1
|
Anaya ES, de Groot EL, Lydon JP, Pangas SA, Hartig SM. Contributions of white adipose tissue to energy requirements for female reproduction. Trends Endocrinol Metab 2024; 35:809-820. [PMID: 38749883 PMCID: PMC11387141 DOI: 10.1016/j.tem.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/13/2024] [Accepted: 04/15/2024] [Indexed: 09/12/2024]
Abstract
Body composition impacts female fertility and there are established relationships between adipose tissue and the reproductive system. Maintaining functional adipose tissue is vital for meeting the energetic demands during the reproductive process, from ovulation to delivery and lactation. White adipose tissue (WAT) shows plastic responses to daily physiology and secretes diverse adipokines that affect the hypothalamic-pituitary-ovarian axis, but many other interorgan interactions remain to be determined. This review summarizes the current state of research on the dialogue between WAT and the female reproductive system, focusing on the impact of this crosstalk on ovarian and endometrial factors essential for fecundity.
Collapse
Affiliation(s)
- Elizabeth S Anaya
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - Evelyn L de Groot
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Stephanie A Pangas
- Cancer and Cellular Biology Program, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Sean M Hartig
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
2
|
Akbarinejad V, Cushman RA. Developmental programming of reproduction in the female animal. Anim Reprod Sci 2024; 263:107456. [PMID: 38503204 DOI: 10.1016/j.anireprosci.2024.107456] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/21/2024]
Abstract
Successful reproduction is a cornerstone in food animal industry in order to sustain food production for human. Therefore, various methods focusing on genetics and postnatal environment have been identified and applied to improve fertility in livestock. Yet there is evidence indicating that environmental factors during prenatal and/or neonatal life can also impact the function of reproductive system and fertility in the animals during adulthood, which is called the developmental programming of reproduction. The current review summarizes data associated with the developmental origins of reproduction in the female animals. In this regard, this review focuses on the effect of plane of nutrition, maternal body condition, hypoxia, litter size, maternal age, parity, level of milk production and milk components, lactocrine signaling, stress, thermal stress, exposure to androgens, endocrine disrupting chemicals, mycotoxins and pollutants, affliction with infection and inflammation, and maternal gut microbiota during prenatal and neonatal periods on the neuroendocrine system, puberty, health of reproductive organs and fertility in the female offspring. It is noteworthy that these prenatal and neonatal factors do not always exert their effects on the reproductive performance of the female by compromising the development of organs directly related to reproductive function such as hypothalamus, pituitary, ovary, oviduct and uterus. Since they can impair the development of non-reproductive organs and systems modulating reproductive function as well (e.g., metabolic system and level of milk yield in dairy animals). Furthermore, when these factors affect the epigenetics of the offspring, their adverse effects will not be limited to one generation and can transfer transgenerationally. Hence, pinpointing the factors influencing developmental programming of reproduction and considering them in management of livestock operations could be a potential strategy to help improve fertility in food animals.
Collapse
Affiliation(s)
- Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Robert A Cushman
- USDA, Agricultural Research Service, US. Meat Animal Research Center, Clay Center, NE 68933-0166, United States
| |
Collapse
|
3
|
Jiang H, Chen L, Tian T, Shi H, Huang N, Chi H, Yang R, Long X, Qiao J. Inflammation mediates the effect of adiposity and lipid metabolism indicators on the embryogenesis of PCOS women undergoing in vitro fertilization/intracytoplasmic sperm injection. Front Endocrinol (Lausanne) 2023; 14:1198602. [PMID: 37560312 PMCID: PMC10408295 DOI: 10.3389/fendo.2023.1198602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/26/2023] [Indexed: 08/11/2023] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is a complex reproductive endocrine and metabolic disease affecting women of reproductive age. The low-grade chronic inflammation in PCOS is considered to be associated with obesity and dyslipidemia. We aim to investigate the potential mediating role of white blood cell (WBC) count, a representative inflammatory marker, in the effect of adiposity and lipid metabolism indicators on IVF/ICSI outcomes in PCOS women. Methods We conducted a retrospective cohort study of 1,534 PCOS women who underwent their first IVF/ICSI cycles with autologous oocytes at a reproductive center from January 2018 to December 2020. The associations between PCOS women's adiposity and lipid metabolism indicators and WBC count and IVF/ICSI outcomes were examined using multivariable generalized linear models. Mediation analyses were conducted to evaluate the possible mediating role of WBC count. Results We found significant dose-dependent correlations between adiposity and lipid metabolism indicators and IVF/ICSI outcomes (i.e., hormone levels on the ovulatory triggering day, oocyte development outcomes, fertilization, early embryo development outcomes, and pregnancy outcomes) (all p < 0.05), as well as between adiposity and lipid metabolism indicators and WBC count (all p < 0.001). Increasing WBC count was associated with adverse oocyte and embryonic development outcomes (all p < 0.05). Mediation analyses suggested that increasing serum TG and LDL-C levels and decreasing serum HDL-C level were significantly associated with reduced high-quality Day 3 embryo count in PCOS women, with 21.51%, 9.75%, and 14.10% mediated by WBC count, respectively (all p < 0.05). Conclusions We observed significant associations between lipid metabolism indicators and high-quality Day 3 embryo count in PCOS women, partially mediated by inflammation-related mechanisms, suggesting the potential intervention target for improving embryo quality in PCOS women.
Collapse
Affiliation(s)
- Huahua Jiang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Lixue Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Tian Tian
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Huifeng Shi
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Centre for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Ning Huang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Hongbin Chi
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Rui Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Xiaoyu Long
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
4
|
Wei W, Qin F, Gao J, Chang J, Pan X, Jiang X, Che L, Zhuo Y, Wu D, Xu S. The effect of maternal consumption of high-fat diet on ovarian development in offspring. Anim Reprod Sci 2023; 255:107294. [PMID: 37421833 DOI: 10.1016/j.anireprosci.2023.107294] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/10/2023]
Abstract
The environment encountered by the fetus during its development exerts a profound influence on its physiological function and disease risk in adulthood. Women's intake of high-fat diet during pregnancy and lactation has gradually become an issue of widespread concern. Maternal high-fat diet will not only cause abnormal neurological development and metabolic syndrome symptoms in the offspring, but also affect the fertility of female offspring. Maternal high-fat diet affects the expression of genes related to follicle growth in offspring, such as AAT, AFP and GDF-9, which reduces the number of follicles and impairs follicle development. Additionally, maternal high-fat diet also affects ovarian health by inducing ovarian oxidative stress and cell apoptosis, which collectively can impair the reproductive potential of female offspring. Reproductive potential carries significant importance for both humans and animals. Therefore, this review aims to describe the effect of maternal exposure to high-fat diet on the ovarian development of offspring and to discuss possible mechanisms by which maternal diet affects the growth and metabolism of offspring.
Collapse
Affiliation(s)
- Wenyan Wei
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Feng Qin
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Junjie Gao
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Junlei Chang
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Xujing Pan
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Xuemei Jiang
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Lianqiang Che
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Yong Zhuo
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - De Wu
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China
| | - Shengyu Xu
- Animal Nutrition Institute, Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130 Sichuan, PR China.
| |
Collapse
|
5
|
Han Y, Wu H, Sun S, Zhao R, Deng Y, Zeng S, Chen J. Effect of High Fat Diet on Disease Development of Polycystic Ovary Syndrome and Lifestyle Intervention Strategies. Nutrients 2023; 15:2230. [PMID: 37432488 PMCID: PMC10180647 DOI: 10.3390/nu15092230] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 07/12/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder that affects premenopausal women. The etiology of PCOS is multifaceted, involving various genetic and epigenetic factors, hypothalamic-pituitary-ovarian dysfunction, androgen excess, insulin resistance, and adipose-related mechanisms. High-fat diets (HFDs) has been linked to the development of metabolic disorders and weight gain, exacerbating obesity and impairing the function of the hypothalamic-pituitary-ovarian axis. This results in increased insulin resistance, hyperinsulinemia, and the release of inflammatory adipokines, leading to heightened fat synthesis and reduced fat breakdown, thereby worsening the metabolic and reproductive consequences of PCOS. Effective management of PCOS requires lifestyle interventions such as dietary modifications, weight loss, physical activity, and psychological well-being, as well as medical or surgical interventions in some cases. This article systematically examines the pathological basis of PCOS and the influence of HFDs on its development, with the aim of raising awareness of the connection between diet and reproductive health, providing a robust approach to lifestyle interventions, and serving as a reference for the development of targeted drug treatments.
Collapse
Affiliation(s)
- Yingxue Han
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Hao Wu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Siyuan Sun
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Rong Zhao
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yifan Deng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shenming Zeng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Juan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| |
Collapse
|
6
|
Di Berardino C, Peserico A, Capacchietti G, Zappacosta A, Bernabò N, Russo V, Mauro A, El Khatib M, Gonnella F, Konstantinidou F, Stuppia L, Gatta V, Barboni B. High-Fat Diet and Female Fertility across Lifespan: A Comparative Lesson from Mammal Models. Nutrients 2022; 14:nu14204341. [PMID: 36297035 PMCID: PMC9610022 DOI: 10.3390/nu14204341] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022] Open
Abstract
Female reproduction focuses mainly on achieving fully grown follicles and competent oocytes to be successfully fertilized, as well as on nourishing the developing offspring once pregnancy occurs. Current evidence demonstrates that obesity and/or high-fat diet regimes can perturbate these processes, leading to female infertility and transgenerational disorders. Since the mechanisms and reproductive processes involved are not yet fully clarified, the present review is designed as a systematic and comparative survey of the available literature. The available data demonstrate the adverse influences of obesity on diverse reproductive processes, such as folliculogenesis, oogenesis, and embryo development/implant. The negative reproductive impact may be attributed to a direct action on reproductive somatic and germinal compartments and/or to an indirect influence mediated by the endocrine, metabolic, and immune axis control systems. Overall, the present review highlights the fragmentation of the current information limiting the comprehension of the reproductive impact of a high-fat diet. Based on the incidence and prevalence of obesity in the Western countries, this topic becomes a research challenge to increase self-awareness of dietary reproductive risk to propose solid and rigorous preventive dietary regimes, as well as to develop targeted pharmacological interventions.
Collapse
Affiliation(s)
- Chiara Di Berardino
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alessia Peserico
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Correspondence:
| | - Giulia Capacchietti
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alex Zappacosta
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council, A. Buzzati-Traverso Campus, via E. Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy
| | - Valentina Russo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Annunziata Mauro
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Mohammad El Khatib
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Francesca Gonnella
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Fani Konstantinidou
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Barbara Barboni
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
7
|
A Systematic Review of the Effects of High-Fat Diet Exposure on Oocyte and Follicular Quality: A Molecular Point of View. Int J Mol Sci 2022; 23:ijms23168890. [PMID: 36012154 PMCID: PMC9408717 DOI: 10.3390/ijms23168890] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 12/19/2022] Open
Abstract
Worldwide, infertility affects between 10 and 15% of reproductive-aged couples. Female infertility represents an increasing health issue, principally in developing countries, as the current inclinations of delaying pregnancy beyond 35 years of age significantly decrease fertility rates. Female infertility, commonly imputable to ovulation disorders, can be influenced by several factors, including congenital malformations, hormonal dysfunction, and individual lifestyle choices, such as smoking cigarettes, stress, drug use and physical activity. Moreover, diet-related elements play an important role in the regulation of ovulation. Modern types of diet that encourage a high fat intake exert a particularly negative effect on ovulation, affecting the safety of gametes and the implantation of a healthy embryo. Identifying and understanding the cellular and molecular mechanisms responsible for diet-associated infertility might help clarify the confounding multifaceted elements of infertility and uncover novel, potentially curative treatments. In this view, this systematic revision of literature will summarize the current body of knowledge of the potential effect of high-fat diet (HFD) exposure on oocyte and follicular quality and consequent female reproductive function, with particular reference to molecular mechanisms and pathways. Inflammation, oxidative stress, gene expression and epigenetics represent the main mechanisms associated with mammal folliculogenesis and oogenesis.
Collapse
|
8
|
Čekić N, Grgić A, Kokot A, Mujkić R, Šnajder Mujkić D, Bijelić N, Sablić M. Influence of Maternal Diet and Intergenerational Change in Diet Type on Ovarian and Adipose Tissue Morphology in Female Rat Offspring. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:854. [PMID: 35888573 PMCID: PMC9318591 DOI: 10.3390/medicina58070854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022]
Abstract
Background and Objectives: A high-fat diet causes inflammation in the organism and many metabolic disorders. Adipose tissue secretes adipokines that affect the function of many organs. The health status of the mother before and during pregnancy affects the health of the offspring. The aim of this study was to determine how the type of maternal diet and the change in the type of diet in the offspring affects the histological characteristics of the ovaries and subcutaneous and perigonadal adipose tissue in female rat offspring. Materials and Methods: Ten female rats were divided into two groups. One group was fed standard laboratory chow, and the other was fed a high-fat diet and mated with a male of the same breed. The offspring of both groups of dams were divided into four subgroups with different feeding protocols. At 22 weeks of age, the offspring were sacrificed. Ovaries and subcutaneous and perigonadal adipose tissue were isolated. In the ovaries, the presence of cystic formations was investigated. Histomorphometric analysis was performed in two types of adipose tissue. Results: The weight of the ovaries of the offspring of mothers fed a high-fat diet was significantly higher than that of the offspring of mothers fed standard laboratory diets. Cystic formations were found in the ovaries of the offspring of mothers fed a high-fat diet. In subcutaneous adipose tissue, the percentage of small-sized adipocytes was significantly higher in the offspring of mothers fed standard laboratory diets. There were no significant differences in adipocyte surface area and adipocyte number between groups. Conclusion: Maternal diet influences the morphology of the ovaries and adipose tissue of the offspring.
Collapse
Affiliation(s)
- Nenad Čekić
- Department of Anatomy and Neuroscience, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (N.Č.); (D.Š.M.); (M.S.)
- Department of Anatomy, Histology, Embryology, Pathological Anatomy and Pathological Histology, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
- Department of Surgery, National Memorial Hospital Vukovar, 32000 Vukovar, Croatia
| | - Anđela Grgić
- Department of Anatomy, Histology, Embryology, Pathological Anatomy and Pathological Histology, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
| | - Antonio Kokot
- Department of Anatomy and Neuroscience, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (N.Č.); (D.Š.M.); (M.S.)
| | - Robert Mujkić
- Department of Anatomy, Histology, Embryology, Pathological Anatomy and Pathological Histology, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
| | - Darija Šnajder Mujkić
- Department of Anatomy and Neuroscience, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (N.Č.); (D.Š.M.); (M.S.)
- Clinical Institute of Nuclear Medicine, and Radiation Protection, University Hospital Osijek, 31000 Osijek, Croatia
| | - Nikola Bijelić
- Department of Histology and Embriology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
| | - Marko Sablić
- Department of Anatomy and Neuroscience, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (N.Č.); (D.Š.M.); (M.S.)
| |
Collapse
|
9
|
Sinha N, Lydia Walker G, Sen A. Looking at the Future Through the Mother's Womb: Gestational Diabetes and Offspring Fertility. Endocrinology 2021; 162:6379047. [PMID: 34597389 PMCID: PMC8520322 DOI: 10.1210/endocr/bqab209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Indexed: 12/12/2022]
Abstract
Altered nutrition or intrauterine exposure to various adverse conditions during fetal development or earlier in a mother's life can lead to epigenetic changes in fetal tissues, predisposing those tissues to diseases that manifest when offspring become adults. An example is a maternal obesity associated with gestational diabetes (GDM), where fetal exposure to a hyperglycemic, hyperinsulinemic, and/or hyperlipidemic gestational environment can provoke epigenetic changes that predispose offspring to various diseased conditions later in life. While it is now well established that offspring exposed to GDM have an increased risk of developing obesity, metabolic disorders, and/or cardiovascular disease in adult life, there are limited studies assessing the reproductive health of these offspring. This mini-review discusses the long-term effect of in utero exposure to GDM-associated adverse prenatal environment on the reproductive health of the offspring. Moreover, using evidence from various animal models and human epidemiological studies, this review offers molecular insight and understanding of how epigenetic reprogramming of genes culminates in reproductive dysfunction and the development of subfertility or infertility later in adult life.
Collapse
Affiliation(s)
- Niharika Sinha
- Reproductive and Developmental Sciences Program, East Lansing, Michigan 48824, USA
- Department of Animal Sciences, Michigan State University, East Lansing, Michigan 48824, USA
| | - Gretchen Lydia Walker
- Reproductive and Developmental Sciences Program, East Lansing, Michigan 48824, USA
- Department of Animal Sciences, Michigan State University, East Lansing, Michigan 48824, USA
| | - Aritro Sen
- Reproductive and Developmental Sciences Program, East Lansing, Michigan 48824, USA
- Department of Animal Sciences, Michigan State University, East Lansing, Michigan 48824, USA
- Correspondence: Aritro Sen, PhD, Reproductive and Developmental Sciences Program, 3013 Interdisciplinary Science & Technology Building, Michigan State University, 766 Service Rd, East Lansing, MI 48824, USA.
| |
Collapse
|
10
|
Sun H, Chen Z, Ma C, Lian L, Zhao Z, Niu S, Xu L, Sun J. Effects of maternal dietary energy restriction on laying performance, embryonic development, and lipid Metabolism in broilers. Anim Biosci 2021; 35:698-710. [PMID: 34727634 PMCID: PMC9065775 DOI: 10.5713/ab.21.0301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/19/2021] [Indexed: 11/27/2022] Open
Abstract
Objective The objective of this study was to investigate the effects of different degrees of maternal dietary energy restriction on lipid deposition in embryonic tissues during the medium laying period (37 to 39 weeks) in Arbor Acres (AA) broiler breeders. Methods A single factor design was adopted, and 400 AA broiler breeders (20 weeks of age) with a similar weight were randomly allocated into four groups. The birds in the control group were fed a corn-soybean meal based diet, and those in trial groups were fed diets with 80%, 70%, and 50% energy levels of the basal diet. Incubated eggs from the medium laying period were collected. Samples of developing embryos at various stages were prepared for composition analysis. Results The embryo weight in the 80% energy group was higher than those of the other groups on embryonic day (E) 13, but at 21 E, they were significantly decreased with decreasing energy intake of the broiler breeders (p<0.05). Additionally, the levels of crude fat in tissues in the restriction groups were significantly decreased (p<0.05). The long axis and area of adipocytes in breast muscle, thigh muscle and the liver were significantly decreased (p<0.05) at 21 E in the 80%, 70%, and 50% energy groups. Conclusion The effects of the 80% maternal dietary energy restriction energy affects egg production performance, egg quality, and nutrient deposition in egg weights, which then directly impacts on the developmental process of embryos, especially on fat utilization and deposition.
Collapse
Affiliation(s)
- Hao Sun
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Zhihui Chen
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Chenzhan Ma
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Lina Lian
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Zeyu Zhao
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Shupeng Niu
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Liangmei Xu
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Jinhua Sun
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, Heilongjiang, China
| |
Collapse
|
11
|
Environmental Alterations during Embryonic Development: Studying the Impact of Stressors on Pluripotent Stem Cell-Derived Cardiomyocytes. Genes (Basel) 2021; 12:genes12101564. [PMID: 34680959 PMCID: PMC8536136 DOI: 10.3390/genes12101564] [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: 09/08/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/16/2022] Open
Abstract
Non-communicable diseases (NCDs) sauch as diabetes, obesity and cardiovascular diseases are rising rapidly in all countries world-wide. Environmental maternal factors (e.g., diet, oxidative stress, drugs and many others), maternal illnesses and other stressors can predispose the newborn to develop diseases during different stages of life. The connection between environmental factors and NCDs was formulated by David Barker and colleagues as the Developmental Origins of Health and Disease (DOHaD) hypothesis. In this review, we describe the DOHaD concept and the effects of several environmental stressors on the health of the progeny, providing both animal and human evidence. We focus on cardiovascular diseases which represent the leading cause of death worldwide. The purpose of this review is to discuss how in vitro studies with pluripotent stem cells (PSCs), such as embryonic and induced pluripotent stem cells (ESC, iPSC), can underpin the research on non-genetic heart conditions. The PSCs could provide a tool to recapitulate aspects of embryonic development “in a dish”, studying the effects of environmental exposure during cardiomyocyte (CM) differentiation and maturation, establishing a link to molecular mechanism and epigenetics.
Collapse
|
12
|
Identification of Long Non-Coding RNAs Involved in Porcine Fat Deposition Using Two High-Throughput Sequencing Methods. Genes (Basel) 2021; 12:genes12091374. [PMID: 34573356 PMCID: PMC8467702 DOI: 10.3390/genes12091374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022] Open
Abstract
Adipose is an important body tissue in pigs, and fatty traits are critical in pig production. The function of long non-coding RNA (lncRNA) in fat deposition and metabolism has been found in previous studies. In this study, we collected the adipose tissue of six Landrace pigs with contrast backfat thickness (nhigh = 3, nlow = 3), after which we performed strand-specific RNA sequencing (RNA-seq) based on pooling and biological replicate methods. Biological replicate and pooling RNA-seq revealed 1870 and 1618 lncRNAs, respectively. Using edgeR, we determined that 1512 genes and 220 lncRNAs, 2240 genes and 127 lncRNAs were differentially expressed in biological replicate and pooling RNA-seq, respectively. After target gene prediction, we found that ACSL3 was cis-targeted by lncRNA TCONS-00052400 and could activate the conversion of long-chain fatty acids. In addition, lncRNA TCONS_00041740 cis-regulated gene ACACB regulated the rate-limiting enzyme in fatty acid oxidation. Since these genes have necessary functions in fat metabolism, the results imply that the lncRNAs detected in our study may affect backfat deposition in swine through regulation of their target genes. Our study explored the regulation of lncRNA and their target genes in porcine backfat deposition and provided new insights for further investigation of the biological functions of lncRNA.
Collapse
|
13
|
|
14
|
Shi M, Sirard MA. Transcriptome and epigenome analysis of porcine embryos from non-esterified fatty acid-exposed oocytes. Domest Anim Endocrinol 2021; 76:106605. [PMID: 33631700 DOI: 10.1016/j.domaniend.2021.106605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 12/22/2022]
Abstract
Increasing evidence indicates that maternal malnutrition leads to decreased female fertility and dysregulated metabolic homeostasis in offspring. High levels of non-esterified fatty acids (NEFAs) in follicular fluid were reported to be involved in these maternal nutritional effects, but the mechanisms remain unclear. This study explored the mechanisms of action of abnormal NEFA levels during porcine oocyte in vitro maturation (IVM) on early embryo development (blastocysts) using phenotypic, transcriptomic, and epigenetic analysis. The oocytes were treated during IVM with, in addition to the 1% (v/v) porcine follicular fluid in the control group, a combination of 468 μmol/L palmitic acid, 194 μmol/L stearic acid, and 534 μmol/L oleic acid supplemented to North Carolina State University-23 (NCSU-23) maturation medium to achieve a high level of NEFAs. After IVM, oocytes were in vitro fertilized and then cultured in regular conditions for blastocysts. Expanded blastocysts were collected to complete transcriptomic and epigenetic analysis. Macroscopically, high level of NEFAs impaired embryo development by reducing the blastocyst rate. Analysis of the transcriptome revealed that pathways related to inflammation, apoptosis, metabolism, and oxidative stress were the most affected. Moreover, DNA methylation data demonstrated differentially methylated regions in genes related to cellular metabolism and inflammation processes. Therefore, our conclusion is that high level of NEFAs during IVM might affect porcine early embryo development by diminishing blastocyst rate and altering gene expression, especially at the metabolism and cell status levels, which could further decrease the embryo quality.
Collapse
Affiliation(s)
- M Shi
- Centre de recherche en reproduction, développement et santé intergénérationnelle, Faculté des sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Pavillon INAF, Université Laval, Québec, Québec, Canada
| | - M-A Sirard
- Centre de recherche en reproduction, développement et santé intergénérationnelle, Faculté des sciences de l'Agriculture et de l'Alimentation, Département des Sciences Animales, Pavillon INAF, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
15
|
Xu S, Wu X, Dong Y, Xu M, Li Z, Chen S, Zhuo Y, Lin Y, Che L, Fang Z, Feng B, Li J, Wang J, Wu D, Ren Z. Glucose activates the primordial follicle through the AMPK/mTOR signaling pathway. Clin Transl Med 2020. [PMCID: PMC7418812 DOI: 10.1002/ctm2.122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background We have previously found that the energy level in sows affects the activation of primordial follicles. Glucose is the primary metabolic substrate of dietary energy and its effect and mechanism of action with regards to the activation and development of primordial follicle remain unclear. Studies utilizing several different animal cells have shown that energy stress, induced by glucose starvation, activates AMPK and participates in a variety of cellular processes by regulating the Hippo and mTOR signaling pathways. However, whether glucose can affect primordial follicle activation through the above pathways has not been reported. Methods We developed an in vitro culture system for mouse ovaries to investigate the effects of glucose on the primordial follicle activation. Protein expression of AMPK‐Hippo‐YAP and AMPK‐mTOR pathway was investigated under glucose starvation and optimal glucose level treatment. Then, ovaries were treated with AICAR or Compound C in vitro to explore the effect of AMPK activation or inhibition on primordial follicle activation, and the changes of AMPK‐Hippo‐YAP and AMPK‐mTOR signaling pathways. Finally, investigated the signaling pathways affected by glucose potentially affecting the primordial follicle activation in vivo. Results The glucose was an essential nutrient for primordial follicle activation and we identified 25 mM glucose as the optimal level (P < .05) for the primordial follicle activation in vitro. The glycolysis pathway was involved in primordial follicle activation (P < .05) of ovaries cultured in vitro. The glucose affected the activation of primordial follicles in vitro through AMPK/mTOR signaling pathway by AMPK activation or inhibition treatment and follicle ratio count (P < .05). Moreover, glucose affected the primordial follicle activation of ovary in vivo via mTOR signaling pathway. Conclusions This study demonstrates that glucose affects the primordial follicle activation through the AMPK/mTOR rather than the AMPK/Hippo signaling pathway.
Collapse
Affiliation(s)
- Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Xiaoling Wu
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Yanpeng Dong
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Mengmeng Xu
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Zimei Li
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Sirun Chen
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Jian Li
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Jianping Wang
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University; Key Laboratory of Animal Disease‐resistant Nutrition, Ministry of EducationMinistry of Agriculture and Rural Affairs, Sichuan Province Chengdu Sichuan P. R. China
| | - Zhihua Ren
- College of Veterinary Medicine, Sichuan Province Key Laboratory of Animal Disease and Human Health, Key Laboratory of Environmental Hazard and Human Health of Sichuan ProvinceSichuan Agricultural University Chengdu Sichuan P. R. China
| |
Collapse
|
16
|
Che L, Xu M, Gao K, Wang L, Yang X, Wen X, Xiao H, Jiang Z. Effects of dietary valine supplementation during late gestation on the reproductive performance and mammary gland development of gilts. J Anim Sci Biotechnol 2020; 11:15. [PMID: 32099647 PMCID: PMC7029528 DOI: 10.1186/s40104-019-0420-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/22/2019] [Indexed: 02/07/2023] Open
Abstract
Background Mammary gland development during late gestation in gilts is a major factor that alters the composition of colostrum and growth performance of piglets. Plasma valine is taken up and metabolized extensively by the mammary gland; however, the effects of valine on mammary gland development during late gestation are still unclear. Thirty primiparous gilts were divided into three treatment groups (n = 10) and received one of the three diets starting on day 75 of gestation until the day of farrowing. The total dietary valine to lysine ratio of the three diets was 0.63 (LV), 0.73 (MV), and 0.93 (HV), respectively. Results Dietary valine supplementation during late gestation did not affect (P > 0.05) the litter size and weight at farrowing; however, the piglet weight and average daily gain at weaning were linearly increased (P < 0.05) as the dietary valine increased. The highest piglet weight at weaning was observed when the gilts were provided the HV diet. Dietary valine supplementation linearly elevated (P < 0.05) protein, fat and solids-not-fat and some free amino acids content in colostrum. The concentration of prolactin in plasma of gilts was linearly increased in response to valine supplementation at days 1 and 10 of lactation (P < 0.05). Furthermore, with increasing dietary valine allowance, a linear increase (P < 0.05) was observed in the area of the lumen of alveolus and the content of DNA, RNA, and total protein in the mammary tissues at day 1 of lactation. Moreover, the protein expression of cyclin D1, p-mTOR, p-S6, and p-4EBP1 was also linearly increased (P < 0.05) in the mammary tissue at day 1 of lactation. However, no difference (P > 0.05) was observed in the indices related to mammary development and the mTOR signaling pathway at day 21 of lactation. Conclusion The results revealed that increasing the total dietary valine to lysine ratio to 0.93 during late gestation significantly enhances the piglet weight and average daily gain at weaning probably due to improved development of mammary gland.
Collapse
Affiliation(s)
- Long Che
- 1State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, No.1 Dafeng Street, Wushan Rd, Tianhe District, Guangzhou, 510640 Guangdong China.,2College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, 450046 Henan China
| | - Mengmeng Xu
- 1State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, No.1 Dafeng Street, Wushan Rd, Tianhe District, Guangzhou, 510640 Guangdong China.,2College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, 450046 Henan China
| | - Kaiguo Gao
- 1State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, No.1 Dafeng Street, Wushan Rd, Tianhe District, Guangzhou, 510640 Guangdong China
| | - Li Wang
- 1State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, No.1 Dafeng Street, Wushan Rd, Tianhe District, Guangzhou, 510640 Guangdong China
| | - Xuefen Yang
- 1State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, No.1 Dafeng Street, Wushan Rd, Tianhe District, Guangzhou, 510640 Guangdong China
| | - Xiaolu Wen
- 1State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, No.1 Dafeng Street, Wushan Rd, Tianhe District, Guangzhou, 510640 Guangdong China
| | - Hao Xiao
- 1State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, No.1 Dafeng Street, Wushan Rd, Tianhe District, Guangzhou, 510640 Guangdong China
| | - Zongyong Jiang
- 1State Key Laboratory of Livestock and Poultry Breeding; Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture; Guangdong Public Laboratory of Animal Breeding and Nutrition; Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, No.1 Dafeng Street, Wushan Rd, Tianhe District, Guangzhou, 510640 Guangdong China
| |
Collapse
|
17
|
Maternal energy insufficiency affects testicular development of the offspring in a swine model. Sci Rep 2019; 9:14533. [PMID: 31601864 PMCID: PMC6787339 DOI: 10.1038/s41598-019-51041-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022] Open
Abstract
We determined the effects of insufficient maternal energy on testicular development in offspring in a swine model. Thirty-six sows were divided into control (CON) and low-energy diet (LE) groups during gestation. We observed that the number of Sertoli, germ, and Leydig cells in the offspring of the CON group were significantly higher than those in the LE group at 28 and 120 d after birth. Furthermore, the percentage of apoptotic testis cells was significantly higher in the offspring of the LE group than in the CON group. Transcriptome analysis of differentially expressed mRNAs and long noncoding RNAs in offspring testes indicated that these RNAs were mainly involved in lipid metabolism, apoptosis, cell proliferation, and some pivotal regulatory pathways. Results revealed that AMPK-PI3K-mTOR, MAPK, and oxidative phosphorylation signaling pathways play an important role in mediating the programming effect of insufficient maternal energy on testicular development, and that this effect occurs mainly at an early stage in life. mRNA and protein expression analyses confirmed the importance of certain signaling pathways in the regulation of testicular development. This study provides insights into the influence and possible mechanism underlying the effect of inadequate maternal energy intake on testicular development in the offspring.
Collapse
|
18
|
Maternal betaine protects rat offspring from glucocorticoid-induced activation of lipolytic genes in adipose tissue through modification of DNA methylation. Eur J Nutr 2019; 59:1707-1716. [DOI: 10.1007/s00394-019-02025-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 06/08/2019] [Indexed: 12/19/2022]
|
19
|
Mehri K, Banan Khojasteh SM, Seyed Mahdi BK, Fereshteh F, Zavvari Oskuye Z, Ebrahimi H, Diba R, Bayandor P, Hosseindoost M, Babri S. Effect of troxerutin on apelin-13, apelin receptors (APJ), and ovarian histological changes in the offspring of high-fat diet fed rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:637-642. [PMID: 31231491 PMCID: PMC6570758 DOI: 10.22038/ijbms.2019.34158.8123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Maternal high-fat diet (HFD) consumption has been linked to metabolic disorders and reproductive dysfunctions in offspring. Troxerutin (TRO) has anti-hyperlipidemic, anti-oxidant, and anti-inflammatory effects. This study examined the effects of TRO on apelin-13, its receptors mRNA and ovarian histological changes in the offspring of HFD fed rats. MATERIALS AND METHODS Female Wistar rats were randomly divided into control diet (CD) or HFD groups and received these diets for eight weeks. After mating, dams were assigned into four subgroups: CD, CD + TRO, HFD, and HFD + TRO, and received their respective diets until the end of lactation. Troxerutin (150 mg/kg/day) was gavaged in the CD + TRO and HFD + TRO groups during pregnancy. On the postnatal day (PND) 21 all female offspring were separated and fed CD until PND 90. On PND 90 animals were sacrificed and ovarian tissue samples were collected for further evaluation. RESULTS Results showed that HFD significantly decreased serum apelin-13 in the female offspring of the HFD dams, which was significantly reversed by TRO. Moreover, real-time polymerase chain reaction (PCR) analysis revealed that TRO treatment significantly decreased the ovarian mRNA expression of the apelin-13 receptor in the troxerutin-received offspring. Furthermore, histological examination revealed that TRO increased the number of atretic follicles in the ovaries of HFD+TRO offspring. CONCLUSION Maternal high fat feeding compromises ovarian health including follicular growth and development in the adult offspring and troxerutin treatment improved negative effects of maternal HFD on the apelin-13 level and ovarian development of offspring.
Collapse
Affiliation(s)
- Keyvan Mehri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Fereshteh Fereshteh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hadi Ebrahimi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roghaye Diba
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Bayandor
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hosseindoost
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Babri
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
20
|
Effect of Sweet Potato Vine on the Onset of Puberty and Follicle Development in Chinese Meishan Gilts. Animals (Basel) 2019; 9:ani9060297. [PMID: 31151241 PMCID: PMC6617383 DOI: 10.3390/ani9060297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/26/2019] [Accepted: 05/27/2019] [Indexed: 01/22/2023] Open
Abstract
This study was conducted to evaluate the effects of sweet potato vine on the onset of puberty and the follicular development in the ovaries of Chinese Meishan gilts. A total of 20 Meishan gilts (initial body weight at 30 ± 0.18 kg) were randomly fed a control (CON) or sweet potato vine (SPV) supplemented diet until 19 days following the third estrous. Sweet potato vine was instead of part of basal diet with the same amount of energy and protein in the sweet potato vine group. The results indicate that gilts fed with sweet potato vine reached puberty 9.4 days later. The development of ovaries was enhanced by sweet potato vine supplementation, characterized by an increase (p < 0.05) in the relative weight of the ovaries and the number of large follicles (>5 mm). Sweet potato vine supplementation increased (p < 0.05) the total superoxide dismutase (T-SOD) and reduced (p < 0.05) the concentration of malondialdehyde (MDA) in the serum of the gilts. Also, the expression of superoxide dismutase-1 (SOD1) and luteinizing hormone receptor (LHR) mRNA in the granulosa cells from the large follicle (>5 mm) of gilts in the SPV group were increased (p < 0.05) as compared with the CON group. These results indicate that gilts fed with sweet potato vine exhibited delayed puberty as well as improved follicular development, which may contribute to the reproductive performance of Chinese Meishan gilts.
Collapse
|
21
|
Effects of Dietary Taurine Supplementation to Gilts during Late Gestation and Lactation on Offspring Growth and Oxidative Stress. Animals (Basel) 2019; 9:ani9050220. [PMID: 31064160 PMCID: PMC6562957 DOI: 10.3390/ani9050220] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/25/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Previous studies showed that gilts had elevated oxidative stress during late gestation and lactation, and could affect offspring growth. Taurine (Tau) is an important regulator of oxidative stress and possesses growth-enhancing properties. Our results suggested that taurine supplementation during late gestation and lactation of gilts increased growth performance in piglets through improved milk quality of gilts and intestinal morphology and barrier function of offspring. Abstract Birth is one of the most important events of animal production agriculture, as newborns are abruptly forced to adapt to environmental and nutritional disruptions that can lead to oxidative damage and delay in growth. Taurine (Tau) is an important regulator of oxidative stress and possesses growth-enhancing properties. In the present study, we investigated the effects of dietary Tau supplementation in gilts during late gestation and lactation on the growth performance of piglets by assessing intestinal morphology and barrier function, and oxidative stress status. Sixteen gilts were randomly allocated to the Con (basal diet) and Tau (basal diet with 1% Tau) groups from 75 d of gestation to weaning. Maternal dietary Tau supplementation significantly increased weaning weight and average daily gain weight in piglets. Piglets in the Tau group had higher villus height and villus height-to-crypt depth ratio (VCR), ZO-1 protein expression, and secretory immunoglobulin A (sIgA) content in the jejunum. Meanwhile, Tau bebeficial affected the milk quality of gilts, as indicated by decreased malondialdehyde (MDA) concentration and increased total superoxide dismutase (T-SOD), total antioxidative capability (T-AOC), glutathione peroxidase (GPx), and catalase (CAT) activity. Furthermore, Tau supplementation increased T-SOD activity in plasma and SOD2 protein expression in the jejunum in the piglets. In conclusion, this study provides evidence that dietary Tau supplementation to gilts improves growth performance in piglets, owing to improved intestinal morphology and barrier function, as well as inhibition of oxidative stress.
Collapse
|
22
|
Maternal β-hydroxy-β-methylbutyrate (HMB) supplementation during pregnancy affects early folliculogenesis in the ovary of newborn piglets. Theriogenology 2019; 128:91-100. [PMID: 30743108 DOI: 10.1016/j.theriogenology.2019.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 01/15/2019] [Accepted: 02/01/2019] [Indexed: 12/22/2022]
Abstract
Beta-hydroxy-beta-methylbutyrate (HMB) is a leucine metabolite with protein anabolic effects. This study was designed to determine whether prenatal HMB treatment has an effect on oogenesis and folliculogenesis in the ovary of newborn piglets. HMB decreased the number of egg nests and primordial follicles and increased the pool of developing follicles compared to the control group. Although the percentage of TUNEL-positive oocytes within the egg nests was higher in HMB-treated group no increase in the Bax/Bcl-2 ratio and active caspase-3 expression was observed. Moreover, the granulosa cell proliferation index and StAR protein expression were higher in HMB-treated group. In contrast to the control group, the expression of E-cadherins was reduced after the HMB treatment. In addition, a significant increase in the serum level of gonadotropins and steroid hormones was detected in HMB-treated piglets. In conclusion, prenatal HMB treatment dysregulates hormonal homeostasis which impairs early folliculogenesis in piglets.
Collapse
|
23
|
El-Sayyad HIH, El-Shershaby EMF, El-Mansi AA, El-Ashry NE. Anti-hypercholesterolemic impacts of barley and date palm fruits on the ovary of Wistar albino rats and their offspring. Reprod Biol 2018; 18:236-251. [PMID: 30005909 DOI: 10.1016/j.repbio.2018.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/25/2018] [Accepted: 07/05/2018] [Indexed: 12/23/2022]
Abstract
A high cholesterol diet is related to ovarian dysfunction and infertility which has been increased among young ages consuming processed food products. The present study was conducted to evaluate the role of a high cholesterol diet on the ovaries of young female rats via assessments of histopathology, immunohistochemistry, oxidative stress and apoptic markers. Also, mating of hypercholesterolemic female rats was carried out to measure the fertility and numbers of their offspring. At the same time, phytotherapy was carried out through supplementing the diet with barley and/ or date palm fruits (10%) during the experiment to assess the phyto-therapeutic impacts in attenuation of drastic hypercholesterolemic effects. Hypercholesterolemic diet-fed rats exhibited damage of the ovarian follicles and increased follicular atresia. Furthermore, expression of cleaved caspase-3 was upregulated, while PCNA was downregulated in granulosa, theca and stroma cells. Hypercholesterolemic female rats showed marked depletion of antioxidative enzymes, increased lipid peroxidation and apoptotic markers. Alterations to the female serum hormones were detected. Offspring maternally fed on hypercholesterolemic diet showed a significant decrease of body weight and altered sex ratio. However, concomitant supplementation of barley and or date fruits to hypercholesterolemic groups revealed marked improvement of ovarian structure and function. On the basis of these evidences, it is believed that the enhanced synergistic effects of barley and/or date palm fruits in the amelioration of ovarian structure and functions were elicited by the potential antioxidant activity of their phytomicronutrients, polyphenols, β-glucan and trace elements. These materials scavenge free radicals from inflamed cells that can be used to establish an effective and novel therapeutic strategy for activating ovarian cell regeneration.
Collapse
Affiliation(s)
| | | | - Ahmed A El-Mansi
- Zoology Dept., Faculty of Science, Mansoura University, Mansoura, Egypt; Biology Dept., Faculty of Science, King Khalid University, Abha, Saudi Arabia.
| | | |
Collapse
|
24
|
Archibong AE, Rideout ML, Harris KJ, Ramesh A. OXIDATIVE STRESS IN REPRODUCTIVE TOXICOLOGY. CURRENT OPINION IN TOXICOLOGY 2017; 7:95-101. [PMID: 30105313 DOI: 10.1016/j.cotox.2017.10.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Oxidative stress (OS) has been implicated in the causation of environmentally-induced diseases. However, the role of toxicants in the pathophysiology of disorders and diseases affecting the reproductive system are less understood. This review focuses on some of the mechanisms that underlie OS-induced reproductive toxicity at the cellular- and organ levels (germ cell damage and perturbed organ responses to endocrine stimuli). While most of the reproductive and developmental studies conducted in adult animals and transgenerational adult animals point to the involvement of genotoxicity, the part played by epigenetic alterations is accorded a recent recognition, thus warranting more studies in this area. Additionally, metabolomic, proteomic and transcriptomic approaches need to be employed to advance our understanding of key metabolites formed and the expression of anti-OS genes at the molecular level that are necessary for combating reactive oxygen species formation. The resulting data could be analyzed using bioinformatics tools to identify the pathways linked to disease causation and as a consequence, the adoption of therapeutic strategies, including but not limited to administering phytochemicals (many of which possess antioxidant properties) to improve disease outcomes.
Collapse
Affiliation(s)
| | | | - Kenneth J Harris
- Department of Biochemistry & Cancer Biology, Meharry Medical College, Nashville TN 37208
| | - Aramandla Ramesh
- Department of Biochemistry & Cancer Biology, Meharry Medical College, Nashville TN 37208
| |
Collapse
|
25
|
Akbarinejad V, Gharagozlou F, Vojgani M. Temporal effect of maternal heat stress during gestation on the fertility and anti-Müllerian hormone concentration of offspring in bovine. Theriogenology 2017; 99:69-78. [DOI: 10.1016/j.theriogenology.2017.05.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 01/28/2023]
|