1
|
Mbegbu EC, Salavati M, Aka LO, Obidike IR, Tang JCY, Fraser WD, Hanson MA, Green LR, Fouladi-Nashta AA. Impact of Periconceptional and Gestational Vitamin D3 Restriction on Fetal Folliculogenesis and Anti-Mullerian Hormone Secretion Using Sheep as a Model. Reprod Domest Anim 2024; 59:e14717. [PMID: 39234989 DOI: 10.1111/rda.14717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/06/2024]
Abstract
Ovarian reserve is a reflection of the overall female reproductive potential. Vitamin D status has been suspected to influence fetal development and female fertility. As maternal diet during pregnancy can affect fetal development and future fertility, we hypothesised that periconceptional and gestational Vitamin D restriction could affect folliculogenesis and AMH secretion in the offspring. Nineteen sexually mature Welsh mountain ewes were randomly assigned to Vitamin D3 deficient (VDD, n = 10) and Vitamin D3 control (VDC, n = 9) diets from 17 days (d) before mating, up to 127-130 days of gestation, when fetal ovaries were collected (3 from VDC and 6 from VDD). Serum 25(OH)D3 concentrations were lower in VDD compared with VDC (p < 0.05). Relative to total follicle number, the percentage of primordial follicles was higher (p < 0.05), while the percentage of primary follicles was lower (p < 0.05) in VDD group compared with VDC group fetal ovaries. The integrated density value and percentage of affected area in TUNEL staining in VDD group did not vary from VDC group fetal ovaries (p > 0.05). Relative expression of AMH mRNA and AMH protein in VDD fetal ovaries were not statistically different compared with controls (p > 0.05). The relative expression of VDR mRNA were lower in VDD compared with VDC group fetal ovaries (p < 0.05). These data indicate that maternal Vitamin D dietary restriction is associated with ovarian tissue stemness and increased primordial follicle number but does not promote normal follicle recruitment or development in sheep fetal ovaries.
Collapse
Affiliation(s)
- E C Mbegbu
- Veterinary Physiology and Biochemistry, University of Nigeria, Nsukka, Nigeria
- Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - M Salavati
- Comparative Biomedical Sciences, Royal Veterinary College, London, UK
- Dairy Research Innovation Centre, South and West Faculty, Scotland's Rural College, Dumfries, UK
| | - L O Aka
- Veterinary Biosciences, St Matthew's University, Grand Cayman, Cayman Islands
| | - I R Obidike
- Veterinary Physiology and Biochemistry, University of Nigeria, Nsukka, Nigeria
| | - J C Y Tang
- Bioanalytical Facility, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
- Department of Clinical Biochemistry, Diabetes and Endocrinology, Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, UK
| | - W D Fraser
- Bioanalytical Facility, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
- Department of Clinical Biochemistry, Diabetes and Endocrinology, Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, UK
| | - M A Hanson
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - L R Green
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | |
Collapse
|
2
|
Paula VG, Sinzato YK, Gallego FQ, Cruz LL, Aquino AMD, Scarano WR, Corrente JE, Volpato GT, Damasceno DC. Intergenerational Hyperglycemia Impairs Mitochondrial Function and Follicular Development and Causes Oxidative Stress in Rat Ovaries Independent of the Consumption of a High-Fat Diet. Nutrients 2023; 15:4407. [PMID: 37892483 PMCID: PMC10609718 DOI: 10.3390/nu15204407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
We analyzed the influence of maternal hyperglycemia and the post-weaning consumption of a high-fat diet on the mitochondrial function and ovarian development of the adult pups of diabetic rats. Female rats received citrate buffer (Control-C) or Streptozotocin (for diabetes induction-D) on postnatal day 5. These adult rats were mated to obtain female pups (O) from control dams (OC) or from diabetic dams (OD), and they received a standard diet (SD) or high-fat diet (HFD) from weaning to adulthood and were distributed into OC/SD, OC/HFD, OD/SD, and OD/HFD. In adulthood, the OGTT and AUC were performed. These rats were anesthetized and euthanized for sample collection. A high percentage of diabetic rats were found to be in the OD/HFD group (OD/HFD 40% vs. OC/SD 0% p < 0.05). Progesterone concentrations were lower in the experimental groups (OC/HFD 0.40 ± 0.04; OD/SD 0.30 ± 0.03; OD/HFD 0.24 ± 0.04 vs. OC/SD 0.45 ± 0.03 p < 0.0001). There was a lower expression of MFF (OD/SD 0.34 ± 0.33; OD/HFD 0.29 ± 0.2 vs. OC/SD 1.0 ± 0.41 p = 0.0015) and MFN2 in the OD/SD and OD/HFD groups (OD/SD 0.41 ± 0.21; OD/HFD 0.77 ± 0.18 vs. OC/SD 1.0 ± 0.45 p = 0.0037). The number of follicles was lower in the OD/SD and OD/HFD groups. A lower staining intensity for SOD and Catalase and higher staining intensity for MDA were found in ovarian cells in the OC/HFD, OD/SD, and OD/HFD groups. Fetal programming was responsible for mitochondrial dysfunction, ovarian reserve loss, and oxidative stress; the association of maternal diabetes with an HFD was responsible for the higher occurrence of diabetes in female adult pups.
Collapse
Affiliation(s)
- Verônyca Gonçalves Paula
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Yuri Karen Sinzato
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Franciane Quintanilha Gallego
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Larissa Lopes Cruz
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Ariana Musa de Aquino
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| | - Wellerson Rodrigo Scarano
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| | - José Eduardo Corrente
- Research Support Office, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças 78600-000, MG, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu 18618-687, SP, Brazil
| |
Collapse
|
3
|
Perono GA, Petrik JJ, Thomas PJ, Holloway AC. The effects of polycyclic aromatic compounds (PACs) on mammalian ovarian function. Curr Res Toxicol 2022; 3:100070. [PMID: 35492299 PMCID: PMC9043394 DOI: 10.1016/j.crtox.2022.100070] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/24/2022] [Accepted: 04/01/2022] [Indexed: 12/09/2022] Open
Abstract
Toxicity of polycyclic aromatic compounds (PACs) is limited to a subset of PACs. Exposure to these compounds impact major processes necessary for ovarian function. PAC exposure causes follicle loss and aberrant steroid production and angiogenesis. PAC exposure may increase the risk for impaired fertility and ovarian pathologies. The study of PACs as ovarian toxicants should include additional compounds.
Polycyclic aromatic compounds (PACs) are a broad class of contaminants ubiquitously present in the environment due to natural and anthropogenic activities. With increasing industrialization and reliance on petroleum worldwide, PACs are increasingly being detected in different environmental compartments. Previous studies have shown that PACs possess endocrine disruptive properties as these compounds often interfere with hormone signaling and function. In females, the ovary is largely responsible for regulating reproductive and endocrine function and thus, serves as a primary target for PAC-mediated toxicity. Perturbations in the signaling pathways that mediate ovarian folliculogenesis, steroidogenesis and angiogenesis can lead to adverse reproductive outcomes including polycystic ovary syndrome, premature ovarian insufficiency, and infertility. To date, the impact of PACs on ovarian function has focused predominantly on polycyclic aromatic hydrocarbons like benzo(a)pyrene, 3-methylcholanthrene and 7,12-dimethylbenz[a]anthracene. However, investigation into the impact of substituted PACs including halogenated, heterocyclic, and alkylated PACs on mammalian reproduction has been largely overlooked despite the fact that these compounds are found in higher abundance in free-ranging wildlife. This review aims to discuss current literature on the effects of PACs on the ovary in mammals, with a particular focus on folliculogenesis, steroidogenesis and angiogenesis, which are key processes necessary for proper ovarian functions.
Collapse
|
4
|
Ben Maamar M, Nilsson EE, Skinner MK. Epigenetic transgenerational inheritance, gametogenesis and germline development†. Biol Reprod 2021; 105:570-592. [PMID: 33929020 PMCID: PMC8444706 DOI: 10.1093/biolre/ioab085] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/12/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
One of the most important developing cell types in any biological system is the gamete (sperm and egg). The transmission of phenotypes and optimally adapted physiology to subsequent generations is in large part controlled by gametogenesis. In contrast to genetics, the environment actively regulates epigenetics to impact the physiology and phenotype of cellular and biological systems. The integration of epigenetics and genetics is critical for all developmental biology systems at the cellular and organism level. The current review is focused on the role of epigenetics during gametogenesis for both the spermatogenesis system in the male and oogenesis system in the female. The developmental stages from the initial primordial germ cell through gametogenesis to the mature sperm and egg are presented. How environmental factors can influence the epigenetics of gametogenesis to impact the epigenetic transgenerational inheritance of phenotypic and physiological change in subsequent generations is reviewed.
Collapse
Affiliation(s)
- Millissia Ben Maamar
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Eric E Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Michael K Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
5
|
Paula VG, Vesentini G, Sinzato YK, Moraes-Souza RQ, Volpato GT, Damasceno DC. Intergenerational high-fat diet impairs ovarian follicular development in rodents: a systematic review and meta-analysis. Nutr Rev 2021; 80:889-903. [PMID: 34459492 DOI: 10.1093/nutrit/nuab049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
CONTEXT Excessive consumption of high-fat diets has increased in the population over time and is harmful to female fertility. OBJECTIVE To investigate and discuss the effects of a high-fat diet on ovarian follicles in rodents. DATA SOURCE A systematic literature search of PubMed, EMBASE, Web of Science, and SCOPUS was carried out. DATA EXTRACTION Study characteristics, including study design, population, intervention, outcome, and risk of bias were analyzed. DATA ANALYSIS Twenty-two articles were included in a systematic review. Given the availability of studies, a quantitative meta-analysis included 12 studies that were performed for outcomes. There was a decrease in primordial follicles in female rodents that received a high-fat diet compared with the standard diet group. The offspring of mothers exposed to a high-fat diet showed an increased number of cystic follicles and a decreased number of secondary follicles and antral follicles, compared with the control diet group. Therefore, these high-fat diet-induced follicular alterations might impair the fertility of dams and their female newborns. CONCLUSION The consumption of a high-fat diet causes damage to ovarian follicular development, and this commitment will persist in the next generation. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42019133865.
Collapse
Affiliation(s)
- Verônyca G Paula
- V.G. Paula, G. Vesentini, Y.K. Sinzato, R.Q. Moraes-Souza, and D.C. Damasceno are with the Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil. G.T. Volpato and R.Q. Moraes-Souza are with the Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Giovana Vesentini
- V.G. Paula, G. Vesentini, Y.K. Sinzato, R.Q. Moraes-Souza, and D.C. Damasceno are with the Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil. G.T. Volpato and R.Q. Moraes-Souza are with the Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Yuri K Sinzato
- V.G. Paula, G. Vesentini, Y.K. Sinzato, R.Q. Moraes-Souza, and D.C. Damasceno are with the Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil. G.T. Volpato and R.Q. Moraes-Souza are with the Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Rafaianne Q Moraes-Souza
- V.G. Paula, G. Vesentini, Y.K. Sinzato, R.Q. Moraes-Souza, and D.C. Damasceno are with the Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil. G.T. Volpato and R.Q. Moraes-Souza are with the Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Gustavo T Volpato
- V.G. Paula, G. Vesentini, Y.K. Sinzato, R.Q. Moraes-Souza, and D.C. Damasceno are with the Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil. G.T. Volpato and R.Q. Moraes-Souza are with the Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| | - Débora C Damasceno
- V.G. Paula, G. Vesentini, Y.K. Sinzato, R.Q. Moraes-Souza, and D.C. Damasceno are with the Laboratory of Experimental Research on Gynecology and Obstetrics, Gynecology, Postgraduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil. G.T. Volpato and R.Q. Moraes-Souza are with the Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso, Brazil
| |
Collapse
|
6
|
Gonadal development and sex determination in mouse. Reprod Biol 2020; 20:115-126. [DOI: 10.1016/j.repbio.2020.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 01/21/2020] [Accepted: 01/25/2020] [Indexed: 12/18/2022]
|
7
|
Santamaría CG, Abud JE, Luque EH, Kass L, Rodríguez HA. Culturing Rat Whole Ovary for UV Filter Benzophenone-3 Treatment. ACTA ACUST UNITED AC 2019; 82:e89. [PMID: 31765519 DOI: 10.1002/cptx.89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We describe a detailed protocol to establish a newborn rat whole ovary culture, which enables the study of direct effects (independent of hypothalamic-pituitary-gonadal axis) of endocrine disrupting chemicals (EDCs), such as benzophenone-3 (BP-3). This method is useful to understand changes in follicle formation, primordial to primary transition, and expression of regulatory molecules linked to these processes and also provides an alternative to animal models. © 2019 by John Wiley & Sons, Inc. Basic Protocol 1: Rat ovarian surgery Basic Protocol 2: Whole organ/ovarian culture Basic Protocol 3: RNA isolation and quantitative real-time PCR Basic Protocol 4: Histological processing and staining.
Collapse
Affiliation(s)
- Clarisa Guillermina Santamaría
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina
| | - Julián Elías Abud
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina
| | - Enrique Hugo Luque
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina
| | - Laura Kass
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina
| | - Horacio Adolfo Rodríguez
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral (UNL), Santa Fe, Argentina
| |
Collapse
|
8
|
Santamaría C, Abud J, Porporato M, Meyer N, Zenclussen A, Kass L, Rodríguez H. The UV filter benzophenone 3, alters early follicular assembly in rat whole ovary cultures. Toxicol Lett 2019; 303:48-54. [DOI: 10.1016/j.toxlet.2018.12.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 10/27/2022]
|
9
|
Jazwiec PA, Li X, Matushewski B, Richardson BS, Sloboda DM. Fetal Growth Restriction Is Associated With Decreased Number of Ovarian Follicles and Impaired Follicle Growth in Young Adult Guinea Pig Offspring. Reprod Sci 2019; 26:1557-1567. [PMID: 30744513 DOI: 10.1177/1933719119828041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The mechanisms mediating the impacts of fetal growth restriction (FGR) on follicular development are commonly studied in mouse/rat models, where ovarian development occurs largely during the early postnatal period. These models have shown that FGR is associated with premature follicle loss, early pubertal onset, and accelerated ovarian aging. Whether the same occurs in precocious species is unknown. OBJECTIVE Since guinea pig follicle development occurs in utero in a manner consistent with human ovarian development, we sought to determine whether FGR had similar impacts on guinea pig ovarian development. METHODS Dunkin-Hartley guinea pig dams were randomized to receive a control (CON) or a nutrient-restricted diet (FGR) prior to conception until weaning. Offspring ovaries were collected at prepubertal (postnatal day [P] 25) and young adult (P110) time points. RESULTS Prepubertal offspring exposed to FGR showed little differences in ovarian transcript levels and follicle counts. Young adult FGR offspring, however, showed reductions in the number of transitioning, primary, and antral follicles, as well as corpora lutea. This loss in follicles was associated with reduced insulin-like growth factor receptor and growth differentiation factor-9 messenger RNA levels in FGR P110 offspring compared to CON. CONCLUSION We demonstrate that FGR in guinea pigs is accompanied by perturbations in signaling pathways essential for proper follicle growth and manifests as reductions in growing follicles in offspring, but these changes do not manifest until postpuberty. These data support the fact that accelerated reproductive maturation/aging is a conserved phenotype that is associated with in utero nutritional adversity.
Collapse
Affiliation(s)
- Patrycja A Jazwiec
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Xinglin Li
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Brad Matushewski
- Department of Obstetrics and Gynecology, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, University of Western Ontario, London, Ontario, Canada
- Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Bryan S Richardson
- Department of Obstetrics and Gynecology, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, University of Western Ontario, London, Ontario, Canada
- Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- Departments of Pediatrics and Obstetrics and Gynecology, McMaster University, Hamilton, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| |
Collapse
|
10
|
Zhou Z, Lin Q, Xu X, Illahi GS, Dong C, Wu X. Maternal high-fat diet impairs follicular development of offspring through intraovarian kisspeptin/GPR54 system. Reprod Biol Endocrinol 2019; 17:13. [PMID: 30670046 PMCID: PMC6343291 DOI: 10.1186/s12958-019-0457-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/14/2019] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Excessive gestational weight gain (GWG), which is associated with adverse long-term effects on the health of the offspring, has become a major clinical problem. Accumulating evidence indicates that the ovary kisspeptin/GPR54 system directly participates in a series of physiological activities. We used a model of high-fat diet (HFD) during gestational to investigate offspring's ovarian function and whether kisspeptin/GPR54 system is involved. METHODS After introducing the male and confirmation of mating by checking a vaginal sperm plug, female rats were randomized into two groups: control diet called NCD group and high-fat diet called HFD group. After birth, all rats were changed into a control diet and litter size was adjusted to 12 pups per litter. Ovaries were collected for assessment at postnatal day (PND) 4 and PND 30. The timing of vaginal opening was recorded, and the estrous cyclicity was monitored for 2 consecutive weeks immediately. Primary granulosa cells and ovaries which were taken from PND 4 were collected for determination of the direct effect of kisspeptin-10 (kp-10) in vitro. RESULTS Neonatal rats exposed to HFD during gestation had a lower number of secondary follicles in the ovary. The expression of follicle-stimulating hormone receptor (FSHR) and kisspeptin was not altered. At prepuberty, the number of antral follicles and preovulatory follicles was elevated with decreased type III follicles in the HFD group. While the expression of ovulation-related genes was decreased, the expression levels of follicular growth-related genes and steroidogenesis synthesis related genes were elevated. A significant increase in kiss1 mRNA and kisspeptin protein was detected without changes in kiss1r mRNA and GPR54. Maternal high-fat diet during gestation resulted in a significant advanced puberty onset and an irregular estrous cycle in offspring rats. In addition, the administration of kp-10 produced an increase in viability of primary granulosa cells and enlarged the size of oocytes. CONCLUSIONS HFD exposure during maternal gestation had a long-term effect on reproductive function in the offspring and the increased ovarian kisspeptin/GPR54 system might be involved.
Collapse
Affiliation(s)
- Zhiyang Zhou
- 0000 0001 0472 9649grid.263488.3Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, 518055 Guangdong China
- 0000 0001 0472 9649grid.263488.3Shenzhen University Clinical Medical Academy, Shenzhen, 518055 Guangdong China
- 0000 0004 1808 0918grid.414906.eDepartment of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Qi Lin
- 0000 0004 1808 0918grid.414906.eDepartment of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Xinxin Xu
- 0000 0004 1808 0918grid.414906.eDepartment of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Gaby Sukma Illahi
- 0000 0004 1808 0918grid.414906.eDepartment of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Chenle Dong
- 0000 0004 1808 0918grid.414906.eDepartment of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Xueqing Wu
- 0000 0001 0472 9649grid.263488.3Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, 518055 Guangdong China
- 0000 0001 0472 9649grid.263488.3Shenzhen University Clinical Medical Academy, Shenzhen, 518055 Guangdong China
- 0000 0004 1808 0918grid.414906.eDepartment of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| |
Collapse
|
11
|
Morrison JL, Botting KJ, Darby JRT, David AL, Dyson RM, Gatford KL, Gray C, Herrera EA, Hirst JJ, Kim B, Kind KL, Krause BJ, Matthews SG, Palliser HK, Regnault TRH, Richardson BS, Sasaki A, Thompson LP, Berry MJ. Guinea pig models for translation of the developmental origins of health and disease hypothesis into the clinic. J Physiol 2018; 596:5535-5569. [PMID: 29633280 PMCID: PMC6265540 DOI: 10.1113/jp274948] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/19/2018] [Indexed: 12/12/2022] Open
Abstract
Over 30 years ago Professor David Barker first proposed the theory that events in early life could explain an individual's risk of non-communicable disease in later life: the developmental origins of health and disease (DOHaD) hypothesis. During the 1990s the validity of the DOHaD hypothesis was extensively tested in a number of human populations and the mechanisms underpinning it characterised in a range of experimental animal models. Over the past decade, researchers have sought to use this mechanistic understanding of DOHaD to develop therapeutic interventions during pregnancy and early life to improve adult health. A variety of animal models have been used to develop and evaluate interventions, each with strengths and limitations. It is becoming apparent that effective translational research requires that the animal paradigm selected mirrors the tempo of human fetal growth and development as closely as possible so that the effect of a perinatal insult and/or therapeutic intervention can be fully assessed. The guinea pig is one such animal model that over the past two decades has demonstrated itself to be a very useful platform for these important reproductive studies. This review highlights similarities in the in utero development between humans and guinea pigs, the strengths and limitations of the guinea pig as an experimental model of DOHaD and the guinea pig's potential to enhance clinical therapeutic innovation to improve human health.
Collapse
Affiliation(s)
- Janna L. Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health ResearchUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Kimberley J. Botting
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Jack R. T. Darby
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health ResearchUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Anna L. David
- Research Department of Maternal Fetal Medicine, Institute for Women's HealthUniversity College LondonLondonUK
| | - Rebecca M. Dyson
- Department of Paediatrics & Child Health and Centre for Translational PhysiologyUniversity of OtagoWellingtonNew Zealand
| | - Kathryn L. Gatford
- Robinson Research Institute and Adelaide Medical SchoolUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Clint Gray
- Department of Paediatrics & Child Health and Centre for Translational PhysiologyUniversity of OtagoWellingtonNew Zealand
| | - Emilio A. Herrera
- Pathophysiology Program, Biomedical Sciences Institute (ICBM), Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jonathan J. Hirst
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Biomedical Sciences and PharmacyUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Bona Kim
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
| | - Karen L. Kind
- School of Animal and Veterinary SciencesUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Bernardo J. Krause
- Division of Paediatrics, Faculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | | | - Hannah K. Palliser
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Biomedical Sciences and PharmacyUniversity of NewcastleCallaghanNew South WalesAustralia
| | - Timothy R. H. Regnault
- Departments of Obstetrics and Gynaecology, Physiology and PharmacologyWestern University, and Children's Health Research Institute and Lawson Health Research InstituteLondonOntarioCanada
| | - Bryan S. Richardson
- Departments of Obstetrics and Gynaecology, Physiology and PharmacologyWestern University, and Children's Health Research Institute and Lawson Health Research InstituteLondonOntarioCanada
| | - Aya Sasaki
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
| | - Loren P. Thompson
- Department of Obstetrics, Gynecology, and Reproductive SciencesUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Mary J. Berry
- Department of Paediatrics & Child Health and Centre for Translational PhysiologyUniversity of OtagoWellingtonNew Zealand
| |
Collapse
|
12
|
Ur Rehman Z, Khan FA, Farmanullah, Talpur HS, Liu Q, Liu S, Yang L. Transcriptome profiling of anti-müllerian hormone treated preantral/small antral mouse ovary follicles. Oncotarget 2018; 9:30253-30267. [PMID: 30100987 PMCID: PMC6084392 DOI: 10.18632/oncotarget.25572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 05/14/2018] [Indexed: 12/01/2022] Open
Abstract
The predisposition for the initiation of folliculogenesis in mammals including humans is programmed to start at fetal life and continues until reproductive capacity. The follicles grow from a pool of primordial follicles which retain the major functions in the entire reproductive life of a female. Anti-müllerian hormone (AMH), a glycoprotein belonging to the transforming growth factor-beta family, has an inhibitory effect on ovarian follicle development. The key regulatory target genes in primordial follicle development are of paramount importance in reproductive biology of female. A systems biology method was used to find regulatory genes performing critical role in primordial follicle development. A complete in-depth bioinformatics analysis was performed to investigate the changes in transcriptome of preantral to small antral mouse follicles treated for 12 h and 24 h with two different concentrations; 50 and 200 ng/ml of AMH, and thereby identify candidate genes in time and concentration manner. Firstly, we found differentially expressed genes that were time and concentration dependent in response to AMH. The network analysis of these differentially expressed genes provided new candidate genes and pathways associated with inhibitory action of AMH on the primordial follicle development. To further emphasize the function of AMH, the key identified genes’ protein-protein docking was analyzed and found the intracellular and extracellular protein-protein interaction. This study elucidates one of the novel mechanisms of AMH involvement in inhibition of ovarian follicle development which may lead to prolong productive life in female.
Collapse
Affiliation(s)
- Zia Ur Rehman
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China.,Department of Animal Health, Faculty of Animal Husbandry and Veterinary Sciences, The University of Agriculture, Peshawar, Pakistan
| | - Faheem Ahmad Khan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China.,The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Farmanullah
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Hira Sajjad Talpur
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Qing Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, People's Republic of China
| | - Shenhe Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| | - Liguo Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, People's Republic of China
| |
Collapse
|
13
|
Martinez-Pinto J, Piquer B, Tiszavari M, Lara H. Neonatal exposure to estradiol valerate reprograms the rat ovary androgen receptor and anti-Müllerian hormone to a polycystic ovary phenotype. Reprod Toxicol 2018; 75:127-135. [DOI: 10.1016/j.reprotox.2017.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 09/14/2017] [Accepted: 10/17/2017] [Indexed: 01/29/2023]
|
14
|
Laffan SB, Posobiec LM, Uhl JE, Vidal JD. Species Comparison of Postnatal Development of the Female Reproductive System. Birth Defects Res 2017; 110:163-189. [PMID: 29243395 DOI: 10.1002/bdr2.1132] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 01/26/2023]
Abstract
The postnatal development of the female reproductive system in laboratory animals and humans is reviewed. To enable a meaningful species comparison of the developing female reproductive system, common definitions of developmental processes were established with a focus made on aspects that are similar across species. A species comparison of the key endocrine, morphologic, and functional (onset of ovarian cycles and ability to reproduce) features of postnatal development of the female reproductive system is provided for human, nonhuman primate, dog, rat, and also mouse, minipig, and rabbit where possible. Species differences in the timing and control of female sexual maturation are highlighted. Additionally, a species comparison of the type and timing of female reproductive ovarian cycles was compiled. Human development provided the frame of reference, and then other common laboratory species were compared. The comparison has inherent challenges because the processes involved and sequence of events can differ greatly across species. Broad strokes were taken to assign a particular average age to an event and are to be used with caution. Methods of evaluation of postnatal female reproductive development in laboratory animals are discussed. Lastly, control rodent data from one of the author's laboratory on vaginal opening, first estrus, estrous cyclicity, and the histopathology involved with the developing female rat and mouse are presented. The information provided in this review is intended to be a resource for the design and interpretation of juvenile animal toxicity testing and ultimately, the relevance of the data to characterize potential risks for women and girls. Birth Defects Research 110:163-189, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Susan B Laffan
- GlaxoSmithKline Research & Development, King of Prussia, Pennsylvania
| | | | - Jenny E Uhl
- GlaxoSmithKline Research & Development, King of Prussia, Pennsylvania
| | | |
Collapse
|
15
|
Wang C, Zhou B, Xia G. Mechanisms controlling germline cyst breakdown and primordial follicle formation. Cell Mol Life Sci 2017; 74:2547-2566. [PMID: 28197668 PMCID: PMC11107689 DOI: 10.1007/s00018-017-2480-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 12/11/2022]
Abstract
In fetal females, oogonia proliferate immediately after sex determination. The progress of mitosis in oogonia proceeds so rapidly that the incompletely divided cytoplasm of the sister cells forms cysts. The oogonia will then initiate meiosis and arrest at the diplotene stage of meiosis I, becoming oocytes. Within each germline cyst, oocytes with Balbiani bodies will survive after cyst breakdown (CBD). After CBD, each oocyte is enclosed by pre-granulosa cells to form a primordial follicle (PF). Notably, the PF pool formed perinatally will be the sole lifelong oocyte source of a female. Thus, elucidating the mechanisms of CBD and PF formation is not only meaningful for solving mysteries related to ovarian development but also contributes to the preservation of reproduction. However, the mechanisms that regulate these phenomena are largely unknown. This review summarizes the progress of cellular and molecular research on these processes in mice and humans.
Collapse
Affiliation(s)
- Chao Wang
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Bo Zhou
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Guoliang Xia
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
16
|
Abstract
Evaluation of the female reproductive system in a general toxicity setting can be challenging for the toxicologic pathologist due to the cyclic nature of the estrous and menstrual cycles, timing of puberty and reproductive senescence, and species differences. Age in particular can have a significant impact on the histologic appearance of the female reproductive system and create challenges when trying to distinguish test article-related findings from normal developmental or senescent changes. This review describes the key physiologic and histologic features of immaturity, the transition through puberty, sexual maturity, and reproductive senescence in the female reproductive system, with an emphasis on practical applications for the toxicologic pathologist, and includes recommendations for distinguishing and documenting these developmental periods. Rats and cynomolgus monkeys are used as examples throughout with correlations to clinically observed end points to better aid the toxicologic pathologist in understanding how age may impact study interpretation.
Collapse
|
17
|
Zubeldia-Brenner L, Roselli CE, Recabarren SE, Gonzalez Deniselle MC, Lara HE. Developmental and Functional Effects of Steroid Hormones on the Neuroendocrine Axis and Spinal Cord. J Neuroendocrinol 2016; 28:10.1111/jne.12401. [PMID: 27262161 PMCID: PMC4956521 DOI: 10.1111/jne.12401] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 06/03/2016] [Accepted: 06/03/2016] [Indexed: 12/13/2022]
Abstract
This review highlights the principal effects of steroid hormones at central and peripheral levels in the neuroendocrine axis. The data discussed highlight the principal role of oestrogens and testosterone in hormonal programming in relation to sexual orientation, reproductive and metabolic programming, and the neuroendocrine mechanism involved in the development of polycystic ovary syndrome phenotype. Moreover, consistent with the wide range of processes in which steroid hormones take part, we discuss the protective effects of progesterone on neurodegenerative disease and the signalling mechanism involved in the genesis of oestrogen-induced pituitary prolactinomas.
Collapse
Affiliation(s)
- L Zubeldia-Brenner
- Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Argentina
| | - C E Roselli
- Department of Physiology and Pharmacology, Oregon Health and Science University Portland, Portland, OR, USA
| | - S E Recabarren
- Laboratory of Animal Physiology and Endocrinology, Faculty of Veterinary Sciences, University of Concepcion, Chillán, Chile
| | - M C Gonzalez Deniselle
- Instituto de Biología y Medicina Experimental-CONICET, Buenos Aires, Argentina
- Departamento de Ciencias Fisiológicas, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - H E Lara
- Laboratory of Neurobiochemistry Department of Biochemistry and Molecular Biology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| |
Collapse
|
18
|
Zhao L, Du X, Huang K, Zhang T, Teng Z, Niu W, Wang C, Xia G. Rac1 modulates the formation of primordial follicles by facilitating STAT3-directed Jagged1, GDF9 and BMP15 transcription in mice. Sci Rep 2016; 6:23972. [PMID: 27050391 PMCID: PMC4822123 DOI: 10.1038/srep23972] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/01/2016] [Indexed: 11/17/2022] Open
Abstract
The size of the primordial follicle pool determines the reproductive potential of mammalian females, and establishment of the pool is highly dependent on specific genes expression. However, the molecular mechanisms by which the essential genes are regulated coordinately to ensure primordial follicle assembly remain a mystery. Here, we show that the small GTPase Rac1 plays an indispensable role in controlling the formation of primordial follicles in mouse ovary. Employing fetal mouse ovary organ culture system, we demonstrate that disruption of Rac1 retarded the breakdown of germline cell cysts while Rac1 overexpression accelerated the formation of primordial follicles. In addition, in vivo inhibitor injection resulted in the formation of multi-oocyte follicles. Subsequent investigation showed that Rac1 induced nuclear import of STAT3 by physical binding. In turn, nuclear STAT3 directly activated the transcription of essential oocyte-specific genes, including Jagged1, GDF9, BMP15 and Nobox. Further, GDF9 and BMP15 regulated the translation of Notch2 via mTORC1 activation in pregranulosa cells. Overexression or addition of Jagged1, GDF9 and BMP15 not only reversed the effect of Rac1 disruption, but also accelerated primordial follicle formation via Notch2 signaling activation. Collectively, these results indicate that Rac1 plays important roles as a key regulator in follicular assembly.
Collapse
Affiliation(s)
- Lihua Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Xinhua Du
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Kun Huang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Tuo Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Zhen Teng
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Wanbao Niu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
19
|
Tsoulis MW, Chang PE, Moore CJ, Chan KA, Gohir W, Petrik JJ, Vickers MH, Connor KL, Sloboda DM. Maternal High-Fat Diet-Induced Loss of Fetal Oocytes Is Associated with Compromised Follicle Growth in Adult Rat Offspring. Biol Reprod 2016; 94:94. [PMID: 26962114 PMCID: PMC4861169 DOI: 10.1095/biolreprod.115.135004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 02/25/2016] [Indexed: 11/12/2022] Open
Abstract
Maternal obesity predisposes offspring to metabolic and reproductive dysfunction. We have shown previously that female rat offspring born to mothers fed a high-fat (HF) diet throughout pregnancy and lactation enter puberty early and display aberrant reproductive cyclicity. The mechanisms driving this reproductive phenotype are currently unknown thus we investigated whether changes in ovarian function were involved. Wistar rats were mated and randomized to: dams fed a control diet (CON) or dams fed a HF diet from conception until the end of lactation (HF). Ovaries were collected from fetuses at Embryonic Day (E) 20, and neonatal ovaries at Day 4 (P4), prepubertal ovaries at P27 and adult ovaries at P120. In a subset of offspring, the effects of a HF diet fed postweaning were evaluated. The present study shows that fetuses of mothers fed a HF diet had significantly fewer oocytes at E20, and in neonates, have reduced AMH signaling that may facilitate an increased number of assembled primordial follicles. Both prepubertally and in adulthood, ovaries show increased follicular atresia. As adults, offspring have reduced FSH responsiveness, low expression levels of estrogen receptor alpha (Eralpha), the oocyte-secreted factor, Gdf9, oocyte-specific RNA binding protein, Dazl, and high expression levels of the granulosa-cell derived factor, AMH, in antral follicles. Together, these data suggest that ovarian compromise in offspring born to HF-fed mothers may arise from changes already observable in the fetus and neonate and in the long term, associated with increased follicular atresia through adulthood.
Collapse
Affiliation(s)
- Michael W Tsoulis
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Pauline E Chang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Caroline J Moore
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Kaitlyn A Chan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Wajiha Gohir
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - James J Petrik
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Mark H Vickers
- Liggins Institute and Gravida, National Centre for Growth and Development, University of Auckland, Aukland, New Zealand
| | - Kristin L Connor
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
20
|
Silber S. How ovarian transplantation works and how resting follicle recruitment occurs: a review of results reported from one center. ACTA ACUST UNITED AC 2016; 12:217-27. [PMID: 26900727 DOI: 10.2217/whe.15.108] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Ovarian freezing and transplantation has garnered increasing interest as a potential way of preserving fertility in cancer patients. This special report aims to identify the success rate of frozen compared with fresh ovarian cortex transplantation (in one single series from one center for the sake of consistency), as well as potentially provides insight into the mechanism behind ovarian follicle recruitment. A comparison of fresh versus frozen transplantation techniques is presented, highlighting the similarity and differences between the fresh and frozen transplantation procedures. Much of the literature is scattered case reports with different patient populations and different techniques. This represents an effort to simplify and popularize an approach that has yielded favorable results (all cases recovered ovulation and 75% had successful spontaneous pregnancy) in one single, disciplined study.
Collapse
Affiliation(s)
- Sherman Silber
- Infertility Center of St. Louis, 224 S. Woods Mill Road, Suite 730, St. Louis, MO 63017, USA.,Department of Obstetrics & Gynecology, University of Amsterdam, 1012 WX Amsterdam, The Netherlands.,Sun Yat-Sen Medical School, No.74 Zhongshan Rd.2, Guangzhou, P.R. China
| |
Collapse
|
21
|
Bertoldo MJ, Bernard J, Duffard N, Tsikis G, Alves S, Calais L, Uzbekova S, Monniaux D, Mermillod P, Locatelli Y. Inhibitors of c-Jun phosphorylation impede ovine primordial follicle activation. Mol Hum Reprod 2016; 22:338-49. [PMID: 26908644 DOI: 10.1093/molehr/gaw012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/20/2016] [Indexed: 12/23/2022] Open
Abstract
STUDY HYPOTHESIS Is the c-Jun-N-terminal kinase (JNK) pathway implicated in primordial follicle activation? STUDY FINDING Culture of ovine ovarian cortex in the presence of two different c-Jun phosphorylation inhibitors impeded pre-antral follicle activation. WHAT IS KNOWN ALREADY Despite its importance for fertility preservation therapies, the mechanisms of primordial follicle activation are poorly understood. Amongst different signalling pathways potentially involved, the JNK pathway has been previously shown to be essential for cell cycle progression and pre-antral follicle development in mice. STUDY DESIGN, SAMPLES/MATERIALS, METHODS Ovine ovarian cortex pieces were cultured with varying concentrations of SP600125, JNK inhibitor VIII or anti-Mullerian hormone (AMH) in the presence of FSH for 9 days. Follicular morphometry and immunohistochemistry for proliferating cell nuclear antigen (PCNA), apoptosis and follicle activation (Foxo3a) were assessed. MAIN RESULTS AND THE ROLE OF CHANCE Inhibition of primordial follicle activation occurred in the presence of SP600125, JNK inhibitor VIII and AMH when compared with controls (all P < 0.05) after 2 days of culture. However, only in the highest concentrations used was the inhibition of activation associated with induction of follicular apoptosis (P < 0.05). In growing follicles, PCNA antigen expression was reduced when the JNK inhibitors or AMH were used (P < 0.05 versus control), indicating reduced proliferation of the somatic compartment. LIMITATIONS, REASONS FOR CAUTION Although we evaluated the effects of inhibition of c-Jun phosphorylation on primordial follicle development, we did not determine the cellular targets and mechanism of action of the inhibitors. WIDER IMPLICATIONS OF THE FINDINGS These results are the first to implicate the JNK pathway in primordial follicle activation and could have significant consequences for the successful development of fertility preservation strategies and our understanding of primordial follicle activation. LARGE SCALE DATA n/a. STUDY FUNDING AND COMPETING INTERESTS Dr Michael J. Bertoldo and the laboratories involved in the present study were supported by a grant from 'Région Centre' (CRYOVAIRE, Grant number #320000268). There are no conflicts of interest to declare.
Collapse
Affiliation(s)
- Michael J Bertoldo
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly 37380, France School of Women's and Children's Health, Discipline of Obstetrics and Gynaecology, University of New South Wales, Sydney, Australia
| | - Jérémy Bernard
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly 37380, France MNHN, Laboratoire de la Réserve de la Haute Touche, Obterre 36290, France
| | - Nicolas Duffard
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly 37380, France MNHN, Laboratoire de la Réserve de la Haute Touche, Obterre 36290, France
| | - Guillaume Tsikis
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly 37380, France CNRS, UMR7247, Nouzilly 37380, France Université François Rabelais de Tours, Tours 37041, France IFCE, Nouzilly 37380, France
| | - Sabine Alves
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly 37380, France CNRS, UMR7247, Nouzilly 37380, France Université François Rabelais de Tours, Tours 37041, France IFCE, Nouzilly 37380, France
| | - Laure Calais
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly 37380, France
| | - Svetlana Uzbekova
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly 37380, France CNRS, UMR7247, Nouzilly 37380, France Université François Rabelais de Tours, Tours 37041, France IFCE, Nouzilly 37380, France
| | - Danielle Monniaux
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly 37380, France CNRS, UMR7247, Nouzilly 37380, France Université François Rabelais de Tours, Tours 37041, France IFCE, Nouzilly 37380, France
| | - Pascal Mermillod
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly 37380, France CNRS, UMR7247, Nouzilly 37380, France Université François Rabelais de Tours, Tours 37041, France IFCE, Nouzilly 37380, France
| | - Yann Locatelli
- INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly 37380, France MNHN, Laboratoire de la Réserve de la Haute Touche, Obterre 36290, France CNRS, UMR7247, Nouzilly 37380, France Université François Rabelais de Tours, Tours 37041, France IFCE, Nouzilly 37380, France
| |
Collapse
|
22
|
El-Hayek S, Clarke HJ. Control of Oocyte Growth and Development by Intercellular Communication Within the Follicular Niche. Results Probl Cell Differ 2016; 58:191-224. [PMID: 27300180 DOI: 10.1007/978-3-319-31973-5_8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In the mammalian ovary, each oocyte grows and develops within its own structural and developmental niche-the follicle. Together with the female germ cell in the follicle are somatic granulosa cells, specialized companion cells that surround the oocyte and provide support to it, and an outer layer of thecal cells that serve crucial roles including steroid synthesis. These follicular compartments function as a single physiological unit whose purpose is to produce a healthy egg, which upon ovulation can be fertilized and give rise to a healthy embryo, thus enabling the female germ cell to fulfill its reproductive potential. Beginning from the initial stage of follicle formation and until terminal differentiation at ovulation, oocyte and follicle growth depend absolutely on cooperation between the different cellular compartments. This cooperation synchronizes the initiation of oocyte growth with follicle activation. During growth, it enables metabolic support for the follicle-enclosed oocyte and allows the follicle to fulfill its steroidogenic potential. Near the end of the growth period, intra-follicular interactions prevent the precocious meiotic resumption of the oocyte and ensure its nuclear differentiation. Finally, cooperation enables the events of ovulation, including meiotic maturation of the oocyte and expansion of the cumulus granulosa cells. In this chapter, we discuss the cellular interactions that enable the growing follicle to produce a healthy oocyte, focusing on the communication between the germ cell and the surrounding granulosa cells.
Collapse
Affiliation(s)
- Stephany El-Hayek
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
- Department of Biology, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Block E-M0.2218, Montreal, QC, Canada, H4A 3J1
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada.
- Department of Biology, McGill University, Montreal, QC, Canada.
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Block E-M0.2218, Montreal, QC, Canada, H4A 3J1.
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
23
|
Unifying theory of adult resting follicle recruitment and fetal oocyte arrest. Reprod Biomed Online 2015; 31:472-5. [DOI: 10.1016/j.rbmo.2015.06.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/26/2015] [Accepted: 06/30/2015] [Indexed: 01/01/2023]
|
24
|
Yaba A, Ordueri NEG, Tanriover G, Sahin P, Demir N, Celik-Ozenci C. Expression of CCM2 and CCM3 during mouse gonadogenesis. J Assist Reprod Genet 2015; 32:1497-507. [PMID: 26386873 DOI: 10.1007/s10815-015-0559-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/20/2015] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Three cerebral cavernous malformation (CCM) proteins, CCM1, CCM2, and CCM3, regulate cell-cell adhesion, cell shape and polarity, and most likely cell adhesion to extracellular matrix. Recently, CCM2 and CCM3 are known to be expressed in control and varicocele-induced rat testes, but little is known about these proteins during gonadogenesis. This led us to study the CCM proteins during the mouse gonadogenesis. METHODS Neonatal (PND 0), postnatal, and adult mice testes and ovaries were obtained from mice. CCM2 and CCM3 expression were analyzed during mouse testicular and ovarian development by immunohistochemistry and quantitative real-time PCR. RESULTS The results showed that in both sexes, Ccm2 and Ccm3 mRNA and protein were first detectable after gonadogenesis when the gonads were well differentiated and remained present until the adult stage. In the testis, CCM2 and CCM3 expression were restricted to the nuclei of Sertoli cells, suggesting a conserved role in testicular differentiation. In the ovary, the CCM2 and CCM3 proteins were localized in the cytoplasm of oocytes, suggesting an unexpected role during oogenesis. Quantitative real-time PCR (qRT-PCR) results showed that expression of Ccm2 and Ccm3 genes could play a role in the regulation of mouse gonadogenesis translational activation upon testicular and ovarian development. CONCLUSIONS The localization of CCM2 and CCM3 proteins show their different functions for CCM2 and CCM3 which may have important roles in testicular and ovarian differentiation. In conclusion, CCM2 and CCM3 may be involved in establishing the differential expression pattern in developing mouse testis and ovary.
Collapse
Affiliation(s)
- Aylin Yaba
- Department of Histology and Embryology, İstanbul Medipol University School of Medicine, İstanbul, Turkey
| | - N Ece Gungor Ordueri
- Department of Histology and Embryology, Akdeniz University School of Medicine, 07070 Campus, Antalya, Turkey
| | - Gamze Tanriover
- Department of Histology and Embryology, Akdeniz University School of Medicine, 07070 Campus, Antalya, Turkey
| | - Pinar Sahin
- Department of Histology and Embryology, Akdeniz University School of Medicine, 07070 Campus, Antalya, Turkey
| | - Necdet Demir
- Department of Histology and Embryology, Akdeniz University School of Medicine, 07070 Campus, Antalya, Turkey
| | - Ciler Celik-Ozenci
- Department of Histology and Embryology, Akdeniz University School of Medicine, 07070 Campus, Antalya, Turkey.
| |
Collapse
|
25
|
Stanley JA, Arosh JA, Burghardt RC, Banu SK. A fetal whole ovarian culture model for the evaluation of CrVI-induced developmental toxicity during germ cell nest breakdown. Toxicol Appl Pharmacol 2015; 289:58-69. [PMID: 26348139 DOI: 10.1016/j.taap.2015.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 08/18/2015] [Accepted: 09/01/2015] [Indexed: 01/08/2023]
Abstract
Prenatal exposure to endocrine disrupting chemicals (EDCs), including bisphenol A, dioxin, pesticides, and cigarette smoke, has been linked to several ovarian diseases such as premature ovarian failure (POF) and early menopause in women. Hexavalent chromium (CrVI), one of the more toxic heavy metals, is widely used in more than 50 industries. As one of the world's leading producers of Cr compounds, the U.S. is facing growing challenges in protecting human health against adverse effects of CrVI. Our recent findings demonstrated that in vivo CrVI exposure during gestational period caused POF in F1 offspring. Our current research focus is three-fold: (i) to identify the effect of CrVI on critical windows of great vulnerability of fetal ovarian development; (ii) to understand the molecular mechanism of CrVI-induced POF; (iii) to identify potential intervention strategies to mitigate or inhibit CrVI effects. In order to accomplish these goals we used a fetal whole ovarian culture system. Fetuses were removed from the normal pregnant rats on gestational day 13.5. Fetal ovaries were cultured in vitro for 12 days, and treated with or without 0.1 ppm potassium dichromate (CrVI) from culture day 2-8, which recapitulated embryonic day 14.5-20.5, in vivo. Results showed that CrVI increased germ cell/oocyte apoptosis by increasing caspase 3, BAX, p53 and PUMA; decreasing BCL2, BMP15, GDF9 and cKIT; and altering cell cycle regulatory genes and proteins. This model system may serve as a potential tool for high throughput testing of various drugs and/or EDCs in particular to assess developmental toxicity of the ovary.
Collapse
Affiliation(s)
- Jone A Stanley
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Joe A Arosh
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Sakhila K Banu
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
26
|
Wang Z, Niu W, Wang Y, Teng Z, Wen J, Xia G, Wang C. Follistatin288 Regulates Germ Cell Cyst Breakdown and Primordial Follicle Assembly in the Mouse Ovary. PLoS One 2015; 10:e0129643. [PMID: 26076381 PMCID: PMC4468113 DOI: 10.1371/journal.pone.0129643] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 05/11/2015] [Indexed: 11/18/2022] Open
Abstract
In mammals, the primordial follicle pool represents the entire reproductive potential of a female. The transforming growth factor-β (TGF-β) family member activin (ACT) contributes to folliculogenesis, although the exact mechanism is not known. The role of FST288, the strongest ACT-neutralizing isoform of follistatin (FST), during cyst breakdown and primordial follicle formation in the fetal mice ovary was assessed using an in vitro culture system. FST was continuously expressed in the oocytes as well as the cuboidal granulosa cells of growing follicles in perinatal mouse ovaries. Treatment with FST288 delayed germ cell nest breakdown, particularly near the periphery of the ovary, and dramatically decreased the percentage of primordial follicles. In addition, there was a dramatic decrease in proliferation of granulosa cells and somatic cell expression of Notch signaling was impaired. In conclusion, FST288 impacts germ cell nest breakdown and primordial follicle assembly by inhibiting somatic cell proliferation.
Collapse
Affiliation(s)
- Zhengpin Wang
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Wanbao Niu
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Yijing Wang
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Zhen Teng
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Jia Wen
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Guoliang Xia
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Chao Wang
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
- * E-mail:
| |
Collapse
|
27
|
Silber S, Pineda J, Lenahan K, DeRosa M, Melnick J. Fresh and cryopreserved ovary transplantation and resting follicle recruitment. Reprod Biomed Online 2015; 30:643-50. [PMID: 25892498 DOI: 10.1016/j.rbmo.2015.02.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 02/12/2015] [Accepted: 02/17/2015] [Indexed: 01/01/2023]
Abstract
Ovary cryopreservation and transplantation has garnered increasing interest as a possible method to preserve fertility for cancer patients and to study ovarian resting follicle recruitment. Eleven consecutive women underwent fresh donor ovary transplantation, and 11 underwent cryopreserved ovary auto-transplantation in the same centre, with the same surgeon. Of the 11 fresh transplant recipients, who were all young but menopausal, nine women had normal ovarian cortex transplanted from an identical twin sister, and two had a fresh allograft from a non-identical sister. In the second group, 11 women with cancer had ovarian tissue cryopreserved before bone marrow transplant, and then after years of therapeutically induced menopause, underwent cryopreserved ovarian cortex autotransplantation. Recovery of ovarian function and follicle recruitment was assessed in all 22 recipients, and the potential for pregnancy was further investigated in 19 (11 fresh and 8 cryopreserved) with over 1-year follow-up. In all recipients, normal FSH levels and menstruation returned by about 150 days, and anti-Müllerian hormone reached much greater than normal concentrations by about 170 days. Anti-Müllerian hormone levels then fell below normal by about 240 days and remained at that lower level. Seventeen babies have been born to these 11 fresh and eight cryopreserved ovary transplant recipients.
Collapse
Affiliation(s)
- Sherman Silber
- Infertility Center of St Louis, St Luke's Hospital, 224 South Woods Mill Road, Saint Louis, MO 63017, USA.
| | - Jorge Pineda
- Infertility Center of St Louis, St Luke's Hospital, 224 South Woods Mill Road, Saint Louis, MO 63017, USA
| | - Kathleen Lenahan
- Infertility Center of St Louis, St Luke's Hospital, 224 South Woods Mill Road, Saint Louis, MO 63017, USA
| | - Michael DeRosa
- Infertility Center of St Louis, St Luke's Hospital, 224 South Woods Mill Road, Saint Louis, MO 63017, USA
| | - Jeffrey Melnick
- Department of Pathology, St Luke's Hospital, 232 South Woods Mill Road, St Louis, MO 63017, USA
| |
Collapse
|
28
|
Abstract
There is now considerable epidemiological and experimental evidence indicating that early-life environmental conditions, including nutrition, affect subsequent development in later life. These conditions induce highly integrated responses in endocrine-related homeostasis, resulting in persistent changes in the developmental trajectory producing an altered adult phenotype. Early-life events trigger processes that prepare the individual for particular circumstances that are anticipated in the postnatal environment. However, where the intrauterine and postnatal environments differ markedly, such modifications to the developmental trajectory may prove maladaptive in later life. Reproductive maturation and function are similarly influenced by early-life events. This should not be surprising, because the primordial follicle pool is established early in life and is thus vulnerable to early-life events. Results of clinical and experimental studies have indicated that early-life adversity is associated with a decline in ovarian follicular reserve, changes in ovulation rates, and altered age at onset of puberty. However, the underlying mechanisms regulating the relationship between the early-life developmental environment and postnatal reproductive development and function are unclear. This review examines the evidence linking early-life nutrition and effects on the female reproductive system, bringing together clinical observations in humans and experimental data from targeted animal models.
Collapse
Affiliation(s)
- K A Chan
- Departments of Biochemistry and Biomedical SciencesPediatricsObstetrics and GynecologyMcMaster University, 1280 Main Street West HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1
| | - M W Tsoulis
- Departments of Biochemistry and Biomedical SciencesPediatricsObstetrics and GynecologyMcMaster University, 1280 Main Street West HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1
| | - D M Sloboda
- Departments of Biochemistry and Biomedical SciencesPediatricsObstetrics and GynecologyMcMaster University, 1280 Main Street West HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1 Departments of Biochemistry and Biomedical SciencesPediatricsObstetrics and GynecologyMcMaster University, 1280 Main Street West HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1 Departments of Biochemistry and Biomedical SciencesPediatricsObstetrics and GynecologyMcMaster University, 1280 Main Street West HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1
| |
Collapse
|
29
|
Hummitzsch K, Anderson RA, Wilhelm D, Wu J, Telfer EE, Russell DL, Robertson SA, Rodgers RJ. Stem cells, progenitor cells, and lineage decisions in the ovary. Endocr Rev 2015; 36:65-91. [PMID: 25541635 PMCID: PMC4496428 DOI: 10.1210/er.2014-1079] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/15/2014] [Indexed: 01/05/2023]
Abstract
Exploring stem cells in the mammalian ovary has unleashed a Pandora's box of new insights and questions. Recent evidence supports the existence of stem cells of a number of the different cell types within the ovary. The evidence for a stem cell model producing mural granulosa cells and cumulus cells is strong, despite a limited number of reports. The recent identification of a precursor granulosa cell, the gonadal ridge epithelial-like cell, is exciting and novel. The identification of female germline (oogonial) stem cells is still very new and is currently limited to just a few species. Their origins and physiological roles, if any, are unknown, and their potential to produce oocytes and contribute to follicle formation in vivo lacks robust evidence. The precursor of thecal cells remains elusive, and more compelling data are needed. Similarly, claims of very small embryonic-like cells are also preliminary. Surface epithelial cells originating from gonadal ridge epithelial-like cells and from the mesonephric epithelium at the hilum of the ovary have also been proposed. Another important issue is the role of the stroma in guiding the formation of the ovary, ovigerous cords, follicles, and surface epithelium. Immune cells may also play key roles in developmental patterning, given their critical roles in corpora lutea formation and regression. Thus, while the cellular biology of the ovary is extremely important for its major endocrine and fertility roles, there is much still to be discovered. This review draws together the current evidence and perspectives on this topic.
Collapse
Affiliation(s)
- Katja Hummitzsch
- Discipline of Obstetrics and Gynaecology (K.H., D.L.R., S.A.R., R.J.R.), School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia 5005; Medical Research Council Centre for Reproductive Health (R.A.A.), The University of Edinburgh, The Queens Medical Research Institute, Edinburgh EH16 4TJ, United Kingdom; Department of Anatomy and Developmental Biology (D.W.), Monash University, Clayton, Victoria, Australia 3800; Bio-X Institutes (J.W.), Shanghai Jiao Tong University, Shanghai 200240, China; and Institute of Cell Biology and Centre for Integrative Physiology (E.E.T), The University of Edinburgh, Edinburgh EH8 9XE, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Antimüllerian hormone regulates stem cell factor expression in human granulosa cells. Fertil Steril 2014; 102:1742-50.e1. [PMID: 25241372 DOI: 10.1016/j.fertnstert.2014.08.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 08/07/2014] [Accepted: 08/08/2014] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To determine whether there is a correlation between antimüllerian hormone (AMH) and stem cell factor (SCF) in serum, follicular fluid (FF), and granulosa cells (GCs), and to investigate a possible regulatory mechanism of AMH on SCF in human granulosa cells. DESIGN Prospective clinical and experimental study. SETTING Academic center. PATIENT(S) 163 women undergoing IVF. INTERVENTION(S) Serum, FF, and GCs obtained in all women, primary cultures of human GCs. MAIN OUTCOME MEASURE(S) AMH and SCF were analyzed in serum, FF, and GCs, using enzyme-linked immunosorbent assay, reverse-transcription polymerase chain reaction, and immunoblotting. RESULT(S) There was a significant negative correlation between AMH and SCF protein level in FF, and in the mRNA expression of AMH and SCF in GCs. Conversely, there was no correlation between AMH and SCF levels in serum. In primary cultures of human GCs, SCF was down-regulated by treatment with recombinant human AMH and was increased by cyclic adenosine 3':5' monophosphate (cAMP) in a dose-dependent manner. A protein kinase A (PKA) inhibitor (H89) significantly reversed the effects of recombinant human AMH and cAMP on SCF mRNA and protein expression. CONCLUSION(S) This is the first report on a modulatory role for AMH as an ovarian/follicular autocrine/paracrine factor controlling SCF expression via the cAMP/PKA pathway.
Collapse
|
31
|
Bagavandoss P. Temporal expression of tenascin-C and type I collagen in response to gonadotropins in the immature rat ovary. Acta Histochem 2014; 116:1125-33. [PMID: 24998028 DOI: 10.1016/j.acthis.2014.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 05/23/2014] [Accepted: 05/26/2014] [Indexed: 12/25/2022]
Abstract
Ovarian morphogenesis and physiology in mammals take place in the context of hormones, paracrine factors and extracellular matrix molecules. Both fibrillar type I collagen and the multidomain tenascin-C are matrix molecules capable of modulating the behavior of both normal and neoplastic cells in many organs. Therefore, the objective of this qualitative study was to simultaneously examine the distribution of both tenascin-C and type I collagen in ovarian follicles and corpora lutea induced to develop in response to gonadotropin treatments. In preantral follicles both matrix proteins were present in the focimatrix, theca externa and the interstitium. Equine gonadotropin induced the appearance of both proteins in the theca interna. Subsequent to administration with human chorionic gonadotropin, tenascin-C appearance in the thecal capillaries preceded type I collagen expression. Tenascin-C was also observed in the capillaries of functional and regressing corpora lutea, while type I collagen was predominantly present in the interstitium and tunica albuginea. Western blots showed both an increase in and degradation of tenascin-C in the regressing corpora lutea. The ovarian surface epithelium also showed immunoreactivity for both tenascin-C and type I collagen. The study reveals that tenascin-C and type I collagen may participate in the morphogenesis of ovarian follicles, and in the formation and regression of corpora lutea.
Collapse
|
32
|
Cruz G, Foster W, Paredes A, Yi KD, Uzumcu M. Long-term effects of early-life exposure to environmental oestrogens on ovarian function: role of epigenetics. J Neuroendocrinol 2014; 26:613-24. [PMID: 25040227 PMCID: PMC4297924 DOI: 10.1111/jne.12181] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 06/22/2014] [Accepted: 07/15/2014] [Indexed: 12/14/2022]
Abstract
Oestrogens play an important role in development and function of the brain and reproductive tract. Accordingly, it is considered that developmental exposure to environmental oestrogens can disrupt neural and reproductive tract development, potentially resulting in long-term alterations in neurobehaviour and reproductive function. Many chemicals have been shown to have oestrogenic activity, whereas others affect oestrogen production and turnover, resulting in the disruption of oestrogen signalling pathways. However, these mechanisms and the concentrations required to induce these effects cannot account for the myriad adverse effects of environmental toxicants on oestrogen-sensitive target tissues. Hence, alternative mechanisms are assumed to underlie the adverse effects documented in experimental animal models and thus could be important to human health. In this review, the epigenetic regulation of gene expression is explored as a potential target of environmental toxicants including oestrogenic chemicals. We suggest that toxicant-induced changes in epigenetic signatures are important mechanisms underlying the disruption of ovarian follicular development. In addition, we discuss how exposure to environmental oestrogens during early life can alter gene expression through effects on epigenetic control potentially leading to permanent changes in ovarian physiology.
Collapse
Affiliation(s)
- Gonzalo Cruz
- Centro de Neurobiología y Plasticidad Cerebral, Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Correspondence to: Gonzalo Cruz, Gran Bretaña 1111, Playa Ancha, Valparaíso, Chile. 2360102, Tel. 56 32 2508015,
| | - Warren Foster
- Department of Obstetrics & Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Alfonso Paredes
- Laboratorio de Neurobioquímica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Chile
| | - Kun Don Yi
- Syngenta Crop Protection, LLC. Greensboro, NC
| | - Mehmet Uzumcu
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
33
|
Picut CA, Dixon D, Simons ML, Stump DG, Parker GA, Remick AK. Postnatal ovary development in the rat: morphologic study and correlation of morphology to neuroendocrine parameters. Toxicol Pathol 2014; 43:343-53. [PMID: 25107574 DOI: 10.1177/0192623314544380] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Histopathologic examination of the immature ovary is a required end point on juvenile toxicity studies and female pubertal and thyroid function assays. To aid in this evaluation and interpretation of the immature ovary, the characteristic histologic features of rat ovary through the developmental periods are described. These histologic features are correlated with published changes in neuroendocrine profiles as the hypothalamic-pituitary-gonadal axis matures. During the neonatal stage (postnatal day [PND] 0-7), ovarian follicle development is independent of pituitary gonadotropins (luteinizing hormone [LH] or follicle-stimulating hormone [FSH]), and follicles remain preantral. Antral development of "atypical" follicles occurs in the early infantile period (PND 8-14) when the ovary becomes responsive to pituitary gonadotropins. In the late infantile period (PND 15-20), the zona pellucida appears, the hilus forms, and antral follicles mature by losing their "atypical" appearance. The juvenile stage (PND 21-32) is the stage when atresia of medullary follicles occurs corresponding to a nadir in FSH levels. In the peripubertal period (PND 33-37), atresia subsides as FSH levels rebound, and LH begins its bimodal surge pattern leading to ovulation. This report will provide pathologists with baseline morphologic and endocrinologic information to aid in identification and interpretation of xenobiotic effects in the ovary of the prepubertal rat.
Collapse
Affiliation(s)
| | - Darlene Dixon
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, Division of the NTP, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | | | | | | | | |
Collapse
|
34
|
Nilsson EE, Larsen G, Skinner MK. Roles of Gremlin 1 and Gremlin 2 in regulating ovarian primordial to primary follicle transition. Reproduction 2014; 147:865-74. [PMID: 24614542 DOI: 10.1530/rep-14-0005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A network of extracellular signaling factors has previously been shown to act in concert to control the ovarian primordial to primary follicle transition. The current study was designed to investigate the roles of the endogenous bone morphogenetic protein (BMP) inhibitors Gremlin 1 (GREM1) and GREM2 in primordial follicle transition in the rat ovary. GREM1 and GREM2 treatments were found to reverse the effects of anti-Müllerian hormone (AMH) to inhibit follicle transition in a whole-ovary culture system. GREM1 reversed the effect of BMP4 to stimulate primordial follicle transition. Immunohistochemical studies showed that GREM2, but not GREM1, was present in primordial follicles suggesting that GREM2 may regulate primordial follicle transition in vivo. Co-immunoprecipitation studies indicated that GREM2 directly binds to AMH, as well as to BMP4. Transcriptome analyses of ovaries treated with GREM2 or GREM1 yielded negligible numbers of differentially expressed genes, suggesting that the immediate effects of GREM2 or GREM1 appear to be at the level of protein-protein interactions, rather than direct actions on the cells. A number of other ovarian growth factors were found to influence the expression of Grem2. Observations suggest that Grem2 is a part of the signaling network of growth factors that regulate the primordial to primary follicle transition. Insights into the regulatory networks affecting the pool of primordial follicles are important to understand the molecular basis for reproductive diseases such as primary ovarian insufficiency.
Collapse
Affiliation(s)
- Eric E Nilsson
- School of Biological SciencesCenter for Reproductive Biology, Washington State University, Pullman, Washington 99164-4236, USA
| | - Ginger Larsen
- School of Biological SciencesCenter for Reproductive Biology, Washington State University, Pullman, Washington 99164-4236, USA
| | - Michael K Skinner
- School of Biological SciencesCenter for Reproductive Biology, Washington State University, Pullman, Washington 99164-4236, USA
| |
Collapse
|
35
|
Chalmey C, Giton F, Chalmel F, Fiet J, Jégou B, Mazaud-Guittot S. Systemic compensatory response to neonatal estradiol exposure does not prevent depletion of the oocyte pool in the rat. PLoS One 2013; 8:e82175. [PMID: 24358151 PMCID: PMC3864944 DOI: 10.1371/journal.pone.0082175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 10/21/2013] [Indexed: 11/19/2022] Open
Abstract
The formation of ovarian follicles is a finely tuned process that takes place within a narrow time-window in rodents. Multiple factors and pathways have been proposed to contribute to the mechanisms triggering this process but the role of endocrine factors, especially estrogens, remains elusive. It is currently hypothesized that removal from the maternal hormonal environment permits follicle formation at birth. However, experimentally-induced maintenance of high 17β-estradiol (E2) levels leads to subtle, distinct, immediate effects on follicle formation and oocyte survival depending on the species and dose. In this study, we examined the immediate effects of neonatal E2 exposure from post-natal day (PND) 0 to PND2 on the whole organism and on ovarian follicle formation in rats. Measurements of plasma E2, estrone and their sulfate conjugates after E2 exposure showed that neonatal female rats rapidly acquire the capability to metabolize and clear excessive E2 levels. Concomitant modifications to the mRNA content of genes encoding selected E2 metabolism enzymes in the liver and the ovary in response to E2 exposure indicate that E2 may modify the neonatal maturation of these organs. In the liver, E2 treatment was associated with lower acquisition of the capability to metabolize E2. In the ovary, E2 depleted the oocyte pool in a dose dependent manner by PND3. In 10 µg/day E2-treated ovaries, apoptotic oocytes were observed in newly formed follicles in addition to areas of ovarian cord remodeling. At PND6, follicles without any visible oocyte were present and multi-oocyte follicles were not observed. Our study reveals a major species-difference. Indeed, neonatal exposure to E2 depletes the oocyte pool in the rat ovary, whereas in the mouse it is well known to increase oocyte survival.
Collapse
Affiliation(s)
- Clémentine Chalmey
- Institut National de la Santé et de la Recherche Médicale, Unité 1085 Institut de Recherche en Santé Environnement et Travail, Institut Fédératif de Recherche 140, Université de Rennes 1, Rennes, France
| | - Franck Giton
- AP-HP, Hôpital H. Mondor - A. Chenevier, service de Biochimie et de Génétique, Créteil, France
- Institut National de la Santé et de la Recherche Médicale, U955 Équipe 07, Créteil, France
| | - Frédéric Chalmel
- Institut National de la Santé et de la Recherche Médicale, Unité 1085 Institut de Recherche en Santé Environnement et Travail, Institut Fédératif de Recherche 140, Université de Rennes 1, Rennes, France
| | - Jean Fiet
- Institut National de la Santé et de la Recherche Médicale, U955 Équipe 07, Créteil, France
| | - Bernard Jégou
- Institut National de la Santé et de la Recherche Médicale, Unité 1085 Institut de Recherche en Santé Environnement et Travail, Institut Fédératif de Recherche 140, Université de Rennes 1, Rennes, France
- Ecole des Hautes Études en Santé Publique, Rennes, France
| | - Séverine Mazaud-Guittot
- Institut National de la Santé et de la Recherche Médicale, Unité 1085 Institut de Recherche en Santé Environnement et Travail, Institut Fédératif de Recherche 140, Université de Rennes 1, Rennes, France
- * E-mail:
| |
Collapse
|
36
|
Jorgensen JS. Defining the neighborhoods that escort the oocyte through its early life events and into a functional follicle. Mol Reprod Dev 2013; 80:960-76. [PMID: 24105719 PMCID: PMC3980676 DOI: 10.1002/mrd.22232] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 08/15/2013] [Indexed: 01/19/2023]
Abstract
The ovary functions to chaperone the most precious cargo for female individuals, the oocyte, thereby allowing the passage of genetic material to subsequent generations. Within the ovary, single oocytes are surrounded by a legion of granulosa cells inside each follicle. These two cell types depend upon one another to support follicle formation and oocyte survival. The infrastructure and events that work together to ultimately form these functional follicles within the ovary are unprecedented, given that the oocyte originates as a cell like all other neighboring cells within the embryo prior to gastrulation. This review discusses the journey of the germ cell in the context of the developing female mouse embryo, with a focus on specific signaling events and cell-cell interactions that escort the primordial germ cell as it is specified into the germ cell fate, migrates through the hindgut into the gonad, differentiates into an oocyte, and culminates upon formation of the primordial and then primary follicle.
Collapse
Affiliation(s)
- Joan S Jorgensen
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
37
|
McFee RM, Cupp AS. Vascular contributions to early ovarian development: potential roles of VEGFA isoforms. Reprod Fertil Dev 2013; 25:333-42. [PMID: 23021322 DOI: 10.1071/rd12134] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 08/21/2012] [Indexed: 12/25/2022] Open
Abstract
Vascularisation is an essential component of ovarian morphogenesis; however, little is known regarding factors regulating the establishment of vasculature in the ovary. Angiogenesis involving extensive endothelial cell migration is a critical component of vessel formation in the embryonic testis but vasculogenic mechanisms appear to play a prominent role in ovarian vascularisation. Vasculature has a strong influence on the formation of ovarian structures, and the early developmental processes of ovigerous cord formation, primordial follicle assembly and follicle activation are all initiated in regions of the ovary that are in close association with the highly vascular medulla. The principal angiogenic factor, vascular endothelial growth factor A (VEGFA), has an important role in both endothelial cell differentiation and vascular pattern development. Expression of VEGFA has been localised to ovigerous cords and follicles in developing ovaries and an increased expression of pro-angiogenic Vegfa isoform mRNA in relation to anti-angiogenic isoform mRNA occurs at the same time-point as the peak of primordial follicle assembly in perinatal rats. Elucidation of specific genes that affect vascular development within the ovary may be critical for determining not only the normal mechanisms of ovarian morphogenesis, but also for understanding certain ovarian reproductive disorders.
Collapse
Affiliation(s)
- Renee M McFee
- Department of Animal Science, University of Nebraska-Lincoln, 3940 Fair Street, Lincoln, NB 68583-0908, USA
| | | |
Collapse
|
38
|
Abstract
Accumulating evidence suggest that the concept of programming can also be applied to reproductive development and function, representing an ever expanding research area. Recently issues such as peri- or even preconceptional nutrition, transgenerational effects and underlying mechanisms have received considerable attention. The present chapter presents the existed evidence and reviews the available data from numerous animal and human studies on the effects of early life nutritional environment on adult reproductive function. Specific outcomes depend on the severity, duration and stage of development when nutritional perturbations are imposed, while sex-specific effects are also manifested. Apart from undernutrition, effects of relative overnutrition as well as the complex interactions between pre- and postnatal nutrition is of high importance, especially in the context of our days obesity epidemic. Mechanisms underlying reproductive programming are yet unclear, but may include a role for epigenetic modifications. Epigenetic modulation of critical genes involved in the control of reproductive function and potential intergenerational effects represent an exciting area of interdisciplinary research toward the development of new nutritional approaches during pre- and postnatal periods to ensure reproductive health in later life.
Collapse
|
39
|
Nilsson E, Zhang B, Skinner MK. Gene bionetworks that regulate ovarian primordial follicle assembly. BMC Genomics 2013; 14:496. [PMID: 23875758 PMCID: PMC3726361 DOI: 10.1186/1471-2164-14-496] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 07/04/2013] [Indexed: 11/16/2022] Open
Abstract
Background Primordial follicle assembly is the process by which ovarian primordial follicles are formed. During follicle assembly oocyte nests break down and a layer of pre-granulosa cells surrounds individual oocytes to form primordial follicles. The pool of primordial follicles formed is the source of oocytes for ovulation during a female’s reproductive life. Results The current study utilized a systems approach to detect all genes that are differentially expressed in response to seven different growth factor and hormone treatments known to influence (increase or decrease) primordial follicle assembly in a neonatal rat ovary culture system. One novel factor, basic fibroblast growth factor (FGF2), was experimentally determined to inhibit follicle assembly. The different growth factor and hormone treatments were all found to affect similar physiological pathways, but each treatment affected a unique set of differentially expressed genes (signature gene set). A gene bionetwork analysis identified gene modules of coordinately expressed interconnected genes and it was found that different gene modules appear to accomplish distinct tasks during primordial follicle assembly. Predictions of physiological pathways important to follicle assembly were validated using ovary culture experiments in which ERK1/2 (MAPK1) activity was increased. Conclusions A number of the highly interconnected genes in these gene networks have previously been linked to primary ovarian insufficiency (POI) and polycystic ovarian disease syndrome (PCOS). Observations have identified novel factors and gene networks that regulate primordial follicle assembly. This systems biology approach has helped elucidate the molecular control of primordial follicle assembly and provided potential therapeutic targets for the treatment of ovarian disease.
Collapse
Affiliation(s)
- Eric Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA 99164-4236, USA
| | | | | |
Collapse
|
40
|
Effects of culture and transplantation on follicle activation and early follicular growth in neonatal mouse ovaries. Cell Tissue Res 2013; 354:609-21. [PMID: 23824101 PMCID: PMC3836445 DOI: 10.1007/s00441-013-1678-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 06/03/2013] [Indexed: 11/30/2022]
Abstract
Mouse models have been widely utilized to elucidate the basic principles and regulatory mechanisms of primordial follicle activation. Outside their natural environment, the growth of follicles might be affected by unknown factors in vitro and the elimination of regulation in vivo. Currently, in vitro culture and transplantation of ovaries under the kidney capsule are two commonly used incubation methods. However, the limited number of studies that have been published compare various incubation systems and reveal differences between ovaries that are incubated and grown in vivo. We compare the number of primordial, primary and secondary follicles in cultured, transplanted and in-vivo-grown ovaries. We investigate the expression levels of four genes, including zona pellucida 3 (ZP3), growth and differentiation factor-9 (GDF-9), proliferating cell nuclear antigen (PCNA) and anti-Müllerian hormone (AMH). Our results suggest that in vitro culture accelerates follicle activation, delays the transition from primary to secondary follicles and affects the expression patterns of ZP3, GDF-9, PCNA and AMH. A larger number of secondary follicles in ovaries cultured in alpha-minimal essential medium (α-MEM) had intact zona pellucida compared with those grown in Dulbecco’s modified Eagle medium containing Ham’s F-12 nutrient mixture (D/F12), suggesting that α-MEM is a better basal medium. The transplanted ovaries demonstrated the most similar characteristics to the in-vivo-grown ovaries, indicating that transplantation provided an optimal environment for ovarian incubation. This study has thus established the similarities and differences between in-vivo-grown and incubated ovaries, demonstrated that transplantation can mostly mimic the environment of ovarian growth in vivo and determined the optimal basal culture medium between α-MEM and D/F12.
Collapse
|
41
|
Sominsky L, Sobinoff AP, Jobling MS, Pye V, McLaughlin EA, Hodgson DM. Immune regulation of ovarian development: programming by neonatal immune challenge. Front Neurosci 2013; 7:100. [PMID: 23781169 PMCID: PMC3679471 DOI: 10.3389/fnins.2013.00100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/22/2013] [Indexed: 12/20/2022] Open
Abstract
Neonatal immune challenge by administration of lipopolysaccharide (LPS) produces enduring alterations in the development and activity of neuroendocrine, immune and other physiological systems. We have recently reported that neonatal exposure to an immune challenge by administration of LPS results in altered reproductive development in the female Wistar rat. Specifically, LPS-treated animals exhibited diminished ovarian reserve and altered reproductive lifespan. In the current study, we examined the cellular mechanisms that lead to the previously documented impaired ovulation and reduced follicular pool. Rats were administered intraperitoneally either 0.05 mg/kg of LPS (Salmonella Enteritidis) or an equivalent volume of non-pyrogenic saline on postnatal days (PNDs) 3 and 5, and ovaries were obtained on PND 7. Microarray analysis revealed a significant upregulation in transcript expression (2-fold change; p < 0.05) for a substantial number of genes in the ovaries of LPS-treated animals, implicated in immune cell signaling, inflammatory responses, reproductive system development and disease. Several canonical pathways involved in immune recognition were affected by LPS treatment, such as nuclear factor-κB (NF-κB) activation and LPS-stimulated mitogen-activated protein kinase (MAPK) signaling. Quantitative Real-time PCR analysis supported the microarray results. Protein expression analysis of several components of the MAPK signaling pathway revealed a significant upregulation in the expression of Toll-like receptor 4 (TLR4) in the neonatal ovary of LPS-treated animals. These results indicate that neonatal immune challenge by administration of LPS has a direct effect on the ovary during the sensitive period of follicular formation. Given the pivotal role of inflammatory processes in the regulation of reproductive health, our findings suggest that early life immune activation via TLR signaling may have significant implications for the programming of ovarian development and fertility.
Collapse
Affiliation(s)
- Luba Sominsky
- Laboratory of Neuroimmunology, Faculty of Science and IT, School of Psychology, The University of Newcastle Callaghan, NSW, Australia
| | | | | | | | | | | |
Collapse
|
42
|
A new model of development of the mammalian ovary and follicles. PLoS One 2013; 8:e55578. [PMID: 23409002 PMCID: PMC3567121 DOI: 10.1371/journal.pone.0055578] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 01/03/2013] [Indexed: 01/15/2023] Open
Abstract
Ovarian follicular granulosa cells surround and nurture oocytes, and produce sex steroid hormones. It is believed that during development the ovarian surface epithelial cells penetrate into the ovary and develop into granulosa cells when associating with oogonia to form follicles. Using bovine fetal ovaries (n = 80) we identified a novel cell type, termed GREL for Gonadal Ridge Epithelial-Like. Using 26 markers for GREL and other cells and extracellular matrix we conducted immunohistochemistry and electron microscopy and chronologically tracked all somatic cell types during development. Before 70 days of gestation the gonadal ridge/ovarian primordium is formed by proliferation of GREL cells at the surface epithelium of the mesonephros. Primordial germ cells (PGCs) migrate into the ovarian primordium. After 70 days, stroma from the underlying mesonephros begins to penetrate the primordium, partitioning the developing ovary into irregularly-shaped ovigerous cords composed of GREL cells and PGCs/oogonia. Importantly we identified that the cords are always separated from the stroma by a basal lamina. Around 130 days of gestation the stroma expands laterally below the outermost layers of GREL cells forming a sub-epithelial basal lamina and establishing an epithelial-stromal interface. It is at this stage that a mature surface epithelium develops from the GREL cells on the surface of the ovary primordium. Expansion of the stroma continues to partition the ovigerous cords into smaller groups of cells eventually forming follicles containing an oogonium/oocyte surrounded by GREL cells, which become granulosa cells, all enclosed by a basal lamina. Thus in contrast to the prevailing theory, the ovarian surface epithelial cells do not penetrate into the ovary to form the granulosa cells of follicles, instead ovarian surface epithelial cells and granulosa cells have a common precursor, the GREL cell.
Collapse
|
43
|
Sominsky L, Meehan CL, Walker AK, Bobrovskaya L, McLaughlin EA, Hodgson DM. Neonatal immune challenge alters reproductive development in the female rat. Horm Behav 2012; 62:345-55. [PMID: 22366707 DOI: 10.1016/j.yhbeh.2012.02.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 02/04/2012] [Accepted: 02/06/2012] [Indexed: 10/14/2022]
Abstract
Neonatal lipopolysaccharide (LPS) exposure alters neuroendocrine, immune and behavioural responses in adult rats. Recent findings indicate that neonatal LPS treatment may have a more pronounced effect on the mating behaviours of females compared to males. The current study further explored the impact of neonatal inflammation on reproductive development in the female rat. Wistar rats were administered LPS (0.05 mg/kg, i.p.) or saline (equivolume) on postnatal days (PNDs) 3 and 5. The immediate effect of treatment was assessed on plasma corticosterone and tyrosine hydroxylase (TH) phosphorylation in the adrenal medulla. Weight gain and vaginal opening were recorded, and oestrous cyclicity was monitored post-puberty and in late adulthood. Blood and ovaries were collected throughout development to assess HPA and HPG hormones and to examine ovarian morphology. Reproductive success in the first (F1) generation and reproductive development in the second (F2) generation were also assessed. Neonatal LPS exposure resulted in increased TH phosphorylation in the neonatal adrenals. LPS treatment increased the corticosterone concentrations of females as juveniles, adolescents and adults, and reduced FSH in adolescence. Increased catch-up growth was evident in LPS-treated females, prompting earlier onset of puberty. Diminished follicular reserve was observed in neonatally LPS-treated females along with the advanced reproductive senescence. While fertility rates were not compromised, higher mortality and morbidity were observed in litters born to LPS-treated mothers. Female offspring of LPS-treated mothers displayed increased corticosterone on PND 14, increased catch-up growth and delayed emergence of the first oestrous cycle. No differences in any of the parameters assessed were observed in F2 males. These data suggest that neonatal immunological challenge has a profound impact on the female reproductive development, via the alteration of metabolic and neuroendocrine factors which regulate sexual maturation. Evidence of altered development in the female, but not male offspring of LPS-treated dams suggests increased susceptibility of females to the deleterious effects of neonatal immunological stress and its possible transferability to a subsequent generation.
Collapse
Affiliation(s)
- Luba Sominsky
- Laboratory of Neuroimmunology, School of Psychology, Faculty of Science and IT, The University of Newcastle, Australia.
| | | | | | | | | | | |
Collapse
|
44
|
Kiroshka VV, Tishchenko YO. Dynamics of folliculogenesis of sexually mature and neonatal ovarian tissue under conditions of long-term heterotopical transplantation. J EVOL BIOCHEM PHYS+ 2012. [DOI: 10.1134/s0022093012020089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Ungewitter EK, Yao HHC. How to make a gonad: cellular mechanisms governing formation of the testes and ovaries. Sex Dev 2012; 7:7-20. [PMID: 22614391 PMCID: PMC3474884 DOI: 10.1159/000338612] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Sex determination of the gonad is an extraordinary process by which a single organ anlage is directed to form one of two different structures, a testis or an ovary. Morphogenesis of these two organs utilizes many common cellular events; differences in the timing and execution of these events must combine to generate sexually dimorphic structures. In this chapter, we review recent research on the cellular processes of gonad morphogenesis, focusing on data from mouse models. We highlight the shared cellular mechanisms in testis and ovary morphogenesis and examine the differences that enable formation of the two organs responsible for the perpetuation of all sexually reproducing species.
Collapse
Affiliation(s)
- E K Ungewitter
- Reproductive Developmental Biology Group, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
46
|
Galas J, Słomczyńska M, Knapczyk-Stwora K, Durlej M, Starowicz A, Tabarowski Z, Rutka K, Szołtys M. Steroid levels and the spatiotemporal expression of steroidogenic enzymes and androgen receptor in developing ovaries of immature rats. Acta Histochem 2012; 114:207-16. [PMID: 21620445 DOI: 10.1016/j.acthis.2011.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 04/27/2011] [Accepted: 04/28/2011] [Indexed: 10/18/2022]
Abstract
Immunoexpression of 3β-hydroxysteroid dehydrogenase (3β-HSD), cytochrome P450c17 (P450c17), androgen receptor (AR), and steroid contents were studied in the ovaries of immature female Wistar rats killed between postnatal days 1 and 30. During days 1-7, ovarian somatic structures lacked AR, 3β-HSD and P450c17, except for the surface epithelium, which featured the presence of these three proteins, suggestive of its androgen responsiveness and steroidogenic function. On day 10, AR appeared in many somatic structures, including the granulosa layers, which coincided with the P450c17 immunoexpression in some theca/interstitial cells, and an increase in ovarian androgen concentration. On the following days a further rise in ovarian androgen and progesterone contents paralleled an increase in 3β-HSD and P450c17 immunoexpression in the theca layer cells and primary interstitial cells. However, the development of the follicles constituting the first follicular wave was aberrant, since they lacked AR expression until the preantral stage and were characterized by a delayed onset and much lower expression of the thecal P450c17. They could not ovulate, since ovarian content of estradiol was too low to evoke the LH surge. The clusters of the secondary interstitial cells found on day 30 exhibited predominant expression of 3β-HSD over P450c17, suggesting more intensive progesterone than androgen synthesis in these structures.
Collapse
|
47
|
The balance of proangiogenic and antiangiogenic VEGFA isoforms regulate follicle development. Cell Tissue Res 2012; 349:635-47. [PMID: 22322423 PMCID: PMC3429770 DOI: 10.1007/s00441-012-1330-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/29/2011] [Indexed: 01/13/2023]
Abstract
Vascular endothelial growth factor A (VEGFA) has been extensively studied because of its role in follicular development and is a principal angiogenic factor essential for angiogenesis. Since vascularization of the theca layer increases as follicles progress in size through preantral and antral stages, VEGFA might influence follicle growth via the regulation of angiogenesis. However, VEGFA might also influence follicular development through nonangiogenic mechanisms, since its expression has been localized in nonvascular follicles and cells. Alternative mRNA splicing of eight exons from the VEGFA gene results in the formation of various VEGFA isoforms. Each isoform has unique properties and is identified by the number of amino acids within the mature protein. Proangiogenic isoforms (VEGFA_XXX) are encoded by exon 8a, whereas a sister set of isoforms (VEGFA_XXXB) with antiangiogenic properties is encoded by exon 8b. The antiangiogenic VEGFA_XXXB isoforms comprise the majority of VEGFA expressed in most tissues, whereas expression of the proangiogenic VEGFA isoforms is upregulated in tissues undergoing active angiogenesis. Although proangiogenic and antiangiogenic isoforms can now be distinguished from one another, many studies evaluating VEGFA in ovarian and follicular development up to now have not differentiated proangiogenic VEGFA from antiangiogenic VEGFA. Experiments from our laboratory indicate that proangiogenic VEGFA promotes follicle recruitment and early follicular development and antiangiogenic VEGFA inhibits these processes. The balance of proangiogenic versus antiangiognic VEGFA isoforms is thus of importance during follicle development. Further studies are warranted to elucidate the way that this balance regulates follicular formation and progression.
Collapse
|
48
|
Cossigny DA, Findlay JK, Drummond AE. The effects of FSH and activin A on follicle development in vitro. Reproduction 2012; 143:221-9. [DOI: 10.1530/rep-11-0105] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Numerous studies have reported on the roles of activins in gonadal regulation; however, little is known about their specific roles in early folliculogenesis. Ovarian follicular growth was investigated in 10-day cultures of day 4 postnatal whole ovaries treated with activin A (ActA; 50 ng/ml), with or without FSH (100 ng/ml) in vitro. We hypothesized that treatment with ActA±FSH would affect rates of growth and atresia in follicles. None of the treatments affected primordial follicle activation, and antral follicles were not observed after 10 days in culture. Primordial follicle numbers from all treatment groups were ∼20% of those in day 4 fresh ovaries, indicating that activation had occurred. In the presence of ActA, preantral follicle numbers increased significantly (P<0.0001). ActA alone decreased the proportion of atretic follicles in the primary and preantral classes, whereas the combined treatment of ActA+FSH increased the proportion of atretic preantral oocytes. Real-time PCR analysis revealed that follistatin, FSH receptor, and activin βA and βB subunits were all expressed at significantly higher levels in the ActA-only treated group but not in the ActA+FSH group. Here, we report novel findings supporting the role of FSH in primordial follicle survival through an action on apoptosis and a stimulatory role of ActA in the primordial to primary and preantral stages of follicle development, suggesting an inhibitory action of activin on oocyte apoptosis.
Collapse
|
49
|
Guigon CJ, Cohen-Tannoudji M. [Reconsidering the roles of female germ cells in ovarian development and folliculogenesis]. Biol Aujourdhui 2012; 205:223-33. [PMID: 22251857 DOI: 10.1051/jbio/2011022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Indexed: 11/15/2022]
Abstract
The production of fertilizable ova is the consequence of multiple events that start as soon as ovarian development and culminate at the time of ovulation. Throughout their development, germ cells are associated with companion somatic cells, which ensure germ cell survival, growth and maturation. Data obtained in vitro and in vivo on several animal models of germ cell depletion have led to uncover the many roles of germ cells on both ovarian development and folliculogenesis. During ovarian development, germ cells become progressively enclosed within epithelial structures called "ovigerous cords" constituted by pregranulosa cells, lined by a basement membrane. At the end of ovarian development, ovigerous cords fragment into primordial follicles, which are epithelial units constituted by an oocyte surrounded by a single layer of granulosa cells. Germ cells are necessary for the fragmentation of ovigerous cords into follicles, since in their absence, no follicle will form. Germ cells also ensure the differentiation of the ovarian somatic lineage, and they may inhibit the testis-differentiating pathway by preventing the conversion of pregranulosa cells into Sertoli cells, their counterpart in the testis. Regularly, primordial follicles are recruited into the growing follicle pool and initiate their growth. They develop through primary, preantral, antral and preovulatory stages before being ovulated. Interestingly, the action of the oocyte on companion somatic cells tightly depends on the follicular stage. In primordial follicles, the oocyte prevents the transdifferentiation of granulosa cells into cells resembling Sertoli cells. By contrast, as soon as the follicle enters growth, the oocyte regulates the functional differentiation of granulosa cells and at the latest stages, it prevents their premature maturation into luteal cells. Overall, these data demonstrate that the female germ cell act on companion somatic cells to regulate ovarian development and folliculogenesis, thereby actively supporting its own maturation.
Collapse
Affiliation(s)
- Céline J Guigon
- Université Paris Diderot, Sorbonne Paris Cité, Équipe Physiologie de l'Axe Gonadotrope, Unité Biologie Fonctionnelle et Adaptative, Paris, France.
| | | |
Collapse
|
50
|
Abstract
The differentiation of primordial germ cells (PGCs) into functional oocytes is important for the continuation of species. In mammals, PGCs begin to differentiate into oocytes during embryonic development. Oocytes develop in clusters called germ line cysts. During fetal or neonatal development, germ cell cysts break apart into single oocytes that become surrounded by pregranulosa cells to form primordial follicles. During the process of cyst breakdown, a subset of cells in each cyst undergoes cell death with only one-third of the initial number of oocytes surviving to form primordial follicles. The mechanisms that control cyst breakdown, oocyte survival, and follicle assembly are currently under investigation. This review describes the mechanisms that have been implicated in the control of primordial follicle formation, which include programmed cell death regulation, growth factor and other signaling pathways, regulation by transcription factors and hormones, meiotic progression, and changes in cell adhesion. Elucidation of mechanisms leading to formation of the primordial follicle pool will help research efforts in ovarian biology and improve treatments of female infertility, premature ovarian failure, and reproductive cancers.
Collapse
Affiliation(s)
- Melissa E Pepling
- Department of Biology, Syracuse University, Syracuse, New York 13244, USA.
| |
Collapse
|