change the conclusions. Conclusions Increases in the mean infant birthweight associated with maternal obesity are attenuated when the father is obese. While maternal BMI contributed more to the mean infant birthweight than paternal BMI, a couple-centered approach to preconception health advice is recommended, given the documented relationships between parental obesity and childhood weight beyond infancy. Further studies in both assisted reproductive technology and general population cohorts assessing the parental BMI interaction on infant birthweight are warranted.
Collapse
Affiliation(s)
- Nicole O McPherson
- Freemasons Center for Male Health and Wellbeing, University of Adelaide, South Australia, Australia.,Robinson Research Institute, University of Adelaide, South Australia, Australia.,Repromed, Dulwich, South Australia, Australia.,Faculty of Health and Medical Sciences, University of Adelaide, South Australia, Australia
| | - Andrew D Vincent
- Freemasons Center for Male Health and Wellbeing, University of Adelaide, South Australia, Australia.,Faculty of Health and Medical Sciences, University of Adelaide, South Australia, Australia.,South Australian Health and Medical Research Institute, South Australia, Australia
| | - Deirdre Zander-Fox
- Robinson Research Institute, University of Adelaide, South Australia, Australia.,Repromed, Dulwich, South Australia, Australia.,Monash IVF Group, Clayton, Victoria, Australia.,Faculty of Science, Monash University, Clayton, Victoria, Australia.,Future Industries Institute, University of South Australia, Mawson Lakes, South Australia, Australia
| | - Jessica A Grieger
- Robinson Research Institute, University of Adelaide, South Australia, Australia.,Faculty of Health and Medical Sciences, University of Adelaide, South Australia, Australia
| |
Collapse
|
17
|
César H, Sertorio MN, de Souza EA, Jamar G, Santamarina A, Jucá A, Casagrande BP, Pisani LP. Parental high-fat high-sugar diet programming and hypothalamus adipose tissue axis in male Wistar rats. Eur J Nutr 2021; 61:523-537. [PMID: 34657184 DOI: 10.1007/s00394-021-02690-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 09/28/2021] [Indexed: 01/04/2023]
Abstract
PURPOSE Maternal nutrition during early development and paternal nutrition pre-conception can programme offspring health status. Hypothalamus adipose axis is a target of developmental programming, and paternal and maternal high-fat, high-sugar diet (HFS) may be an important factor that predisposes offspring to develop obesity later in life. This study aims to investigate Wistar rats' maternal and paternal HFS differential contribution on the development, adiposity, and hypothalamic inflammation in male offspring from weaning until adulthood. METHODS Male progenitors were fed a control diet (CD) or HFS for 10 weeks before mating. After mating, dams were fed CD or HFS only during pregnancy and lactation. Forming the following male offspring groups: CD-maternal and paternal CD; MH-maternal HFS and paternal CD; PH-maternal CD and paternal HFS; PMH-maternal and paternal HFS. After weaning, male offspring were fed CD until adulthood. RESULTS Maternal HFS diet increased weight, visceral adiposity, and serum total cholesterol levels, and decreased hypothalamic weight in weanling male rats. In adult male offspring, maternal HFS increased weight, glucose levels, and hypothalamic NFκBp65. Paternal HFS diet lowered hypothalamic insulin receptor levels in weanling offspring and glucose and insulin levels in adult offspring. The combined effects of maternal and paternal HFS diets increased triacylglycerol, leptin levels, and hypothalamic inflammation in weanling rats, and increased visceral adiposity in adulthood. CONCLUSION Male offspring intake of CD diet after weaning reversed part of the effects of parental HFS diet during the perinatal period. However, maternal and paternal HFS diet affected adiposity and hypothalamic inflammation, which remained until adulthood.
Collapse
Affiliation(s)
- Helena César
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo-UNIFESP, Santos, SP, Brazil
| | | | - Esther Alves de Souza
- Programa de Pós-Graduação em Nutrição, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Giovana Jamar
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Aline Santamarina
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Andrea Jucá
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Breno Picin Casagrande
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Luciana Pellegrini Pisani
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil.
| |
Collapse
|
18
|
Callet T, Li H, Surget A, Terrier F, Sandres F, Lanuque A, Panserat S, Marandel L. No adverse effect of a maternal high carbohydrate diet on their offspring, in rainbow trout ( Oncorhynchus mykiss). PeerJ 2021; 9:e12102. [PMID: 34589301 PMCID: PMC8434805 DOI: 10.7717/peerj.12102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/11/2021] [Indexed: 01/09/2023] Open
Abstract
In order to develop a sustainable salmonid aquaculture, it is essential to continue to reduce the use of the protein-rich fishmeal. One promising solution to do so is the use of plant-derived carbohydrates in diet destined to broodstock. However, in mammals, the reduction of protein content (replaced by carbohydrates) in parental diet is known to have strong adverse effects on offspring phenotypes and metabolism. For the first time, the effect of a paternal and a maternal high carbohydrate-low protein diet was assessed on progeny at long term in the rainbow trout. A 30% protein diminution in both males and females broodstock diet during 10 month and 5 months, respectively, did not trigger adverse consequences on their offspring. At the molecular level, offspring transcriptomes were not significantly altered, emphasizing no effect on metabolism. Tenuous differences in the biochemical composition of the liver and the viscera were observed. The recorded effects remained in the normal range of value and accordingly offspring growth were not negatively affected over the long term. Overall, we demonstrated here that a 30% protein diminution during gametogenesis is feasible, confirming the possibility to increase the proportion of plant-derived carbohydrates in female broodstock diets to replace fishmeal proteins.
Collapse
Affiliation(s)
- Therese Callet
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Hongyan Li
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France.,State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Anne Surget
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Frederic Terrier
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Franck Sandres
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Anthony Lanuque
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Stephane Panserat
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Lucie Marandel
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| |
Collapse
|
19
|
Rutkowska J, Lagisz M, Bonduriansky R, Nakagawa S. Mapping the past, present and future research landscape of paternal effects. BMC Biol 2020; 18:183. [PMID: 33246472 PMCID: PMC7694421 DOI: 10.1186/s12915-020-00892-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Although in all sexually reproducing organisms an individual has a mother and a father, non-genetic inheritance has been predominantly studied in mothers. Paternal effects have been far less frequently studied, until recently. In the last 5 years, research on environmentally induced paternal effects has grown rapidly in the number of publications and diversity of topics. Here, we provide an overview of this field using synthesis of evidence (systematic map) and influence (bibliometric analyses). RESULTS We find that motivations for studies into paternal effects are diverse. For example, from the ecological and evolutionary perspective, paternal effects are of interest as facilitators of response to environmental change and mediators of extended heredity. Medical researchers track how paternal pre-fertilization exposures to factors, such as diet or trauma, influence offspring health. Toxicologists look at the effects of toxins. We compare how these three research guilds design experiments in relation to objects of their studies: fathers, mothers and offspring. We highlight examples of research gaps, which, in turn, lead to future avenues of research. CONCLUSIONS The literature on paternal effects is large and disparate. Our study helps in fostering connections between areas of knowledge that develop in parallel, but which could benefit from the lateral transfer of concepts and methods.
Collapse
Affiliation(s)
- Joanna Rutkowska
- Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, Kraków, Poland
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| | - Malgorzata Lagisz
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| | - Russell Bonduriansky
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| | - Shinichi Nakagawa
- Evolution & Ecology Research Centre, School of Biological, Earth and Environmental Sciences, BEES, The University of New South Wales, Sydney, Australia
| |
Collapse
|
20
|
Monarca RI, Speakman JR, Mathias ML. Effects of predation risk on the body mass regulation of growing wood mice. J Zool (1987) 2020. [DOI: 10.1111/jzo.12811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- R. I. Monarca
- CESAM – Center for Environmental and Marine Studies Universidade de Aveiro Aveiro Portugal
- Departamento de Biologia Animal Faculdade de Ciências da Universidade de Lisboa Lisbon Portugal
| | - J. R. Speakman
- Institute of Biological and Environmental Sciences University of Aberdeen Aberdeen UK
- State Key Laboratory of Molecular Developmental Biology Institute of Genetics and Developmental Biology Chinese Academy of Sciences Beijing China
| | - M. L. Mathias
- CESAM – Center for Environmental and Marine Studies Universidade de Aveiro Aveiro Portugal
- Departamento de Biologia Animal Faculdade de Ciências da Universidade de Lisboa Lisbon Portugal
| |
Collapse
|
21
|
Gyllenhammer LE, Entringer S, Buss C, Wadhwa PD. Developmental programming of mitochondrial biology: a conceptual framework and review. Proc Biol Sci 2020; 287:20192713. [PMID: 32345161 PMCID: PMC7282904 DOI: 10.1098/rspb.2019.2713] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Research on mechanisms underlying the phenomenon of developmental programming of health and disease has focused primarily on processes that are specific to cell types, organs and phenotypes of interest. However, the observation that exposure to suboptimal or adverse developmental conditions concomitantly influences a broad range of phenotypes suggests that these exposures may additionally exert effects through cellular mechanisms that are common, or shared, across these different cell and tissue types. It is in this context that we focus on cellular bioenergetics and propose that mitochondria, bioenergetic and signalling organelles, may represent a key cellular target underlying developmental programming. In this review, we discuss empirical findings in animals and humans that suggest that key structural and functional features of mitochondrial biology exhibit developmental plasticity, and are influenced by the same physiological pathways that are implicated in susceptibility for complex, common age-related disorders, and that these targets of mitochondrial developmental programming exhibit long-term temporal stability. We conclude by articulating current knowledge gaps and propose future research directions to bridge these gaps.
Collapse
Affiliation(s)
- Lauren E Gyllenhammer
- Development, Health and Disease Research Program, School of Medicine, Irvine, CA, USA.,Department of Pediatrics, School of Medicine, Irvine, CA, USA
| | - Sonja Entringer
- Development, Health and Disease Research Program, School of Medicine, Irvine, CA, USA.,Department of Pediatrics, School of Medicine, Irvine, CA, USA.,Charité-Universitätsmedizin Berlin, Institute of Medical Psychology, Berlin, Germany
| | - Claudia Buss
- Development, Health and Disease Research Program, School of Medicine, Irvine, CA, USA.,Department of Pediatrics, School of Medicine, Irvine, CA, USA.,Charité-Universitätsmedizin Berlin, Institute of Medical Psychology, Berlin, Germany
| | - Pathik D Wadhwa
- Development, Health and Disease Research Program, School of Medicine, Irvine, CA, USA.,Department of Pediatrics, School of Medicine, Irvine, CA, USA.,Department of Psychiatry and Human Behaviour, School of Medicine, Irvine, CA, USA.,Department of Obstetrics and Gynecology, School of Medicine, Irvine, CA, USA.,Department of Epidemiology, University of California, School of Medicine, Irvine, CA, USA
| |
Collapse
|
22
|
Nicholas LM, Ozanne SE. Early life programming in mice by maternal overnutrition: mechanistic insights and interventional approaches. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180116. [PMID: 30966886 DOI: 10.1098/rstb.2018.0116] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Animal models have been indispensable in elucidating the potential causative mechanisms underlying the effects of maternal diet on offspring health. Of these, the mouse has been widely used to model maternal overnutrition and/or maternal obesity and to study its effects across one or more generations. This review discusses recent findings from mouse models, which resemble the human situation, i.e. overnutrition/obesity across pregnancy and lactation. It also highlights the importance of embryo transfer models in identifying critical developmental period(s) during which specific metabolic changes are programmed in the offspring. The mouse is also an excellent tool for maternal intervention studies aimed at elucidating the longer-term effects on the offspring and for defining possible maternal factors underling the programming of metabolic adversity in offspring. While knowledge of the mouse genome and the molecular tools available have allowed great progress to be made in the field, it is clear that we need to define if the effects on the offspring are mediated by maternal obesity per se or if specific components of the maternal metabolic environment are more important. We can then begin to identify at-risk offspring and to design more effective interventions for the mother and/or her child. This article is part of the theme issue 'Developing differences: early-life effects and evolutionary medicine'.
Collapse
Affiliation(s)
- Lisa M Nicholas
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge , Addenbrooke's Hospital, Cambridge , UK
| | - Susan E Ozanne
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge , Addenbrooke's Hospital, Cambridge , UK
| |
Collapse
|
23
|
Yun Y, Wei Z, Hunter N. Maternal obesity enhances oocyte chromosome abnormalities associated with aging. Chromosoma 2019; 128:413-421. [PMID: 31286204 DOI: 10.1007/s00412-019-00716-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 05/22/2019] [Accepted: 06/17/2019] [Indexed: 12/18/2022]
Abstract
Obesity is increasing globally, and maternal obesity has adverse effects on pregnancy outcomes and the long-term health of offspring. Maternal obesity has been associated with pregnancy failure through impaired oogenesis and embryogenesis. However, whether maternal obesity causes chromosome abnormalities in oocytes has remained unclear. Here we show that chromosome abnormalities are increased in the oocytes of obese mice fed a high-fat diet and identify weakened sister-chromatid cohesion as the likely cause. Numbers of full-grown follicles retrieved from obese mice were the same as controls and the efficiency of in vitro oocyte maturation remained high. However, chromosome abnormalities presenting in both metaphase-I and metaphase-II were elevated, most prominently the premature separation of sister chromatids. Weakened sister-chromatid cohesion in oocytes from obese mice was manifested both as the terminalization of chiasmata in metaphase-I and as increased separation of sister centromeres in metaphase II. Obesity-associated abnormalities were elevated in older mice implying that maternal obesity exacerbates the deterioration of cohesion seen with advancing age.
Collapse
Affiliation(s)
- Yan Yun
- Howard Hughes Medical Institute, University of California, Davis, Davis, CA, USA.,Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, CA, USA
| | - Zijie Wei
- Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, CA, USA
| | - Neil Hunter
- Howard Hughes Medical Institute, University of California, Davis, Davis, CA, USA. .,Department of Microbiology & Molecular Genetics, University of California, Davis, Davis, CA, USA. .,Department of Molecular & Cellular Biology, University of California, Davis, Davis, CA, USA. .,Department of Cell Biology & Human Anatomy, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
24
|
Crean AJ, Senior AM. High-fat diets reduce male reproductive success in animal models: A systematic review and meta-analysis. Obes Rev 2019; 20:921-933. [PMID: 30756459 DOI: 10.1111/obr.12827] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/13/2018] [Accepted: 11/27/2018] [Indexed: 12/11/2022]
Abstract
Sperm counts have shown a progressive decline across the world since the mid-1900s. Global rates of obesity have been climbing at a similarly alarming rate, suggesting that these two factors may be linked. However, studies examining the relationship between body mass index (BMI) and male fertility have produced conflicting results. These discrepancies among studies are commonly attributed to limitations that are inherent in human studies such as correlational data and confounding factors-limitations that are absent from animal studies. Hence, we conducted a systematic review and meta-analysis of animal studies that experimentally induced obesity with a high-fat diet and measured impacts on sperm traits and/or male fertility. Overall, we found strong, consistent evidence in animal studies that high-fat diets are detrimental for male fertility. Compared with controls, males fed a high-fat diet had smaller testes and sex accessory glands relative to body size, reduced semen quality, reduced mating success, and reduced fertilization success. This is the first time that experimental data of high-fat diet effects on male reproduction have been synthesized in a meta-analysis, and thus, our results provide novel insight to the complex question of how dietary-induced obesity affects male fertility. The strong negative effects detected in animal models suggest that the inconsistent results plaguing human studies are an artefact of limitations inherent in correlational studies rather than a true effect.
Collapse
Affiliation(s)
- Angela J Crean
- Sydney School of Veterinary Science, The University of Sydney, Sydney, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney, Sydney, Australia.,School of Mathematics and Statistics, The University of Sydney, Sydney, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| |
Collapse
|
25
|
da Cruz RS, Clarke J, Curi ACP, Al-Yawar A, Jin L, Baird A, Cruz MI, Kallakury B, de Assis S. Parental obesity programs pancreatic cancer development in offspring. Endocr Relat Cancer 2019; 26:511-523. [PMID: 30865925 PMCID: PMC6717698 DOI: 10.1530/erc-19-0016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022]
Abstract
Epidemiological studies suggest that timing of obesity onset - and underlying metabolic dysfunction - is important in determining pancreatic cancer rates: early and young adult abdominal overweight/obesity is more strongly associated with this cancer than obesity that develops later in life. Parental obesity and overweight are associated with metabolic dysfunction and obesity in their children. Here, we evaluated the impact of parental overweight on offspring's susceptibility of pancreatic cancer using the P48Cre/+/KrasG12D/+ mouse model. Male mice were fed an obesity-inducing diet (OID) before conception and mated with females raised on a control diet (CO) to generate the offspring. In a separate experiment, pregnant dams were fed CO or OID throughout gestation. The resulting OID offspring from the maternal (OID-m) or paternal lineage (OID-p) were used to study body weight, metabolic parameters and pancreatic cancer development and for molecular analysis. Parental obesity increased offspring's body weight at birth, weaning and in adulthood compared to CO, with gender- and genotype-specific differences. OID-p and OID-m offspring showed metabolic disorder and accelerated development of high-grade PanIN/PDAC. OID offspring also had higher rates of acinar-to-ductal reprogramming assessed by CPA1+/SOX9+-positive pancreatic cells. Levels of Tenascin C (TNC), an ECM glycoprotein shown to suppress apoptosis, were elevated in OID offspring, particularly females. In line with that, OID offspring displayed increased collagen content and decreased apoptosis in pancreatic lesions compared to CO. An ancestral history of obesity through either the paternal or maternal lineages increases offspring's susceptibility to pancreatic cancer development.
Collapse
|
26
|
Raad G, Azouri J, Rizk K, Zeidan NS, Azouri J, Grandjean V, Hazzouri M. Adverse effects of paternal obesity on the motile spermatozoa quality. PLoS One 2019; 14:e0211837. [PMID: 30742661 PMCID: PMC6370200 DOI: 10.1371/journal.pone.0211837] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022] Open
Abstract
Growing evidence suggests that paternal obesity may decrease male fertility potential. During infertility treatment with intra-cytoplasmic sperm injection (ICSI), a morphologically normal motile spermatozoon is injected into a mature egg, when possible. However, sperm motility and morphology per se do not reflect the sperm molecular composition. In this study, we aimed to assess the quality of motile spermatozoa in the context of obesity by analysing their conventional and molecular characteristics as well as their ability to promote early embryonic development. A prospective study was conducted on 128 infertile men divided into three groups: 40 lean, 42 overweight, and 46 obese men. Conventional sperm parameters (concentration, motility and morphology) and sperm molecular status (chromatin composition and integrity, 5-methycytosine (5-mC) and 5-hydroxycytosine (5-hmC) contents and oxidative stress level) were analysed on raw semen and/or on motile spermatozoa selected by density gradient or swim-up techniques. Morphokinetic analysis of the embryos derived from ICSI was performed using the Embryoviewer software. Our results showed that the motile sperm-enriched fraction from obese men exhibited higher levels of retained histones (p<0.001), elevated percentage of altered chromatin integrity (p<0.001), and decreased contents of 5-hmC (p<0.001), and 5-mC (p<0.05) levels as compared to that from lean men. Importantly, there were no statistically significant correlations between these molecular parameters and the percentages of morphologically normal motile spermatozoa. Regarding embryo morphokinetics, the CC1 (p<0.05) and CC3 (p<0.05) embryonic cell cycles were significantly delayed in the cleavage embryos of the obese group as compared to the embryos of the lean group. Our data is of particular interest because, besides demonstrating the negative impacts of obesity on motile spermatozoa molecular composition, it also highlights the possible risk of disturbing early embryonic cell cycles kinetics in the context of paternal obesity.
Collapse
Affiliation(s)
- Georges Raad
- Azoury-IVF clinic, Mount Lebanon Hospital, Camille Chamoun Boulevard, Beirut, Lebanon
- Lebanese University, Faculty of Sciences 2, Fanar, Lebanon
| | - Joseph Azouri
- Azoury-IVF clinic, Mount Lebanon Hospital, Camille Chamoun Boulevard, Beirut, Lebanon
| | - Kamal Rizk
- Azoury-IVF clinic, Mount Lebanon Hospital, Camille Chamoun Boulevard, Beirut, Lebanon
| | - Nina S. Zeidan
- Lebanese University, Faculty of Sciences 2, Fanar, Lebanon
| | - Jessica Azouri
- Azoury-IVF clinic, Mount Lebanon Hospital, Camille Chamoun Boulevard, Beirut, Lebanon
| | - Valérie Grandjean
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 10 “Control of gene expression “, Nice, France and University of Nice Sophia Antipolis, Faculty of Medecine, Nice, France
- * E-mail:
| | - Mira Hazzouri
- Lebanese University, Faculty of Sciences 2, Fanar, Lebanon
| |
Collapse
|
27
|
Portha B, Grandjean V, Movassat J. Mother or Father: Who Is in the Front Line? Mechanisms Underlying the Non-Genomic Transmission of Obesity/Diabetes via the Maternal or the Paternal Line. Nutrients 2019; 11:E233. [PMID: 30678214 PMCID: PMC6413176 DOI: 10.3390/nu11020233] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/01/2019] [Accepted: 01/09/2019] [Indexed: 02/06/2023] Open
Abstract
Extensive epidemiological and experimental evidence have shown that exposure to an adverse intrauterine environment as observed in offspring of pregnancies complicated by obesity or diabetes, can program susceptibility to metabolic, endocrine and cardiovascular disorders later in life. Although most studies have concentrated on the maternal environment, it is also becoming evident that paternal exposure to obesity or diabetes can result in the later development of metabolic disorders in the offspring. Such programmed effects might not be limited to the first directly exposed generation, but could be transmitted to subsequent generations. This suggests the existence of mechanisms by which metabolic changes in parental phenotype are transmissible to offspring. The mechanisms which underpin the transmission of the programmed effects across generations are still unclear. However, epigenetic regulation of transcription has emerged as a strong candidate for mediating the heritability of metabolic diseases. Here, we review the most relevant evidence from human and animal studies showing transmission of programming effects of obesity or diabetes across generations, and the current mechanisms underlying either maternal or paternal influences on the metabolic status of offspring.
Collapse
Affiliation(s)
- Bernard Portha
- Sorbonne-Paris-Cité, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptative), Université Paris-Diderot, CNRS UMR 8251, F-75205 Paris CEDEX 13, France.
| | - Valérie Grandjean
- Inserm U1065 C3M, Team Control of Gene Expression (10), Université Côte d'Azur, 151 Route de Ginestière, 06204 Nice CEDEX 3, France.
| | - Jamileh Movassat
- Sorbonne-Paris-Cité, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptative), Université Paris-Diderot, CNRS UMR 8251, F-75205 Paris CEDEX 13, France.
| |
Collapse
|
28
|
Bromfield EG, Dowland SN, Dunleavy JEM, Dunning KR, Holland OJ, Houston BJ, Pankhurst MW, Richani D, Riepsamen AH, Rose R, Bertoldo MJ. Fifty years of reproductive biology in Australia: highlights from the 50th Annual Meeting of the Society for Reproductive Biology (SRB). Reprod Fertil Dev 2019; 31:829-836. [PMID: 30636191 DOI: 10.1071/rd18436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/03/2018] [Indexed: 11/23/2022] Open
Abstract
The 2018 edition of the Society for Reproductive Biology's (SRB) Annual Meeting was a celebration of 50 years of Australian research into reproductive biology. The past 50 years has seen many important contributions to this field, and these advances have led to changes in practice and policy, improvements in the efficiency of animal reproduction and improved health outcomes. This conference review delivers a dedicated summary of the symposia, discussing emerging concepts, raising new questions and proposing directions forward. Notably, the symposia discussed in this review emphasised the impact that reproductive research can have on quality of life and the health trajectories of individuals. The breadth of the research discussed encompasses the central regulation of fertility and cyclicity, life course health and how the environment of gametes and embryos can affect subsequent generations, significant advances in our understanding of placental biology and pregnancy disorders and the implications of assisted reproductive technologies on population health. The importance of a reliable food supply and protection of endangered species is also discussed. The research covered at SRB's 2018 meeting not only recognised the important contributions of its members over the past 50 years, but also highlighted key findings and avenues for innovation moving forward that will enable the SRB to continue making significant contributions for the next 50 years.
Collapse
Affiliation(s)
- Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, NSW 2308, Australia
| | - Samson N Dowland
- School of Medical Sciences (Discipline of Anatomy and Histology) and The Bosch Institute, F13 Anderson Stuart Building, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Kylie R Dunning
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, SA 5005, Australia
| | - Olivia J Holland
- School of Medical Science, Griffith University, Gold Coast Campus, Southport, Qld 5005, Australia
| | - Brendan J Houston
- School of Biological Sciences, Monash University, Clayton, Vic. 3168, Australia
| | - Michael W Pankhurst
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Dulama Richani
- Fertility and Research Centre, School of Women's and Children's Health, UNSW, Randwick 2052, NSW 4222, Australia
| | - Angelique H Riepsamen
- Fertility and Research Centre, School of Women's and Children's Health, UNSW, Randwick 2052, NSW 4222, Australia
| | - Ryan Rose
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Michael J Bertoldo
- Fertility and Research Centre, School of Women's and Children's Health, UNSW, Randwick 2052, NSW 4222, Australia
| |
Collapse
|
29
|
Perez MF, Lehner B. Intergenerational and transgenerational epigenetic inheritance in animals. Nat Cell Biol 2019; 21:143-151. [PMID: 30602724 DOI: 10.1038/s41556-018-0242-9] [Citation(s) in RCA: 300] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Animals transmit not only DNA but also other molecules, such as RNA, proteins and metabolites, to their progeny via gametes. It is currently unclear to what extent these molecules convey information between generations and whether this information changes according to their physiological state and environment. Here, we review recent work on the molecular mechanisms by which 'epigenetic' information is transmitted between generations over different timescales, and the importance of this information for development and physiology.
Collapse
Affiliation(s)
- Marcos Francisco Perez
- Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Ben Lehner
- Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain. .,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
30
|
Norman RJ, Mol BWJ. Successful weight loss interventions before in vitro fertilization: fat chance? Fertil Steril 2018; 110:581-586. [DOI: 10.1016/j.fertnstert.2018.05.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 05/23/2018] [Indexed: 12/17/2022]
|
31
|
Cardenas-Perez RE, Fuentes-Mera L, de la Garza AL, Torre-Villalvazo I, Reyes-Castro LA, Rodriguez-Rocha H, Garcia-Garcia A, Corona-Castillo JC, Tovar AR, Zambrano E, Ortiz-Lopez R, Saville J, Fuller M, Camacho A. Maternal overnutrition by hypercaloric diets programs hypothalamic mitochondrial fusion and metabolic dysfunction in rat male offspring. Nutr Metab (Lond) 2018; 15:38. [PMID: 29991958 PMCID: PMC5987395 DOI: 10.1186/s12986-018-0279-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
Abstract
Background Maternal overnutrition including pre-pregnancy, pregnancy and lactation promotes a lipotoxic insult leading to metabolic dysfunction in offspring. Diet-induced obesity models (DIO) show that changes in hypothalamic mitochondria fusion and fission dynamics modulate metabolic dysfunction. Using three selective diet formula including a High fat diet (HFD), Cafeteria (CAF) and High Sugar Diet (HSD), we hypothesized that maternal diets exposure program leads to selective changes in hypothalamic mitochondria fusion and fission dynamics in male offspring leading to metabolic dysfunction which is exacerbated by a second exposure after weaning. Methods We exposed female Wistar rats to nutritional programming including Chow, HFD, CAF, or HSD for 9 weeks (pre-mating, mating, pregnancy and lactation) or to the same diets to offspring after weaning. We determined body weight, food intake and metabolic parameters in the offspring from 21 to 60 days old. Hypothalamus was dissected at 60 days old to determine mitochondria-ER interaction markers by mRNA expression and western blot and morphology by transmission electron microscopy (TEM). Mitochondrial-ER function was analyzed by confocal microscopy using hypothalamic cell line mHypoA-CLU192. Results Maternal programming by HFD and CAF leads to failure in glucose, leptin and insulin sensitivity and fat accumulation. Additionally, HFD and CAF programming promote mitochondrial fusion by increasing the expression of MFN2 and decreasing DRP1, respectively. Further, TEM analysis confirms that CAF exposure after programing leads to an increase in mitochondria fusion and enhanced mitochondrial-ER interaction, which partially correlates with metabolic dysfunction and fat accumulation in the HFD and CAF groups. Finally, we identified that lipotoxic palmitic acid stimulus in hypothalamic cells increases Ca2+ overload into mitochondria matrix leading to mitochondrial dysfunction. Conclusions We concluded that maternal programming by HFD induces hypothalamic mitochondria fusion, metabolic dysfunction and fat accumulation in male offspring, which is exacerbated by HFD or CAF exposure after weaning, potentially due to mitochondria calcium overflux.
Collapse
Affiliation(s)
- Robbi E Cardenas-Perez
- 1Departmento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico.,2Unidad de Neurometabolismo, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autónoma de Nuevo Leon, Monterrey, Mexico
| | - Lizeth Fuentes-Mera
- 1Departmento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Ana Laura de la Garza
- 3Centro de Investigacion en Nutricion y Salud Publica, Facultad de Salud Publica y Nutricion, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Ivan Torre-Villalvazo
- 4Departamento Fisiología de la Nutrición, Instituto Nacional de Ciencias Medicas y Nutrición, Mexico City, Mexico
| | - Luis A Reyes-Castro
- 5Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Medicas y Nutrición Salvador Zubiran, México City, Mexico
| | - Humberto Rodriguez-Rocha
- 6Departmento de Histología, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | - Aracely Garcia-Garcia
- 6Departmento de Histología, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico
| | | | - Armando R Tovar
- 4Departamento Fisiología de la Nutrición, Instituto Nacional de Ciencias Medicas y Nutrición, Mexico City, Mexico
| | - Elena Zambrano
- 5Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Medicas y Nutrición Salvador Zubiran, México City, Mexico
| | - Rocio Ortiz-Lopez
- 8Escuela de Medicina y Ciencias de la Salud, Instituto Tecnologico de Monterrey, Monterrey, Mexico
| | - Jennifer Saville
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, University of Adelaide, Adelaide, Australia
| | - Maria Fuller
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, University of Adelaide, Adelaide, Australia
| | - Alberto Camacho
- 1Departmento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico.,2Unidad de Neurometabolismo, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autónoma de Nuevo Leon, Monterrey, Mexico.,10Departamento de Bioquimica y Medicina Molecular. Facultad de Medicina, Universidad Autónoma de Nuevo León, Ave. Francisco I Madero y Dr. Eduardo Aguirre Pequeño s/n. Colonia Mitras Centro, C.P. 64460 Monterrey, Nuevo Leon Mexico
| |
Collapse
|
32
|
Krout D, Roemmich JN, Bundy A, Garcia RA, Yan L, Claycombe-Larson KJ. Paternal exercise protects mouse offspring from high-fat-diet-induced type 2 diabetes risk by increasing skeletal muscle insulin signaling. J Nutr Biochem 2018; 57:35-44. [PMID: 29669306 DOI: 10.1016/j.jnutbio.2018.03.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 02/06/2023]
Abstract
Paternal obesity increases, while paternal exercise decreases, offspring obesity and type 2 diabetes (T2D) risk; however, no studies have determined whether a paternal high-fat (HF) diet and exercise interact to alter offspring body weight (BW), adiposity and T2D risk. Three-week-old male C57BL/6 mice were fed a normal-fat (NF) diet (16% fat) or an HF diet (45% fat) and assigned to either voluntary wheel running exercise or cage activity for 3 months prior to mating with NF-diet-fed dams. After weaning, male offspring were fed an NF or HF diet for an additional 3 months. F1 male mice whose fathers ate an HF diet had decreased % body fat accompanied by decreased gene expression of beige adipocyte marker FGF21. However, paternal HF-diet-induced reductions in F1 offspring % body fat normalized but did not reduce T2D risk. Exercise was protective against paternal HF-diet-induced insulin resistance by increasing the expression of insulin signaling (GLUT4, IRS1 and PI3K) markers in skeletal muscle resulting in normal T2D risk. When fathers were fed an HF diet and exercised, a postnatal HF diet increased beiging (PPARγ). Thus, these findings show that increases in T2D risk in male offspring when the father consumes an HF diet can be normalized when the father also exercises preconception and that this protection may occur by increases in insulin signaling potential within offspring skeletal muscle. Future studies should further determine the physiological mechanism(s) underlying the beneficial effects of exercise through the paternal lineage.
Collapse
Affiliation(s)
- Danielle Krout
- U.S. Department of Agriculture Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - James N Roemmich
- U.S. Department of Agriculture Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - Amy Bundy
- U.S. Department of Agriculture Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - Rolando A Garcia
- U.S. Department of Agriculture Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - Lin Yan
- U.S. Department of Agriculture Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA
| | - Kate J Claycombe-Larson
- U.S. Department of Agriculture Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA.
| |
Collapse
|
33
|
Embryonic defects induced by maternal obesity in mice derive from Stella insufficiency in oocytes. Nat Genet 2018; 50:432-442. [DOI: 10.1038/s41588-018-0055-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 01/03/2018] [Indexed: 12/18/2022]
|
34
|
Raad G, Hazzouri M, Bottini S, Trabucchi M, Azoury J, Grandjean V. Paternal obesity: how bad is it for sperm quality and progeny health? Basic Clin Androl 2017; 27:20. [PMID: 29123667 PMCID: PMC5657098 DOI: 10.1186/s12610-017-0064-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
There is substantial evidence that paternal obesity is associated not only with an increased incidence of infertility, but also with an increased risk of metabolic disturbance in adult offspring. Apparently, several mechanisms may contribute to the sperm quality alterations associated with paternal obesity, such as physiological/hormonal alterations, oxidative stress, and epigenetic alterations. Along these lines, modifications of hormonal profiles namely reduced androgen levels and elevated estrogen levels, were found associated with lower sperm concentration and seminal volume. Additionally, oxidative stress in testis may induce an increase of the percentage of sperm with DNA fragmentation. The latter, relate to other peculiarities such as alteration of the embryonic development, increased risk of miscarriage, and development of chronic morbidity in the offspring, including childhood cancers. Undoubtedly, epigenetic alterations (ie, DNA methylation, chromatin modifications, and small RNA deregulation) of sperm related to paternal obesity and their consequences on the progeny are poorly understood determinants of paternal obesity-induced transmission. In this review, we summarize and discuss the data available in the literature regarding the biological, physiological, and molecular consequences of paternal obesity on male fertility potential and ultimately progeny health.
Collapse
Affiliation(s)
- Georges Raad
- Azoury-IVF clinic, Mount Lebanon Hospital, 5th floor, Camille Chamoun bvd, Beirut, Lebanon.,Université Cote d'Azur, Inserm, C3M (team 10), Nice, France
| | - Mira Hazzouri
- Faculty of Sciences 2, Lebanese University, Fanar, Lebanon
| | - Silvia Bottini
- Université Cote d'Azur, Inserm, C3M (team 10), Nice, France
| | | | - Joseph Azoury
- Azoury-IVF clinic, Mount Lebanon Hospital, 5th floor, Camille Chamoun bvd, Beirut, Lebanon
| | | |
Collapse
|
35
|
Abstract
Early - intrauterine - environmental factors are linked to the development of cardiovascular disease in later life. Traditionally, these factors are considered to be maternal factors such as maternal under and overnutrition, exposure to toxins, lack of micronutrients, and stress during pregnancy. However, in the recent years, it became obvious that also paternal environmental factors before conception and during sperm development determine the health of the offspring in later life. We will first describe clinical observational studies providing evidence for paternal programming of adulthood diseases in progeny. Next, we describe key animal studies proving this relationship, followed by a detailed analysis of our current understanding of the underlying molecular mechanisms of paternal programming. Alterations of noncoding sperm micro-RNAs, histone acetylation, and targeted as well as global DNA methylation seem to be in particular involved in paternal programming of offspring's diseases in later life.
Collapse
|
36
|
Schwartz MW, Seeley RJ, Zeltser LM, Drewnowski A, Ravussin E, Redman LM, Leibel RL. Obesity Pathogenesis: An Endocrine Society Scientific Statement. Endocr Rev 2017; 38:267-296. [PMID: 28898979 PMCID: PMC5546881 DOI: 10.1210/er.2017-00111] [Citation(s) in RCA: 386] [Impact Index Per Article: 55.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 02/07/2023]
Abstract
Obesity is among the most common and costly chronic disorders worldwide. Estimates suggest that in the United States obesity affects one-third of adults, accounts for up to one-third of total mortality, is concentrated among lower income groups, and increasingly affects children as well as adults. A lack of effective options for long-term weight reduction magnifies the enormity of this problem; individuals who successfully complete behavioral and dietary weight-loss programs eventually regain most of the lost weight. We included evidence from basic science, clinical, and epidemiological literature to assess current knowledge regarding mechanisms underlying excess body-fat accumulation, the biological defense of excess fat mass, and the tendency for lost weight to be regained. A major area of emphasis is the science of energy homeostasis, the biological process that maintains weight stability by actively matching energy intake to energy expenditure over time. Growing evidence suggests that obesity is a disorder of the energy homeostasis system, rather than simply arising from the passive accumulation of excess weight. We need to elucidate the mechanisms underlying this "upward setting" or "resetting" of the defended level of body-fat mass, whether inherited or acquired. The ongoing study of how genetic, developmental, and environmental forces affect the energy homeostasis system will help us better understand these mechanisms and are therefore a major focus of this statement. The scientific goal is to elucidate obesity pathogenesis so as to better inform treatment, public policy, advocacy, and awareness of obesity in ways that ultimately diminish its public health and economic consequences.
Collapse
Affiliation(s)
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109
| | - Lori M Zeltser
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, New York 10032
| | - Adam Drewnowski
- Center for Public Health Nutrition, University of Washington, Seattle, Washington 98195
| | - Eric Ravussin
- John S. McIlhenny Skeletal Muscle Physiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808
| | - Leanne M Redman
- John S. McIlhenny Skeletal Muscle Physiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808
| | - Rudolph L Leibel
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, New York 10032.,Division of Molecular Genetics, Department of Pediatrics, Columbia University, New York, New York 10032
| |
Collapse
|
37
|
Abiad F, Awwad J, Abbas HA, Zebian D, Ghazeeri G. Management of weight loss in obesity-associated male infertility: a spotlight on bariatric surgery. HUM FERTIL 2017; 20:227-235. [PMID: 28460579 DOI: 10.1080/14647273.2017.1317369] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In recent years, there has been a paralleled increase between male obesity and infertility rates. Obesity is associated with impaired hypothalamic-pituitary-gonadal axis, aberrant semen parameters, and subfertility or infertility. Weight loss is strongly recommended for the management of obesity-associated infertility. Lifestyle modifications that include caloric restriction and increased physical activity have a short-lived impact. Bariatric surgery is a better and more durable weight loss alternative. Comprehensive information about the benefits of weight loss on obesity-associated male infertility following bariatric surgery is still emerging. In this review, we discuss the hormonal, physical and environmental mechanism contributing to obesity-associated infertility. We then assess weight loss approaches, which include lifestyle modification, medical and surgical approaches, that can improve fertility in obese men. This review focuses also on bariatric surgery for the management of obese men seeking fertility treatment. Anecdotal evidence suggesting that bariatric surgery can impair fertility is also discussed.
Collapse
Affiliation(s)
- Firass Abiad
- a Department of General Surgery , American University of Beirut Medical Center , Beirut , Lebanon
| | - Johnny Awwad
- b Department of Obstetrics and Gynecology , American University of Beirut Medical Center , Beirut , Lebanon
| | - Hussein A Abbas
- b Department of Obstetrics and Gynecology , American University of Beirut Medical Center , Beirut , Lebanon
| | - Dina Zebian
- c Department of Emergency Medicine , American University of Beirut , Beirut , Lebanon
| | - Ghina Ghazeeri
- b Department of Obstetrics and Gynecology , American University of Beirut Medical Center , Beirut , Lebanon
| |
Collapse
|
38
|
Wallace JM, Milne JS, Adam CL, Aitken RP. Impact of donor and recipient adiposity on placental and fetal growth in adolescent sheep. Reproduction 2017; 153:381-394. [DOI: 10.1530/rep-16-0590] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/09/2016] [Accepted: 01/09/2017] [Indexed: 01/04/2023]
Abstract
The influence of maternal obesity during oocyte development and its putative interaction with nutrient reserves at conception on pregnancy outcome were examined in an adolescent sheep model. Donor ewes were nutritionally managed to achieve contrasting adiposity (control (CD)/obese (ObD)) for 6 weeks prior to superovulation and inseminated by a non-obese sire. Morulae from 6 CD and 7 ObD were transferred in singleton into adolescent recipients of identical age but differing adiposity, classified as relatively fat or thin respectively. Thereafter, all were overnourished to promote rapid growth/adiposity (2 × 2 design, 13/14 pregnancies/group). A fifth recipient group of intermediate adiposity received embryos from another 5 CD, was offered a moderate intake to maintain adiposity throughout gestation and acted as controls for normal pregnancy outcome (optimally treated control (OTC), 19 pregnancies). Donor obesity did not influence ovulation, fertilisation or recovery rates or impact embryo morphology. Gestation length and colostrum yield were unaffected by donor or recipient adiposity and were reduced relative to OTC. Total fetal cotyledon and lamb birth weights were independent of initial donor adiposity but reduced in relatively thin vs relatively fat recipients and lower than those in the OTC group. In spite of high placental efficiency, the incidence of fetal growth restriction was greatest in the thin recipients. Thus, maternal adiposity at conception, but not pre-conception maternal obesity, modestly influences the feto-placental growth trajectory, whereas comparison with the OTC indicates that high gestational intakes to promote rapid maternal growth remain the dominant negative influence on pregnancy outcome in young adolescents. These findings inform dietary advice for pregnant adolescent girls.
Collapse
|
39
|
Yeung EH, Sundaram R, Ghassabian A, Xie Y, Buck Louis G. Parental Obesity and Early Childhood Development. Pediatrics 2017; 139:peds.2016-1459. [PMID: 28044047 PMCID: PMC5260147 DOI: 10.1542/peds.2016-1459] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/03/2016] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Previous studies identified associations between maternal obesity and childhood neurodevelopment, but few examined paternal obesity despite potentially distinct genetic/epigenetic effects related to developmental programming. METHODS Upstate KIDS (2008-2010) recruited mothers from New York State (excluding New York City) at ∼4 months postpartum. Parents completed the Ages and Stages Questionnaire (ASQ) when their children were 4, 8, 12, 18, 24, 30, and 36 months of age corrected for gestation. The ASQ is validated to screen for delays in 5 developmental domains (ie, fine motor, gross motor, communication, personal-social functioning, and problem-solving ability). Analyses included 3759 singletons and 1062 nonrelated twins with ≥1 ASQs returned. Adjusted odds ratios (aORs) and 95% confidence intervals were estimated by using generalized linear mixed models accounting for maternal covariates (ie, age, race, education, insurance, marital status, parity, and pregnancy smoking). RESULTS Compared with normal/underweight mothers (BMI <25), children of obese mothers (26% with BMI ≥30) had increased odds of failing the fine motor domain (aOR 1.67; confidence interval 1.12-2.47). The association remained after additional adjustment for paternal BMI (1.67; 1.11-2.52). Paternal obesity (29%) was associated with increased risk of failing the personal-social domain (1.75; 1.13-2.71), albeit attenuated after adjustment for maternal obesity (aOR 1.71; 1.08-2.70). Children whose parents both had BMI ≥35 were likely to additionally fail the problem-solving domain (2.93; 1.09-7.85). CONCLUSIONS Findings suggest that maternal and paternal obesity are each associated with specific delays in early childhood development, emphasizing the importance of family information when screening child development.
Collapse
Affiliation(s)
| | | | | | | | - Germaine Buck Louis
- Office of the Director, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Rockville, Maryland
| |
Collapse
|
40
|
The Role of Maternal Nutrition During the Periconceptional Period and Its Effect on Offspring Phenotype. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1014:87-105. [DOI: 10.1007/978-3-319-62414-3_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
41
|
A review of fundamental principles for animal models of DOHaD research: an Australian perspective. J Dev Orig Health Dis 2016; 7:449-472. [DOI: 10.1017/s2040174416000477] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Epidemiology formed the basis of ‘the Barker hypothesis’, the concept of ‘developmental programming’ and today’s discipline of the Developmental Origins of Health and Disease (DOHaD). Animal experimentation provided proof of the underlying concepts, and continues to generate knowledge of underlying mechanisms. Interventions in humans, based on DOHaD principles, will be informed by experiments in animals. As knowledge in this discipline has accumulated, from studies of humans and other animals, the complexity of interactions between genome, environment and epigenetics, has been revealed. The vast nature of programming stimuli and breadth of effects is becoming known. As a result of our accumulating knowledge we now appreciate the impact of many variables that contribute to programmed outcomes. To guide further animal research in this field, the Australia and New Zealand DOHaD society (ANZ DOHaD) Animals Models of DOHaD Research Working Group convened at the 2nd Annual ANZ DOHaD Congress in Melbourne, Australia in April 2015. This review summarizes the contributions of animal research to the understanding of DOHaD, and makes recommendations for the design and conduct of animal experiments to maximize relevance, reproducibility and translation of knowledge into improving health and well-being.
Collapse
|
42
|
Heard ME, Melnyk SB, Simmen FA, Yang Y, Pabona JMP, Simmen RCM. High-Fat Diet Promotion of Endometriosis in an Immunocompetent Mouse Model is Associated With Altered Peripheral and Ectopic Lesion Redox and Inflammatory Status. Endocrinology 2016; 157:2870-82. [PMID: 27175969 PMCID: PMC4929556 DOI: 10.1210/en.2016-1092] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Endometriosis is a benign gynecological condition that causes considerable morbidity due to associated infertility, debilitating pelvic pain and inflammatory dysfunctions. Diet is a highly modifiable risk factor for many chronic diseases, but its contribution to endometriosis has not been extensively investigated, due partly to the paradoxical inverse association between obesity and disease incidence. Nevertheless, chronic exposure to dietary high-fat intake has been linked to greater systemic inflammation and oxidative stress, both features of women with endometriosis. Here, we evaluated the effects of a high-fat diet (HFD) (45% fat kcal) on endometriosis progression using an immunocompetent mouse model where ectopic lesion incidence was induced in wild-type recipients by ip administration of endometrial fragments from transcription factor Krüppel-like factor 9-null donor mice. We show that HFD significantly increased ectopic lesion numbers in recipient mice with no significant weight gain and modifications in systemic ovarian steroid hormone and insulin levels, relative to control diet-fed (17% fat kcal) mice. HFD promotion of lesion establishment was associated with reductions in stromal estrogen receptor 1 isoform and progesterone receptor expression, increased F4/80-positive macrophage infiltration, higher stromal but not glandular epithelial proliferation, and enhanced expression of proinflammatory and prooxidative stress pathway genes. Lesion-bearing HFD-fed mice also displayed higher peritoneal fluid TNFα and elevated local and systemic redox status than control diet-fed counterparts. Our results suggest that HFD intake exacerbates endometriosis outcome in the absence of ovarian dysfunction and insulin resistance in mice and warrants further consideration with respect to clinical management of endometriosis progression and recurrence in nonobese patients.
Collapse
Affiliation(s)
- Melissa E Heard
- Departments of Physiology and Biophysics (M.E.H., F.A.S., Y.Y., J.M.P.P., R.C.M.S.) and Pediatrics (S.B.M.) and Arkansas Children's Hospital Research Institute (S.B.M.), University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205; and Department of Obstetrics and Gynecology (Y.Y.), The Aerospace Central Hospital, Beijing 100049, China
| | - Stepan B Melnyk
- Departments of Physiology and Biophysics (M.E.H., F.A.S., Y.Y., J.M.P.P., R.C.M.S.) and Pediatrics (S.B.M.) and Arkansas Children's Hospital Research Institute (S.B.M.), University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205; and Department of Obstetrics and Gynecology (Y.Y.), The Aerospace Central Hospital, Beijing 100049, China
| | - Frank A Simmen
- Departments of Physiology and Biophysics (M.E.H., F.A.S., Y.Y., J.M.P.P., R.C.M.S.) and Pediatrics (S.B.M.) and Arkansas Children's Hospital Research Institute (S.B.M.), University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205; and Department of Obstetrics and Gynecology (Y.Y.), The Aerospace Central Hospital, Beijing 100049, China
| | - Yanqing Yang
- Departments of Physiology and Biophysics (M.E.H., F.A.S., Y.Y., J.M.P.P., R.C.M.S.) and Pediatrics (S.B.M.) and Arkansas Children's Hospital Research Institute (S.B.M.), University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205; and Department of Obstetrics and Gynecology (Y.Y.), The Aerospace Central Hospital, Beijing 100049, China
| | - John Mark P Pabona
- Departments of Physiology and Biophysics (M.E.H., F.A.S., Y.Y., J.M.P.P., R.C.M.S.) and Pediatrics (S.B.M.) and Arkansas Children's Hospital Research Institute (S.B.M.), University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205; and Department of Obstetrics and Gynecology (Y.Y.), The Aerospace Central Hospital, Beijing 100049, China
| | - Rosalia C M Simmen
- Departments of Physiology and Biophysics (M.E.H., F.A.S., Y.Y., J.M.P.P., R.C.M.S.) and Pediatrics (S.B.M.) and Arkansas Children's Hospital Research Institute (S.B.M.), University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205; and Department of Obstetrics and Gynecology (Y.Y.), The Aerospace Central Hospital, Beijing 100049, China
| |
Collapse
|
43
|
Nigg JT. Where Do Epigenetics and Developmental Origins Take the Field of Developmental Psychopathology? JOURNAL OF ABNORMAL CHILD PSYCHOLOGY 2016; 44:405-19. [PMID: 26758288 PMCID: PMC4822998 DOI: 10.1007/s10802-015-0121-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The time is ripe for upgrading or rethinking the assumed paradigms for how we study developmental psychopathology. The classic transactional models appear robust but need specification in terms of biological and psychosocial processes. That specification is increasingly tractable due to developments in genetics, epigenetics, the measurement of psychosocial processes, and theory and data on developmental origins of health and disease. This essay offers a high-level view of where the field has been and where it may be going in regard to nosology and conceptions of etiology. Remarks seek to consider rapidly evolving contexts not only for children, but also for the science itself due to progress in our field and in neighboring fields. Illustrations are provided as to how syndromal nosology can be enriched and advanced by careful integration with biologically relevant behavioral dimensions and application of quantitative methods. It is concluded that a revised, forward-looking, transactional model of abnormal child psychology will incorporate prenatal and postnatal developmental programming, epigenetic mechanisms and their associated genotype x environment interactions, and inflammatory processes as a potential common mediator influencing numerous health and mental health conditions.
Collapse
Affiliation(s)
- Joel T Nigg
- Oregon Health & Science University, Oregon, Portland, USA.
| |
Collapse
|
44
|
Moran LJ, Spencer L, Russell DL, Hull ML, Robertson SA, Varcoe TJ, Davies MJ, Brown HM, Rodgers RJ. Research Priorities for Fertility and Conception Research as Identified by Multidisciplinary Health Care Practitioners and Researchers. Nutrients 2016; 8:nu8010035. [PMID: 26771633 PMCID: PMC4728649 DOI: 10.3390/nu8010035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/19/2022] Open
Abstract
The Robinson Research Institute of the University of Adelaide convened a multidisciplinary group of n = 33 clinicians, researchers and representatives of government organisations on the 2 October 2014 for a workshop entitled “Promoting fertility and healthy conception. How do we generate greater reproductive health awareness?” The key aim of the workshop was to assess the body of knowledge that informs clinical practice and government policy, and to identify questions and additional information needed by health practitioners and government representatives working in the field of reproductive health and to frame future research and policy. The workshop identified topics that fell mostly into three categories: lifestyle-related, societal and biological factors. The lifestyle topics included nutrition and diet, exercise, obesity, shift work and other factors deemed to be modifiable at the level of the individual. The societal topics included discussions of matters that are structural, and resistant to change by individuals, including specific ethical issues, social disadvantage, government and educational policies. The biological factors are intrinsic physical states of the individual, and included many factors where there is a dense body of scientific knowledge which may not be readily accessible in less academic language. This workshop thus provided an opportunity to identify further actions that could be undertaken to meet the needs of diverse organisations and groups of professionals with an interest in human fertility. Since so many factors in our social and biological environment can impact fertility and preconception health, it is imperative to involve many disciplines or levels of government or societal organisations that have not traditionally been involved in this area.
Collapse
Affiliation(s)
- Lisa J Moran
- The Robinson Research Institute, University of Adelaide, 55 King William Road, North Adelaide, South Australia 5006, Australia.
- Monash Centre for Health Research Implementation, School of Public Health and Preventative Medicine, Monash University, Melbourne 3004, Australia.
| | - Laura Spencer
- The Robinson Research Institute, University of Adelaide, 55 King William Road, North Adelaide, South Australia 5006, Australia.
| | - Darryl L Russell
- The Robinson Research Institute, University of Adelaide, 55 King William Road, North Adelaide, South Australia 5006, Australia.
| | - Mary Louise Hull
- The Robinson Research Institute, University of Adelaide, 55 King William Road, North Adelaide, South Australia 5006, Australia.
| | - Sarah A Robertson
- The Robinson Research Institute, University of Adelaide, 55 King William Road, North Adelaide, South Australia 5006, Australia.
| | - Tamara J Varcoe
- The Robinson Research Institute, University of Adelaide, 55 King William Road, North Adelaide, South Australia 5006, Australia.
| | - Michael J Davies
- The Robinson Research Institute, University of Adelaide, 55 King William Road, North Adelaide, South Australia 5006, Australia.
| | - Hannah M Brown
- The Robinson Research Institute, University of Adelaide, 55 King William Road, North Adelaide, South Australia 5006, Australia.
| | - Raymond J Rodgers
- The Robinson Research Institute, University of Adelaide, 55 King William Road, North Adelaide, South Australia 5006, Australia.
| |
Collapse
|
45
|
Ornellas F, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Combined parental obesity augments single-parent obesity effects on hypothalamus inflammation, leptin signaling (JAK/STAT), hyperphagia, and obesity in the adult mice offspring. Physiol Behav 2015; 153:47-55. [PMID: 26485293 DOI: 10.1016/j.physbeh.2015.10.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/07/2015] [Accepted: 10/16/2015] [Indexed: 11/29/2022]
Abstract
We aimed to evaluate the effects of maternal and/or paternal obesity on offspring body mass, leptin signaling, appetite-regulating neurotransmitters and local inflammatory markers. C57BL/6 mice received standard chow (SC, lean groups) or high-fat diet (HF, obese groups) starting from one month of age. At three months, HF mice became obese relative to SC mice. They were then mated as follows: lean mother and lean father, lean mother and obese father, obese mother and lean father, and obese mother and obese father. The offspring received the SC diet from weaning until three months of age, when they were sacrificed. In the offspring, paternal obesity did not lead to changes in the Janus kinase (JAK)/signal transducer and activation of the transcription (STAT) pathway or feeding behavior but did induce hypothalamic inflammation. On the other hand, maternal obesity resulted in increased weight gain, hyperleptinemia, decreased leptin OBRb receptor expression, JAK/STAT pathway impairment, and increased SOCS3 signaling in the offspring. In addition, maternal obesity elevated inflammatory markers and altered NPY and POMC expression in the hypothalamus. Interestingly, combined parental obesity exacerbated the deleterious outcomes compared to single-parent obesity. In conclusion, while maternal obesity is known to program metabolic changes and obesity in offspring, the current study demonstrated that obese fathers induce hypothalamus inflammation in offspring, which may contribute to the development of metabolic syndromes in adulthood.
Collapse
Affiliation(s)
- Fernanda Ornellas
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|