1
|
Cai J, Liang X, Sun Y, Bao S. Beneficial effects of human umbilical cord mesenchymal stem cell (HUCMSC) transplantation on cyclophosphamide (CTX)-induced premature ovarian failure (POF) in Tibetan miniature pigs. Transpl Immunol 2024; 84:102051. [PMID: 38744348 DOI: 10.1016/j.trim.2024.102051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 05/11/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Premature ovarian failure (POF), also known as primary ovarian insufficiency, is a common endocrine disease in young women. The emergence of regenerative medicine using stem cells may improve ovarian function and structure, and represents a promising prospect for POF treatment. In his study, we explored the therapeutic effects of human umbilical cord mesenchymal stem cell (HUCMSC) transplantation in a Tibetan miniature pig model of cyclophosphamide (CTX)-induced POF. METHODS We cultured and identified HUCMSCs, labeled them with DiR iodide red dye, and implanted them into a CTX-induced model of POF in Tibetan miniature pigs. The daily weight changes were recorded, and the levels of estradiol (E2) and follicle-stimulating hormone (FSH) were measured on days 0, 7, and 14. At the end of the 21-day observation period, in vivo imaging of the bilateral ovaries was performed, and the ovarian index was measured. Ovarian tissue morphology and follicles were examined by hematoxylin-eosin staining. The terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay was employed to assess cell apoptosis, and immunohistochemistry was used to determine the levels of p-AKT, p-ERK1/2, BAX, and BCL2 expression. RESULTS Our analysis indicated successful delivery of HUCMSCs to the ovaries of the POF pig model. Significant increases were observed in body weight, E2 levels, ovarian index, and number of normal follicles (all p < 0.05). Moreover, FSH levels reduced and ovarian tissue morphology improved following HUCMSCs transplantation (all p < 0.05). Importantly, upregulated p-AKT, p-ERK1/2, and BCL2 expression were observed, whereas the expression of BAX was suppressed (all p < 0.05), suggesting the inhibition of ovarian cell apoptosis. CONCLUSION Our study highlights the significant therapeutic effects of HUCMSC transplantation on CTX-induced POF in a Tibetan miniature pig model.
Collapse
Affiliation(s)
- Junhong Cai
- Medical Laboratory Central, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Xiaochen Liang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan 571199, PR China; Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, Hainan 571199, PR China
| | - Yuting Sun
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan 571199, PR China; Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, Hainan 571199, PR China
| | - Shan Bao
- Department of Gynaecology and Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China.
| |
Collapse
|
2
|
Cai Y, Liu Z, Gao T, Hu G, Yin W, Wāng Y, Zhao L, Xu D, Wang H, Wei T. Newly discovered developmental and ovarian toxicity of 3-monochloro-1,2-propanediol in Drosophila melanogaster and cyanidin-3-O-glucoside's protective effect. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 874:162474. [PMID: 36863584 DOI: 10.1016/j.scitotenv.2023.162474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 06/18/2023]
Abstract
3-Monochloro-1,2-propanediol (3-MCPD) is a pervasive environmental pollutant that is unintentionally produced during industrial production and food processing. Although some studies reported the carcinogenicity and male reproduction toxicity of 3-MCPD thus far, it remains unexplored whether 3-MCPD hazards to female fertility and long-term development. In this study, the model Drosophila melanogaster was employed to evaluate risk assessment of emerging environmental contaminants 3-MCPD at various levels. We found that flies on dietary exposure to 3-MCPD incurred lethality in a concentration- and time-dependent way and interfered with metamorphosis and ovarian development, resulting in developmental retardance, ovarian deformity and female fecundity disorders. Mechanistically, 3-MCPD caused redox imbalance observed as a drastically increased oxidative status in ovaries, confirmed by increased reactive oxygen species (ROS) and decreased antioxidant activities, which is probably responsible for female reproductive impairments and developmental retardance. Intriguingly, these defects can be substantially prevented by a natural antioxidant, cyanidin-3-O-glucoside (C3G), further confirming a critical role of ovarian oxidative damage in the developmental and reproductive toxicity of 3-MCPD. The present study expanded the findings that 3-MCPD acts as a developmental and female reproductive toxicant, and our work provides a theoretical basis for the exploitation of a natural antioxidant resource as a dietary antidote for the reproductive and developmental hazards of environmental toxicants that act via increasing ROS in the target organ.
Collapse
Affiliation(s)
- Yang Cai
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Zongzhong Liu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Tiantian Gao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Guoyi Hu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Wenjun Yin
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China
| | - Yán Wāng
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China.
| | - Lingli Zhao
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Dexiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Tian Wei
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China.
| |
Collapse
|
3
|
Wang LP, Liu JX, Shang JL, Kong XZ, Guan BX, Wang J. KGLRR: A low-rank representation K-means with graph regularization constraint method for Single-cell type identification. Comput Biol Chem 2023; 104:107862. [PMID: 37031647 DOI: 10.1016/j.compbiolchem.2023.107862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/26/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
Single-cell RNA sequencing technology provides a tremendous opportunity for studying disease mechanisms at the single-cell level. Cell type identification is a key step in the research of disease mechanisms. Many clustering algorithms have been proposed to identify cell types. Most clustering algorithms perform similarity calculation before cell clustering. Because clustering and similarity calculation are independent, a low-rank matrix obtained only by similarity calculation may be unable to fully reveal the patterns in single-cell data. In this study, to capture accurate single-cell clustering information, we propose a novel method based on a low-rank representation model, called KGLRR, that combines the low-rank representation approach with K-means clustering. The cluster centroid is updated as the cell dimension decreases to better from new clusters and improve the quality of clustering information. In addition, the low-rank representation model ignores local geometric information, so the graph regularization constraint is introduced. KGLRR is tested on both simulated and real single-cell datasets to validate the effectiveness of the new method. The experimental results show that KGLRR is more robust and accurate in cell type identification than other advanced algorithms.
Collapse
Affiliation(s)
- Lin-Ping Wang
- School of Computer Science, Qufu Normal University, Rizhao 276826, China
| | - Jin-Xing Liu
- School of Computer Science, Qufu Normal University, Rizhao 276826, China
| | - Jun-Liang Shang
- School of Computer Science, Qufu Normal University, Rizhao 276826, China
| | - Xiang-Zhen Kong
- School of Computer Science, Qufu Normal University, Rizhao 276826, China
| | - Bo-Xin Guan
- School of Computer Science, Qufu Normal University, Rizhao 276826, China
| | - Juan Wang
- School of Computer Science, Qufu Normal University, Rizhao 276826, China.
| |
Collapse
|
4
|
Athar F, Templeman NM. C. elegans as a model organism to study female reproductive health. Comp Biochem Physiol A Mol Integr Physiol 2022; 266:111152. [PMID: 35032657 DOI: 10.1016/j.cbpa.2022.111152] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 12/17/2022]
Abstract
Female reproductive health has been historically understudied and underfunded. Here, we present the advantages of using a free-living nematode, Caenorhabditis elegans, as an animal system to study fundamental aspects of female reproductive health. C. elegans is a powerful high-throughput model organism that shares key genetic and physiological similarities with humans. In this review, we highlight areas of pressing medical and biological importance in the 21st century within the context of female reproductive health. These include the decline in female reproductive capacity with increasing chronological age, reproductive dysfunction arising from toxic environmental insults, and cancers of the reproductive system. C. elegans has been instrumental in uncovering mechanistic insights underlying these processes, and has been valuable for developing and testing therapeutics to combat them. Adopting a convenient model organism such as C. elegans for studying reproductive health will encourage further research into this field, and broaden opportunities for making advancements into evolutionarily conserved mechanisms that control reproductive function.
Collapse
Affiliation(s)
- Faria Athar
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Nicole M Templeman
- Department of Biology, University of Victoria, Victoria, British Columbia V8P 5C2, Canada.
| |
Collapse
|
5
|
Wang CY, Gao YL, Liu JX, Kong XZ, Zheng CH. Single-Cell RNA Sequencing Data Clustering by Low-Rank Subspace Ensemble Framework. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:1154-1164. [PMID: 33026977 DOI: 10.1109/tcbb.2020.3029187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The rapid development of single-cell RNA sequencing (scRNA-seq)technology reveals the gene expression status and gene structure of individual cells, reflecting the heterogeneity and diversity of cells. The traditional methods of scRNA-seq data analysis treat data as the same subspace, and hide structural information in other subspaces. In this paper, we propose a low-rank subspace ensemble clustering framework (LRSEC)to analyze scRNA-seq data. Assuming that the scRNA-seq data exist in multiple subspaces, the low-rank model is used to find the lowest rank representation of the data in the subspace. It is worth noting that the penalty factor of the low-rank kernel function is uncertain, and different penalty factors correspond to different low-rank structures. Moreover, the single cluster model is difficult to find the cellular structure of all datasets. To strengthen the correlation between model solutions, we construct a new ensemble clustering framework LRSEC by using the low-rank model as the basic learner. The LRSEC framework captures the global structure of data through low-rank subspaces, which has better clustering performance than a single clustering model. We validate the performance of the LRSEC framework on seven small datasets and one large dataset and obtain satisfactory results.
Collapse
|
6
|
Upson K, Weinberg CR, Nichols HB, Dinse GE, D'Aloisio AA, Sandler DP, Baird DD. Early-life Farm Exposure and Ovarian Reserve in a US Cohort of Women. Epidemiology 2021; 32:672-680. [PMID: 34039897 PMCID: PMC8370468 DOI: 10.1097/ede.0000000000001376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In a previous exploratory study, we reported lower concentrations of the ovarian reserve biomarker anti-Müllerian hormone (AMH) in adulthood with prenatal farm exposure. We now examine this association as well as childhood farm exposure using enrollment data from the Sister Study, a large US cohort of women. METHODS We collected prenatal and childhood farm exposure data by questionnaire and telephone interview. However, serum AMH data were available only for a nested subset: premenopausal women ages 35-54 subsequently diagnosed with breast cancer (n = 418 cases) and their matched controls (n = 866). To avoid potential bias from restricting analyses to only premenopausal controls, we leveraged the available cohort data. We used data from both premenopausal cases and controls as well as postmenopausal women ages 35-54 (n = 3,526) (all presumed to have undetectable AMH concentrations) and applied weights to produce a sample representative of the cohort ages 35-54 (n = 17,799). The high proportion of undetectable AMH concentrations (41%) was addressed using reverse-scale Cox regression. An adjusted hazard ratio (HR) <1.0 indicates that exposed individuals had lower AMH concentrations than unexposed individuals. RESULTS Prenatal exposure to maternal residence or work on a farm was associated with lower AMH concentrations (HR 0.66; 95% confidence intervals [CI] = 0.48 to 0.90). Associations between childhood farm residence exposures and AMH were null or weak, except childhood contact with pesticide-treated livestock or buildings (HR 0.69; 95% CI = 0.40 to 1.2). CONCLUSIONS Replication of the prenatal farm exposure and lower adult AMH association raises concern that aspects of prenatal farm exposure may result in reduced adult ovarian reserve.
Collapse
Affiliation(s)
- Kristen Upson
- From the Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Clarice R Weinberg
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC
| | - Hazel B Nichols
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Gregg E Dinse
- Clinical and Public Health Sciences, Social & Scientific Systems, Durham, NC
| | - Aimee A D'Aloisio
- Clinical and Public Health Sciences, Social & Scientific Systems, Durham, NC
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC
| | - Donna D Baird
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC
| |
Collapse
|
7
|
Fraser BA, Miller K, Trigg NA, Smith ND, Western PS, Nixon B, Aitken RJ. A novel approach to nonsurgical sterilization; application of menadione-modified gonocyte-targeting M13 bacteriophage for germ cell ablation in utero. Pharmacol Res Perspect 2021; 8:e00654. [PMID: 32930516 PMCID: PMC7507010 DOI: 10.1002/prp2.654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
There remains a compelling need for the development of nonsurgical sterilizing agents to expand the fertility management options for both domestic and feral animal species. We hypothesize that an efficacious sterilization approach would be to selectively ablate nonrenewable cell types that are essential for reproduction, such as the undifferentiated gonocytes within the embryonic gonad. Here, we report a novel strategy to achieve this goal centered on the use of a chemically modified M13 bacteriophage to effect the targeted delivery of menadione, a redox‐cycling naphthoquinone, to mouse gonocytes. Panning of the M13 random peptide ‘phage display library proved effective in the isolation of gonocyte‐specific targeting clones. One such clone was modified via N‐succinimidyl‐S‐acetylthioacetate (SATA) linkage to the N‐terminus of the major PVIII capsid protein. Subsequent deacetylation of the SATA was undertaken to expose a thiol group capable of reacting with menadione through Michael addition. This chemical modification was confirmed using UV spectrophotometry. In proof‐of‐concept experiments we applied the modified ‘phage to primary cultures of fetal germ cells and induced, an approximately, 60% reduction in the viability of the target cell population. These studies pave the way for in vivo application of chemically modified M13 bacteriophage in order to achieve the selective ablation of nonrenewable cell types in the reproductive system, thereby providing a novel nonsurgical approach the regulation of fertility in target species.
Collapse
Affiliation(s)
- Barbara A Fraser
- Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Kasey Miller
- Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Natalie A Trigg
- Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Nathan D Smith
- Analytical and Biomolecular Research Facility, The University of Newcastle, Callaghan, NSW, Australia
| | - Patrick S Western
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Robert J Aitken
- Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
8
|
Molecular Targets and Associated Signaling Pathways of Jingshu Granules in Ovarian Cysts Based on Systemic Pharmacological Analysis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6660087. [PMID: 33623786 PMCID: PMC7875638 DOI: 10.1155/2021/6660087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/09/2021] [Indexed: 11/18/2022]
Abstract
Background More than a third of women could develop ovarian cysts during their lifetime. Jingshu granules are used for the treatment of gynecological disease of primary dysmenorrhea. However, the molecular mechanisms of Jingshu granules in ovarian cysts are still unreported. We aimed to find the active ingredients, molecular targets, and potential signaling pathways of Jingshu granules in ovarian cysts by using the systemic pharmacological analysis. Methods Firstly, the effect of Jingshu granules on female hormones and reproductive organs of young female rats was evaluated. Secondly, candidate pharmaceutical ingredients of Jingshu granules were retrieved from the traditional Chinese medicine systems pharmacology (TCMSP) database and analysis platform. Potential protein targets for the active ingredients in Jingshu granules were then identified according to the oral bioavailability and drug-likeness indices. Thirdly, ovarian cyst-related gene targets were screened based on different databases. Finally, enrichment analysis was used to analyze the potential biological function of intersection targets between Jingshu granules and ovarian cysts. Results In young female rats, Jingshu granules reduced the secretion of estradiol, progesterone, and prolactin and could affect the development of the uterus. This suggested that Jingshu granules played roles in hormone secretion and reproduction. From the TCMSP, a total of 1021 pharmaceutical ingredients of Jingshu granules were retrieved. After further screening, a total of 166 active ingredients and 159 protein targets of Jingshu granules were identified. In addition, 4488 gene targets of ovarian cysts were screened out. After taking the intersection, a total of 110 intersection targets were identified between potential protein targets of Jingshu granules and gene targets of ovarian cysts. In the functional analysis of 110 intersection targets, 8 signaling pathways including progesterone-mediated oocyte maturation (MAPK8 and CDK1 involved), GnRH signaling pathway (JUN involved), T cell receptor signaling pathway and Toll-like receptor signaling pathway (MAPK1 involved), NOD-like receptor signaling pathway (TNF, IL6, and IL1B involved), p53 signaling pathway (CDK2 and CDK4 involved), VEGF signaling pathway (MAPK14 involved), and PPAR signaling pathway (PPARG involved) were obtained. Conclusion Our study revealed that Jingshu granules could function in patients with ovarian cysts through a number of molecular targets and signaling pathways. Our study may provide a new field into the mechanisms of Jingshu granules in ovarian cysts, from the molecular to the signaling pathway level.
Collapse
|
9
|
The Influence of Environmental Factors on Ovarian Function, Follicular Genesis, and Oocyte Quality. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1300:41-62. [PMID: 33523429 DOI: 10.1007/978-981-33-4187-6_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Endocrine-disrupting chemicals (EDCs) exist ubiquitously in the environment. Epidemiological data suggest that the increasing prevalence of infertility may be related to the numerous chemicals. Exposure to EDCs may have significant adverse impacts on the reproductive system including fertility, ovarian reserve, and sex steroid hormone levels. This chapter covers the common exposure ways, the origins of EDCs, and their effects on ovarian function, follicular genesis, and oocyte quality. Furthermore, we will review the origin and the physiology of ovarian development, as well as explore the mechanisms in which EDCs act on the ovary from human and animal data. And then, we will focus on the bisphenol A (BPA), which has been shown to reduce fertility and ovarian reserve, as well as disrupt steroidogenesis in animal and human models. Finally, we will discuss the future direction of prevention and solution methods.
Collapse
|
10
|
Beato S, Toledo-Solís FJ, Fernández I. Vitamin K in Vertebrates' Reproduction: Further Puzzling Pieces of Evidence from Teleost Fish Species. Biomolecules 2020; 10:E1303. [PMID: 32917043 PMCID: PMC7564532 DOI: 10.3390/biom10091303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022] Open
Abstract
Vitamin K (VK) is a fat-soluble vitamin that vertebrates have to acquire from the diet, since they are not able to de novo synthesize it. VK has been historically known to be required for the control of blood coagulation, and more recently, bone development and homeostasis. Our understanding of the VK metabolism and the VK-related molecular pathways has been also increased, and the two main VK-related pathways-the pregnane X receptor (PXR) transactivation and the co-factor role on the γ-glutamyl carboxylation of the VK dependent proteins-have been thoroughly investigated during the last decades. Although several studies evidenced how VK may have a broader VK biological function than previously thought, including the reproduction, little is known about the specific molecular pathways. In vertebrates, sex differentiation and gametogenesis are tightly regulated processes through a highly complex molecular, cellular and tissue crosstalk. Here, VK metabolism and related pathways, as well as how gametogenesis might be impacted by VK nutritional status, will be reviewed. Critical knowledge gaps and future perspectives on how the different VK-related pathways come into play on vertebrate's reproduction will be identified and proposed. The present review will pave the research progress to warrant a successful reproductive status through VK nutritional interventions as well as towards the establishment of reliable biomarkers for determining proper nutritional VK status in vertebrates.
Collapse
Affiliation(s)
- Silvia Beato
- Campus de Vegazana, s/n, Universidad de León (ULE), 24071 León, Spain;
| | - Francisco Javier Toledo-Solís
- Consejo Nacional de Ciencia y Tecnología (CONACYT, México), Av. Insurgentes Sur 1582, Col. Crédito Constructor, Alcaldía Benito Juárez, C.P. 03940 Ciudad de Mexico, Mexico;
- Department of Biology and Geology, University of Almería, 04120 Almería, Spain
| | - Ignacio Fernández
- Center for Aquaculture Research, Agrarian Technological Institute of Castile and Leon, Ctra. Arévalo, s/n, 40196 Zamarramala, Segovia, Spain
| |
Collapse
|
11
|
Kole E, Ozkan SO, Eraldemir C, Akar FY, Ozbek SK, Kole MC, Kum T, Filiz PC. Effects of melatonin on ovarian reserve in cigarette smoking: an experimental study. Arch Med Sci 2020; 16:1376-1386. [PMID: 33224337 PMCID: PMC7667435 DOI: 10.5114/aoms.2019.89409] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/15/2019] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION The aim of the study was to investigate whether melatonin has a protective effect against diminished ovarian reserve induced by smoking. MATERIAL AND METHODS Seventy-two female Wistar-Albino rats were divided into 6 groups: group I (room air), group II (chronic cigarette smoking), group III (room air + 10 mg/kg subcutaneous melatonin), group IV (room air + 20 mg/kg subcutaneous melatonin), group V (chronic cigarette smoking + 10 mg/kg subcutaneous melatonin), group VI (chronic cigarette smoking + 20 mg/kg subcutaneous melatonin). For 45 days, rats were exposed to cigarette smoke through a smoking machine, then subcutaneous melatonin was administered. Apoptotic index, immunohistochemical scoring, ovarian follicle counting, ovarian tissue and serum malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT) analyses were carried out. RESULTS All of the primordial, primary, secondary and mature follicle numbers were found to be significantly lowered in study groups. Increased HSCORE with anti-caspase-3 staining and a high follicular apoptotic index were demonstrated in the smoking group. Serum and ovarian tissue levels of MDA were found to be elevated with smoke exposure whereas lower MDA levels were determined in melatonin treated groups. Serum and tissue levels of SOD, GPx and CAT were shown to be reduced in the smoking group in comparison with melatonin treated and control groups. 20 mg/kg melatonin administration in the smoking group revealed significantly decreased HSCOREs and apoptotic indices. CONCLUSIONS Cigarette smoking has been definitely shown to be associated with impaired ovarian reserve with respect to significantly diminished numbers of primordial, primary, secondary and mature follicles. Dose-related treatment of melatonin in smokers may provide an evidently reduced apoptotic index and improved antioxidant activity in tissue.
Collapse
Affiliation(s)
- Emre Kole
- Department of Gynecology and Obstetrics, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Sebiha Ozdemir Ozkan
- Department of Gynecology and Obstetrics, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Ceyla Eraldemir
- Department of Biochemistry, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Furuzan Yildiz Akar
- Department of Pharmacology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Sema Kurnaz Ozbek
- Department of Histology and Embryology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Merve Cakir Kole
- Department of Gynecology and Obstetrics, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Tugba Kum
- Department of Biochemistry, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | | |
Collapse
|
12
|
Budani MC, D'Aurora M, Stuppia L, Gatta V, Tiboni GM. Whole‐body exposure to cigarette smoke alters oocyte miRNAs expression in C57BL/6 mice. Mol Reprod Dev 2019; 86:1741-1757. [DOI: 10.1002/mrd.23267] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/09/2019] [Accepted: 08/31/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Maria Cristina Budani
- Department of Medicine and Aging SciencesUniversity “G. d'Annunzio” Chieti‐Pescara Chieti Italy
| | - Marco D'Aurora
- Department of Psychological, Health and Territorial Sciences (DISPUTer), Laboratory of Molecular Genetics, School of Medicine and Health SciencesUniversity “G. d'Annunzio” Chieti‐Pescara Chieti Italy
- Center of Excellence on Aging and Translational Medicine ‐ (CeSI‐MeT)University “G. d'Annunzio” Chieti‐Pescara Chieti Italy
| | - Liborio Stuppia
- Department of Psychological, Health and Territorial Sciences (DISPUTer), Laboratory of Molecular Genetics, School of Medicine and Health SciencesUniversity “G. d'Annunzio” Chieti‐Pescara Chieti Italy
- Center of Excellence on Aging and Translational Medicine ‐ (CeSI‐MeT)University “G. d'Annunzio” Chieti‐Pescara Chieti Italy
| | - Valentina Gatta
- Department of Psychological, Health and Territorial Sciences (DISPUTer), Laboratory of Molecular Genetics, School of Medicine and Health SciencesUniversity “G. d'Annunzio” Chieti‐Pescara Chieti Italy
- Center of Excellence on Aging and Translational Medicine ‐ (CeSI‐MeT)University “G. d'Annunzio” Chieti‐Pescara Chieti Italy
| | - Gian Mario Tiboni
- Department of Medical, Oral and Biotechnological SciencesUniversity “G. d'Annunzio” Chieti‐Pescara Chieti Italy
| |
Collapse
|
13
|
Guerreiro DD, de Lima LF, de Sá NAR, Tetaping GM, Alves BG, Lobo CH, Deusdênia Loiola O, Smitz J, de Figueiredo JR, Ribeiro Rodrigues AP. In vitro study of Withanolide D toxicity on goat preantral follicles and its effects on the cell cycle. Reprod Toxicol 2019; 84:18-25. [DOI: 10.1016/j.reprotox.2018.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/15/2018] [Accepted: 12/19/2018] [Indexed: 10/27/2022]
|
14
|
Upson K, Chin HB, Marsh EE, Baird DD. Intrauterine, Infant, and Childhood Factors and Ovarian Reserve in Young African American Women. J Womens Health (Larchmt) 2019; 28:1711-1720. [PMID: 30638418 DOI: 10.1089/jwh.2018.7382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background: Ovarian toxic exposures during early development may contribute to reduced ovarian reserve in adulthood. Materials and Methods: We explored a range of intrauterine, infant, and childhood factors in relation to a biomarker of ovarian reserve, anti-Müllerian hormone (AMH) concentrations, in adulthood. We conducted a cross-sectional exploratory study of 1600 African American women 23-35 years of age residing in the Detroit, Michigan metropolitan area, who had serum AMH measurements (Ansh Labs PicoAMH enzyme-linked immunosorbent assay) and no previous polycystic ovarian syndrome diagnosis. Information on 32 intrauterine, infant, and childhood factors was ascertained by self-administered questionnaires, with 87% of participants receiving assistance from mothers. The percent differences in AMH concentrations in relation to early-life factors and 95% confidence intervals (CIs) were estimated using multivariable linear regression, adjusting for age, current hormonal contraceptive use, and body mass index. Results: Of the early-life factors evaluated in this study, two maternal pregnancy factors were associated with lower AMH concentrations in adult participants. Participants whose mothers lived or worked on a farm (vs. neither lived nor worked on a farm) when pregnant with the participant had 42% lower AMH concentrations (95% CI = -62 to -9). Among participants whose mothers lived in Michigan when pregnant with the participant (n = 1238), maternal residence in Detroit for at least a month was associated with 22% lower AMH concentrations (95% CI = -34 to -8) in the participant. Conclusions: Further research is merited to replicate our findings and identify the aspects of maternal farm exposure and Detroit residence that may be associated with lower AMH concentrations.
Collapse
Affiliation(s)
- Kristen Upson
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Helen B Chin
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Erica E Marsh
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Donna D Baird
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
15
|
Fuller EA, Sominsky L, Sutherland JM, Redgrove KA, Harms L, McLaughlin EA, Hodgson DM. Neonatal immune activation depletes the ovarian follicle reserve and alters ovarian acute inflammatory mediators in neonatal rats. Biol Reprod 2018; 97:719-730. [PMID: 29040417 DOI: 10.1093/biolre/iox123] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 10/07/2017] [Indexed: 12/30/2022] Open
Abstract
Normal ovarian development is crucial for female reproductive success and longevity. Interruptions to the delicate process of initial folliculogenesis may lead to ovarian dysfunction. We have previously demonstrated that an early life immune challenge in the rat, induced by administration of lipopolysaccharide (LPS) on postnatal day (PND) 3 and 5, depletes ovarian follicle reserve long term. Here, we hypothesized that this neonatal immune challenge leads to an increase in peripheral and ovarian inflammatory signaling, contributing to an acute depletion of ovarian follicles. Morphological analysis of neonatal ovaries indicated that LPS administration significantly depleted PND 5 primordial follicle populations and accelerated follicle maturation. LPS exposure upregulated circulating interleukin 6, tumor necrosis factor alpha (TNFa), and C-reactive protein on PND 5, and upregulated ovarian mRNA expression of Tnfa, mitogen-activated protein kinase 8 (Mapk8/Jnk1), and growth differentiation factor 9 (Gdf9) (P < 0.05). Mass spectrometry and cell signaling pathway analysis indicated upregulation of cellular pathways associated with acute phase signaling, and cellular survival and assembly. Apoptosis assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling indicated significantly increased positive staining in the ovaries of LPS-treated neonates. These findings suggest that increased proinflammatory signaling within the neonatal ovary may be responsible for the LPS-induced depletion of the primordial follicle pool. These findings also have implications for female reproductive health, as the ovarian reserve is a major determinate of female reproductive longevity.
Collapse
Affiliation(s)
- Erin A Fuller
- Laboratory of Neuroimmunology, Priority Research Centre for Brain and Mental Health Research, School of Psychology, University of Newcastle, Callaghan, New South Wales, Australia
| | - Luba Sominsky
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Melbourne, Victoria, Australia
| | - Jessie M Sutherland
- School of Environmental and Life Sciences, Priority Research Centre in Chemical Biology, University of Newcastle, Callaghan, New South Wales, Australia
| | - Kate A Redgrove
- School of Environmental and Life Sciences, Priority Research Centre in Chemical Biology, University of Newcastle, Callaghan, New South Wales, Australia
| | - Lauren Harms
- Laboratory of Neuroimmunology, Priority Research Centre for Brain and Mental Health Research, School of Psychology, University of Newcastle, Callaghan, New South Wales, Australia
| | - Eileen A McLaughlin
- School of Environmental and Life Sciences, Priority Research Centre in Chemical Biology, University of Newcastle, Callaghan, New South Wales, Australia.,School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Deborah M Hodgson
- Laboratory of Neuroimmunology, Priority Research Centre for Brain and Mental Health Research, School of Psychology, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
16
|
Miao Y, Zhou C, Bai Q, Cui Z, ShiYang X, Lu Y, Zhang M, Dai X, Xiong B. The protective role of melatonin in porcine oocyte meiotic failure caused by the exposure to benzo(a)pyrene. Hum Reprod 2017; 33:116-127. [DOI: 10.1093/humrep/dex331] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/12/2017] [Indexed: 12/27/2022] Open
Affiliation(s)
- Yilong Miao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Changyin Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Qingyun Bai
- School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Zhaokang Cui
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiayan ShiYang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yajuan Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Mianqun Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoxin Dai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Bo Xiong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
17
|
Molecular Mechanisms Responsible for Increased Vulnerability of the Ageing Oocyte to Oxidative Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4015874. [PMID: 29312475 PMCID: PMC5664291 DOI: 10.1155/2017/4015874] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/03/2017] [Indexed: 12/23/2022]
Abstract
In their midthirties, women experience a decline in fertility, coupled to a pronounced increase in the risk of aneuploidy, miscarriage, and birth defects. Although the aetiology of such pathologies are complex, a causative relationship between the age-related decline in oocyte quality and oxidative stress (OS) is now well established. What remains less certain are the molecular mechanisms governing the increased vulnerability of the aged oocyte to oxidative damage. In this review, we explore the reduced capacity of the ageing oocyte to mitigate macromolecular damage arising from oxidative insults and highlight the dramatic consequences for oocyte quality and female fertility. Indeed, while oocytes are typically endowed with a comprehensive suite of molecular mechanisms to moderate oxidative damage and thus ensure the fidelity of the germline, there is increasing recognition that the efficacy of such protective mechanisms undergoes an age-related decline. For instance, impaired reactive oxygen species metabolism, decreased DNA repair, reduced sensitivity of the spindle assembly checkpoint, and decreased capacity for protein repair and degradation collectively render the aged oocyte acutely vulnerable to OS and limits their capacity to recover from exposure to such insults. We also highlight the inadequacies of our current armoury of assisted reproductive technologies to combat age-related female infertility, emphasising the need for further research into mechanisms underpinning the functional deterioration of the ageing oocyte.
Collapse
|
18
|
Trinh TN, McLaughlin EA, Gordon CP, Bernstein IR, Pye VJ, Redgrove KA, McCluskey A. Small molecule Hedgehog pathway antagonists. Org Biomol Chem 2017; 15:3046-3059. [DOI: 10.1039/c6ob01959e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Leveraging our quinolone-1-(2H)-one based Hedgehog signalling pathway (HSP) inhibitors we have developed two new classes of HSP inhibitors based on: l-tryptophan and benzo[1,3]dioxol-5-ylmethyl-[2-(1H-indol-3-yl)-ethyl]-amine.
Collapse
Affiliation(s)
- Trieu N. Trinh
- Chemistry
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| | - Eileen A. McLaughlin
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| | - Christopher P. Gordon
- Chemistry
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
- Nanoscale Organization and Dynamics Group
| | - Ilana R. Bernstein
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| | - Victoria J. Pye
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| | - Kate A. Redgrove
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| | - Adam McCluskey
- Chemistry
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Australia
| |
Collapse
|
19
|
Hall SE, Nixon B, Aitken RJ. Non-surgical sterilisation methods may offer a sustainable solution to feral horse (Equus caballus) overpopulation. Reprod Fertil Dev 2017; 29:1655-1666. [DOI: 10.1071/rd16200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/09/2016] [Indexed: 11/23/2022] Open
Abstract
Feral horses are a significant pest species in many parts of the world, contributing to land erosion, weed dispersal and the loss of native flora and fauna. There is an urgent need to modify feral horse management strategies to achieve public acceptance and long-term population control. One way to achieve this is by using non-surgical methods of sterilisation, which are suitable in the context of this mobile and long-lived species. In this review we consider the benefits of implementing novel mechanisms designed to elicit a state of permanent sterility (including redox cycling to generate oxidative stress in the gonad, random peptide phage display to target non-renewable germ cells and the generation of autoantibodies against proteins essential for conception via covalent modification) compared with that of traditional immunocontraceptive approaches. The need for a better understanding of mare folliculogenesis and conception factors, including maternal recognition of pregnancy, is also reviewed because they hold considerable potential in providing a non-surgical mechanism for sterilisation. In conclusion, the authors contend that non-surgical measures that are single shot and irreversible may provide a sustainable and effective strategy for feral horse control.
Collapse
|
20
|
Xing X, Su L, Asare PF, Wang L, Li L, Liu E, Yu B, Zhu Y, Gao X, Fan G. Danzhi Qing'e (DZQE) activates AMPK pathway and regulates lipid metabolism in a rat model of perimenopausal hyperlipidaemia. Exp Physiol 2016; 101:1406-1417. [PMID: 27456335 DOI: 10.1113/ep085786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/22/2016] [Indexed: 02/01/2023]
Abstract
What is the central question of this study? Does Danzhi Qing'e (DZQE) regulate lipid metabolism and improve ovarian function in a rat model of perimenopausal hyperlipidaemia, and could this effect be mediated through the AMPK pathway? What is the main finding and its importance? We revealed that DZQE is a pharmacotherapy that could activate the AMPK pathway to improve ovarian function and lipid metabolism during perimenopause complicated with hyperlipidaemia syndrome in an animal model. Thus, this study provides a novel therapeutic option for treating perimenopausal syndrome and highlights the therapeutic potential of DZQE in perimenopausal rats. Menopause is an important event in a woman's life. During perimenopause, accompanied by development of osteoporosis and dyslipidaemia, ovarian function gradually declines. Dyslipidaemia is a risk factor for cardiovascular disorders, cerebrovascular disease and breast cancer in postmenopausal women. All of these contribute to impairment of liver function, particularly fatty liver disease, because liver dysfunction is associated with ovarian dysfunction and hyperlipidaemia. The aim of this study was to define a therapeutic approach to improve ovarian function and attenuate lipid accumulation in order to prevent perimenopause-induced ovarian dysfunction and hyperlipidaemia. Four-week-old female Wistar rats were injected i.p. with 4-vinylcyclohexene diepoxide (4-VCD) and fed with a high-fat diet (HFD) to serve as a model of perimenopause complicated with hyperlipidaemia. The 4-VCD induces perimenopause, while the HFD causes hyperlipidaemia. Five days after administration of 4-VCD, the 4-VCD + HFD-treated rats were assessed daily for oestrous cycle stage by vaginal cytology. Rats were then assigned into groups, in which 2.5, 5.0 or 10.0 g kg-1 Danzhi Qing'e (DZQE) or estradiol valerate was administered intragastrically for 8 weeks. Expression levels of follicle-stimulating hormone (FSH), luteinizing hormone (LH), oestrogen and testosterone measured by enzyme-linked immunosorbent assay served as biomarkers for perimenopause and ovarian dysfunction. The expression levels of AMPK and acetyl-CoA carboxylase in the liver were determined with Western blotting, and aspartate aminotransferase and alanine aminotransferase were analysed using an automated biochemical analyser to examine liver function. The DZQE improved ovarian function by upregulating oestrogen and testosterone concentrations in serum and downregulating FSH and LH serum concentrations. Moreover, DZQE reduced serum concentrations of triglyceride, total cholesterol and low-density lipoprotein in a dose-dependent manner to regulate lipid levels during perimenopause. Furthermore, DZQE increased AMPK at both the transcriptional and translational levels and decreased the expression of SREBP-1c gene as well as its downstream target gene, fatty acid synthase. Danzhi Qing'e improved dyslipidaemia during menopause and also had an effect on liver function. Danzhi Qing'e is an effective Chinese herbal compound, which improves ovarian function and lipid metabolism in perimenopause complicated with hyperlipidaemia at least in part through the AMPK pathway.
Collapse
Affiliation(s)
- Xiaoxue Xing
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.,Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lina Su
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.,Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Patrick Fordjour Asare
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.,Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lingyan Wang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.,Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lan Li
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.,Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Erwei Liu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.,Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Bing Yu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.,Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yan Zhu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.,Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Xiumei Gao
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China. .,Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Guanwei Fan
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China. .,Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
21
|
Redgrove KA, Bernstein IR, Pye VJ, Mihalas BP, Sutherland JM, Nixon B, McCluskey A, Robinson PJ, Holt JE, McLaughlin EA. Dynamin 2 is essential for mammalian spermatogenesis. Sci Rep 2016; 6:35084. [PMID: 27725702 PMCID: PMC5057128 DOI: 10.1038/srep35084] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/26/2016] [Indexed: 11/09/2022] Open
Abstract
The dynamin family of proteins play important regulatory roles in membrane remodelling and endocytosis, especially within brain and neuronal tissues. In the context of reproduction, dynamin 1 (DNM1) and dynamin 2 (DNM2) have recently been shown to act as key mediators of sperm acrosome formation and function. However, little is known about the roles that these proteins play in the developing testicular germ cells. In this study, we employed a DNM2 germ cell-specific knockout model to investigate the role of DNM2 in spermatogenesis. We demonstrate that ablation of DNM2 in early spermatogenesis results in germ cell arrest during prophase I of meiosis, subsequent loss of all post-meiotic germ cells and concomitant sterility. These effects become exacerbated with age, and ultimately result in the demise of the spermatogonial stem cells and a Sertoli cell only phenotype. We also demonstrate that DNM2 activity may be temporally regulated by phosphorylation of DNM2 via the kinase CDK1 in spermatogonia, and dephosphorylation by phosphatase PPP3CA during meiotic and post-meiotic spermatogenesis.
Collapse
Affiliation(s)
- Kate A Redgrove
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,PRC in Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Ilana R Bernstein
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,PRC in Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Victoria J Pye
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,PRC in Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Bettina P Mihalas
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Jessie M Sutherland
- School of Biomedical Sciences &Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Brett Nixon
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,PRC in Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Adam McCluskey
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,PRC in Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Phillip J Robinson
- Cell Signalling Unit, Children's Medical Research Institute, University of Sydney, Sydney, NSW 2145, Australia
| | - Janet E Holt
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,PRC in Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Eileen A McLaughlin
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.,PRC in Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia.,School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| |
Collapse
|
22
|
Liu Y, Wang YL, Chen MH, Zhang Z, Xu BH, Liu R, Xu L, He SW, Li FP, Qi ZQ, Wang HL. Methoxychlor exposure induces oxidative stress and affects mouse oocyte meiotic maturation. Mol Reprod Dev 2016; 83:768-779. [DOI: 10.1002/mrd.22683] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/14/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Yu Liu
- Organ Transplantation Institute, Medical College; Xiamen University; Xiamen City Fujian Province China
| | - Ya-Long Wang
- Organ Transplantation Institute, Medical College; Xiamen University; Xiamen City Fujian Province China
| | - Ming-Huang Chen
- Department of Gynaecology and Obstetrics, Zhongshan Hospital; Xiamen University; Xiamen City Fujian Province China
| | - Zhen Zhang
- Xiamen Institute for Food and Drug Quality Control; Xiamen City Fujian Province China
| | - Bai-Hui Xu
- Organ Transplantation Institute, Medical College; Xiamen University; Xiamen City Fujian Province China
| | - Rui Liu
- Department of Gynaecology and Obstetrics, Zhongshan Hospital; Xiamen University; Xiamen City Fujian Province China
| | - Lin Xu
- Organ Transplantation Institute, Medical College; Xiamen University; Xiamen City Fujian Province China
| | - Shu-Wen He
- Organ Transplantation Institute, Medical College; Xiamen University; Xiamen City Fujian Province China
| | - Fei-Ping Li
- Biological College; Southwest Forestry University; Kunming City Yunnan Province China
| | - Zhong-Quan Qi
- Organ Transplantation Institute, Medical College; Xiamen University; Xiamen City Fujian Province China
| | - Hai-Long Wang
- Organ Transplantation Institute, Medical College; Xiamen University; Xiamen City Fujian Province China
| |
Collapse
|
23
|
Liu W, Wang LY, Xing XX, Fan GW. Conditions and possible mechanisms of VCD-induced ovarian failure. Altern Lab Anim 2016; 43:385-92. [PMID: 26753941 DOI: 10.1177/026119291504300606] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Perimenopause is an important period in women's lives, in which they experience a series of physiological changes. Current animal models of perimenopause fail to adequately replicate this particular stage in female life, while current in vitro models are too simplistic and cannot account for systemic effects. Neither the naturally-ageing animal model, nor the ovariectomised animal model, mimic the natural transitional process that is the menopause. In vivo and in vitro studies have confirmed that the occupational chemical, 4-vinylcyclohexene diepoxide (VCD), can cause selective destruction of the ovarian primordial and primary follicles of rats and mice by accelerating the apoptotic process, which successfully mimics the perimenopausal state in women. However, it is the in vivo VCD-induced rodent perimenopausal models that are currently the most widely used in research, rather than any of the available in vitro models. Studies on the mechanisms involved have found that VCD induces ovotoxicity via interference with the c-kit/kit ligand and apoptotic signalling pathways, among others. Overall, the VCD-induced perimenopausal animal models have provided some insight into female perimenopause, but they are far from ideal models of the human situation.
Collapse
Affiliation(s)
- Wei Liu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ling-Yan Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao-Xue Xing
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guan-Wei Fan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
24
|
Trinh TN, McLaughlin EA, Abdel-Hamid MK, Gordon CP, Bernstein IR, Pye V, Cossar P, Sakoff JA, McCluskey A. Quinolone-1-(2H)-ones as hedgehog signalling pathway inhibitors. Org Biomol Chem 2016; 14:6304-15. [DOI: 10.1039/c6ob00606j] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A series of quinolone-2-(1H)-ones derived from a Ugi-Knoevenagel three- and four-component reaction were prepared exhibiting low micromolar cytotoxicity against a panel of eight human cancer cell lines known to possess the Hedgehog Signalling Pathway (HSP) components, as well as the seminoma TCAM-2 cell line.
Collapse
Affiliation(s)
- Trieu N. Trinh
- Chemistry
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Callaghan
- Australia
| | - Eileen A. McLaughlin
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Callaghan
- Australia
| | - Mohammed K. Abdel-Hamid
- Chemistry
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Callaghan
- Australia
| | - Christopher P. Gordon
- Nanoscale Organization and Dynamics Group
- School of Science and Health
- University of Western Sydney
- Penrith South DC
- Australia
| | - Ilana R. Bernstein
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Callaghan
- Australia
| | - Victoria Pye
- Biology
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Callaghan
- Australia
| | - Peter Cossar
- Chemistry
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Callaghan
- Australia
| | | | - Adam McCluskey
- Chemistry
- Priority Research Centre for Chemical Biology
- University of Newcastle
- Callaghan
- Australia
| |
Collapse
|
25
|
Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, Toppari J, Zoeller RT. EDC-2: The Endocrine Society's Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr Rev 2015; 36:E1-E150. [PMID: 26544531 PMCID: PMC4702494 DOI: 10.1210/er.2015-1010] [Citation(s) in RCA: 1423] [Impact Index Per Article: 142.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 09/01/2015] [Indexed: 02/06/2023]
Abstract
The Endocrine Society's first Scientific Statement in 2009 provided a wake-up call to the scientific community about how environmental endocrine-disrupting chemicals (EDCs) affect health and disease. Five years later, a substantially larger body of literature has solidified our understanding of plausible mechanisms underlying EDC actions and how exposures in animals and humans-especially during development-may lay the foundations for disease later in life. At this point in history, we have much stronger knowledge about how EDCs alter gene-environment interactions via physiological, cellular, molecular, and epigenetic changes, thereby producing effects in exposed individuals as well as their descendants. Causal links between exposure and manifestation of disease are substantiated by experimental animal models and are consistent with correlative epidemiological data in humans. There are several caveats because differences in how experimental animal work is conducted can lead to difficulties in drawing broad conclusions, and we must continue to be cautious about inferring causality in humans. In this second Scientific Statement, we reviewed the literature on a subset of topics for which the translational evidence is strongest: 1) obesity and diabetes; 2) female reproduction; 3) male reproduction; 4) hormone-sensitive cancers in females; 5) prostate; 6) thyroid; and 7) neurodevelopment and neuroendocrine systems. Our inclusion criteria for studies were those conducted predominantly in the past 5 years deemed to be of high quality based on appropriate negative and positive control groups or populations, adequate sample size and experimental design, and mammalian animal studies with exposure levels in a range that was relevant to humans. We also focused on studies using the developmental origins of health and disease model. No report was excluded based on a positive or negative effect of the EDC exposure. The bulk of the results across the board strengthen the evidence for endocrine health-related actions of EDCs. Based on this much more complete understanding of the endocrine principles by which EDCs act, including nonmonotonic dose-responses, low-dose effects, and developmental vulnerability, these findings can be much better translated to human health. Armed with this information, researchers, physicians, and other healthcare providers can guide regulators and policymakers as they make responsible decisions.
Collapse
Affiliation(s)
- A C Gore
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - V A Chappell
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - S E Fenton
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - J A Flaws
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - A Nadal
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - G S Prins
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - J Toppari
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| | - R T Zoeller
- Pharmacology and Toxicology (A.C.G.), College of Pharmacy, The University of Texas at Austin, Austin, Texas 78734; Division of the National Toxicology Program (V.A.C., S.E.F.), National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709; Department of Comparative Biosciences (J.A.F.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61802; Institute of Bioengineering and CIBERDEM (A.N.), Miguel Hernandez University of Elche, 03202 Elche, Alicante, Spain; Departments of Urology, Pathology, and Physiology & Biophysics (G.S.P.), College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612; Departments of Physiology and Pediatrics (J.T.), University of Turku and Turku University Hospital, 20520 Turku, Finland; and Biology Department (R.T.Z.), University of Massachusetts at Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
26
|
Lefèvre PLC, Berger RG, Ernest SR, Gaertner DW, Rawn DFK, Wade MG, Robaire B, Hales BF. Exposure of Female Rats to an Environmentally Relevant Mixture of Brominated Flame Retardants Targets the Ovary, Affecting Folliculogenesis and Steroidogenesis. Biol Reprod 2015; 94:9. [PMID: 26607716 PMCID: PMC4809562 DOI: 10.1095/biolreprod.115.134452] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/13/2015] [Indexed: 11/04/2022] Open
Abstract
Brominated flame retardants (BFRs) are incorporated into various consumer products to prevent flame propagation. These compounds leach into the domestic environment, resulting in chronic exposure and contamination. Pregnancy failure is associated with high levels of BFRs in human follicular fluid, raising serious questions regarding their impact on female reproductive health. The goal of this study is to elucidate the effects of an environmentally relevant BFR mixture on female rat ovarian functions (i.e., folliculogenesis and steroidogenesis). A BFR dietary mixture formulated to mimic the relative BFR congener levels in North American house dust was administered to adult female Sprague-Dawley rats from 2 to 3 wk before mating until Gestational Day 20; these diets were designed to deliver nominal doses of 0, 0.06, 20, or 60 mg/kg/day of the BFR mixture. Exposure to BFRs triggered an approximately 50% increase in the numbers of preantral and antral follicles and an enlargement of the antral follicles in the ovaries of the dams. A significant reduction in the expression of catalase, an antioxidant enzyme, and downregulation of the expression of insulin-like factor 3 (Insl3) and 17alpha-hydroxylase (Cyp17a1) were observed in the ovary. In addition, BFR exposure affected steroidogenesis; we observed a significant decrease in circulating 17-hydroxypregnenolone and an increase in testosterone concentrations in BFR-exposed dams. Thus, BFRs target ovarian function in the rat, adversely affecting both folliculogenesis and steroidogenesis.
Collapse
Affiliation(s)
- Pavine L C Lefèvre
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Robert G Berger
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Sheila R Ernest
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Dean W Gaertner
- Food Research Division, Bureau of Chemical Safety, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada
| | - Dorothea F K Rawn
- Food Research Division, Bureau of Chemical Safety, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada
| | - Michael G Wade
- Environmental Health Science and Research Bureau, Radiation and Research Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Bernard Robaire
- Food Research Division, Bureau of Chemical Safety, Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada Department of Obstetrics and Gynecology, McGill University, Montreal, Quebec, Canada
| | - Barbara F Hales
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
27
|
Knockout of RNA Binding Protein MSI2 Impairs Follicle Development in the Mouse Ovary: Characterization of MSI1 and MSI2 during Folliculogenesis. Biomolecules 2015; 5:1228-44. [PMID: 26131972 PMCID: PMC4598749 DOI: 10.3390/biom5031228] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/29/2015] [Accepted: 06/09/2015] [Indexed: 01/20/2023] Open
Abstract
Characterizing the mechanisms underlying follicle development in the ovary is crucial to understanding female fertility and is an area of increasing research interest. The RNA binding protein Musashi is essential for post-transcriptional regulation of oocyte maturation in Xenopus and is expressed during ovarian development in Drosophila. In mammals Musashi is important for spermatogenesis and male fertility, but its role in the ovary has yet to be characterized. In this study we determined the expression of mammalian Musashi proteins MSI1 and MSI2 during mouse folliculogenesis, and through the use of a MSI2-specific knockout mouse model we identified that MSI2 is essential for normal follicle development. Time-course characterization of MSI1 and MSI2 revealed distinct differences in steady-state mRNA levels and protein expression/localization at important developmental time-points during folliculogenesis. Using a gene-trap mouse model that inactivates Msi2, we observed a significant decrease in ovarian mass, and change in follicle-stage composition due to developmental blocking of antral stage follicles and pre-antral follicle loss through atresia. We also confirmed that hormonally stimulated Msi2-deficient mice produce significantly fewer MII oocytes (60.9% less than controls, p < 0.05). Furthermore, the majority of these oocytes are of poor viability (62.2% non-viable/apoptotic, p < 0.05), which causes a reduction in female fertility evidenced by decreased litter size in Msi2-deficient animals (33.1% reduction to controls, p < 0.05). Our findings indicate that MSI1 and MSI2 display distinct expression profiles during mammalian folliculogenesis and that MSI2 is required for pre-antral follicle development.
Collapse
|
28
|
Sutherland JM, Sobinoff AP, Fraser BA, Redgrove KA, Davidson TL, Siddall NA, Koopman P, Hime GR, McLaughlin EA. RNA binding protein Musashi-1 directly targets Msi2 and Erh during early testis germ cell development and interacts with IPO5 upon translocation to the nucleus. FASEB J 2015; 29:2759-68. [PMID: 25782991 DOI: 10.1096/fj.14-265868] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/26/2015] [Indexed: 12/19/2022]
Abstract
Controlled gene regulation during gamete development is vital for maintaining reproductive potential. During the process of gamete development, male germ cells experience extended periods of inactive transcription despite requirements for continued growth and differentiation. Spermatogenesis therefore provides an ideal model to study the effects of posttranscriptional control on gene regulation. During spermatogenesis posttranscriptional regulation is orchestrated by abundantly expressed RNA-binding proteins. One such group of RNA-binding proteins is the Musashi family, previously identified as a critical regulator of testis germ cell development and meiosis in Drosophila and also shown to be vital to sperm development and reproductive potential in the mouse. We focus in depth on the role and function of the vertebrate Musashi ortholog Musashi-1 (MSI1). Through detailed expression studies and utilizing our novel transgenic Msi1 testis-specific overexpression model, we have identified 2 unique RNA-binding targets of MSI1 in spermatogonia, Msi2 and Erh, and have demonstrated a role for MSI1 in translational regulation. We have also provided evidence to suggest that nuclear import protein, IPO5, facilitates the nuclear translocation of MSI1 to the transcriptionally silenced XY chromatin domain in meiotic pachytene spermatocytes, resulting in the release of MSI1 RNA-binding targets. This firmly establishes MSI1 as a master regulator of posttranscriptional control during early spermatogenesis and highlights the significance of the subcellular localization of RNA binding proteins in relation to their function.
Collapse
Affiliation(s)
- Jessie M Sutherland
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Alexander P Sobinoff
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Barbara A Fraser
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Kate A Redgrove
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Tara-Lynne Davidson
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Nicole A Siddall
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Peter Koopman
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Gary R Hime
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Eileen A McLaughlin
- *School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia; and Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
29
|
American ginseng regulates gene expression to protect against premature ovarian failure in rats. BIOMED RESEARCH INTERNATIONAL 2015; 2015:767124. [PMID: 25705687 PMCID: PMC4330957 DOI: 10.1155/2015/767124] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/30/2014] [Accepted: 12/01/2014] [Indexed: 12/19/2022]
Abstract
Premature ovarian failure (POF) is defined as lost ovarian functions before the age of 40. Three possible molecular markers (PLA2G4A, miR-29a, and miR-144) have been identified in our previous study by integrated analysis of mRNA and miRNA expression profiles. The present study aimed to evaluate American ginseng root's protective potential against POF by studying transcriptional and protein variations between American ginseng treatments and controls in rats. 4-Vinylcyclohexene diepoxide (VCD) was administered to rats for 14 days to induce POF. Additionally, American ginseng was administered to POF rats for one month, and PLA2G4A, miR-29a, and miR-144 expressions were measured in rat ovaries by qRT-PCR. PLA2G4A protein expression was examined by Western Blot, and PGE2, LH, FSH, and E2 serum levels were detected by ELISA. PLA2G4A mRNA and protein were downregulated in American ginseng-treated rats, miR-29a and miR-144 levels increased, and PGE2 serum levels decreased, while LH, FSH, and E2 increased compared to POF induction alone. Analysis of transcriptional and protein variations suggested that American ginseng protects the ovary against POF by regulating prostaglandin biosynthesis, ovulation, and preventing ovarian aging. High hormone levels (PGE2, FSH, and LH) were reduced, and E2 secretion approached normal levels, leading to improved POF symptoms and abnormal ovulation.
Collapse
|
30
|
Sobinoff AP, Sutherland JM, Beckett EL, Stanger SJ, Johnson R, Jarnicki AG, McCluskey A, St John JC, Hansbro PM, McLaughlin EA. Damaging legacy: maternal cigarette smoking has long-term consequences for male offspring fertility. Hum Reprod 2014; 29:2719-35. [PMID: 25269568 DOI: 10.1093/humrep/deu235] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
STUDY QUESTION What are the effects on fertility of cigarette smoke-induced toxicity on male offspring exposed during the gestational/weaning period? SUMMARY ANSWER Maternal cigarette smoke exposure during the gestational/weaning period causes long-term defects in male offspring fertility. WHAT IS KNOWN ALREADY Cigarette smoke is a well-known reproductive toxicant which is particularly harmful to both fetal and neonatal germ cells. However, recent studies suggest a significant portion of young mothers in the developed world still smoke during pregnancy. In the context of male reproductive health, our understanding of the effects of in utero exposure on offspring fertility is limited. STUDY DESIGN, SIZE, DURATION In this study, 27 C57BL/6 5-week-old female mice were exposed via the nose-only to cigarette smoke (treatment) or 27 were exposed to room air (control) for 6 weeks before being housed with stud males to produce litters. In the treatment group, smoke exposure continued throughout mating, pregnancy and lactation until weaning of pups at 21 days post birth. Male offspring were examined at post-natal days 3, 6, 12, 21 and 98 (adult). PARTICIPANTS/MATERIALS, SETTING, METHODS Approximately 108 maternal smoke-exposed C57BL/6 offspring and controls were examined. Spermatogenesis was examined using testicular histology and apoptosis/DNA damage was assessed using caspase immunohistochemistry and TUNEL. Sertoli cell morphology and fluctuations in the spermatogonial stem cell population were also examined using immunohistochemistry. Microarray and QPCR analysis were performed on adult testes to examine specific long-term transcriptomic alteration as a consequence of maternal smoke exposure. Sperm counts and motility, zona/oolemma binding assays, COMET analysis and mitochondrial genomic sequencing were also performed on spermatozoa obtained from adult treated and control mice. Fertility trials using exposed adult male offspring were also performed. MAIN RESULTS AND THE ROLE OF CHANCE Maternal cigarette smoke exposure caused increased gonocyte and meiotic spermatocyte apoptosis (P < 0.01) as well as germ cell depletion in the seminiferous tubules of neonatal and juvenile offspring. Aberrant testicular development characterized by abnormal Sertoli and germ cell organization, a depleted spermatogonial stem cell population (P < 0.01), atrophic seminiferous tubules and increased germ cell DNA damage (P < 0.01) persisted in adult offspring 11 weeks after exposure. Microarray analysis of adult offspring testes associated these defects with meiotic germ cell development, sex hormone metabolism, oxidative stress and Sertoli cell signalling. Next generation sequencing also revealed a high mitochondrial DNA mutational load in the testes of adult offspring (P < 0.01). Adult maternal smoke-exposed offspring also had reduced sperm counts with spermatozoa exhibiting morphological abnormalities (P < 0.01), affecting motility and fertilization potential. Odf2, a spermatozoa flagellum component required for coordinated ciliary beating, was also significantly down-regulated (P < 0.01) in maternal smoke-exposed adult offspring, with aberrant localization along the spermatozoa flagellum. Adult maternal smoke-exposed offspring took significantly longer to impregnate control females and had a slight but significant (P < 0.01) reduction in litter size. LIMITATIONS, REASONS FOR CAUTION This study examined only one species (mouse) using a smoking model which only simulates human cigarette smoke exposure. WIDER IMPLICATIONS OF THE FINDINGS This study represents the first comprehensive animal model of maternal smoking on male offspring reproductive function, suggesting that exposure during the gestational/weaning period causes long-term defects in male offspring fertility. This is due to a compromised spermatogonial stem cell population resulting from gonocyte apoptosis and impaired spermatogenic development. This results in significant germ cell damage and Sertoli cell dysfunction, impacting germ cell number, tubule organization, DNA damage and spermatozoa in adult offspring. This study strengthens the current literature suggesting that maternal exposure impairs male offspring fertility, which is currently debated due to conflicting studies. STUDY FUNDING/COMPETING INTERESTS This study was funded by the Australian Research Council, Hunter Medical Research Institute, National Health and Medical Research Council of Australia and the Newcastle Permanent Building Society Charitable Trust. The authors declare no conflict of interest.
Collapse
Affiliation(s)
- A P Sobinoff
- Reproductive Science Group, School of Environmental & Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia Priority Research Centre for Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia
| | - J M Sutherland
- Reproductive Science Group, School of Environmental & Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia Priority Research Centre for Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia
| | - E L Beckett
- Centre for Asthma and Respiratory Disease, University of Newcastle and Hunter Medical Research Institute, Newcastle, Callaghan, NSW 2308, Australia
| | - S J Stanger
- Reproductive Science Group, School of Environmental & Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia Priority Research Centre for Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia
| | - R Johnson
- Centre for Asthma and Respiratory Disease, University of Newcastle and Hunter Medical Research Institute, Newcastle, Callaghan, NSW 2308, Australia
| | - A G Jarnicki
- Centre for Asthma and Respiratory Disease, University of Newcastle and Hunter Medical Research Institute, Newcastle, Callaghan, NSW 2308, Australia
| | - A McCluskey
- Priority Research Centre for Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia
| | - J C St John
- Centre for Genetic Diseases, MIMR-PHI Institute of Medical Research, Monash University, 27-31 Wright Street, Clayton Vic 3168, Australia
| | - P M Hansbro
- Centre for Asthma and Respiratory Disease, University of Newcastle and Hunter Medical Research Institute, Newcastle, Callaghan, NSW 2308, Australia
| | - E A McLaughlin
- Reproductive Science Group, School of Environmental & Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia Priority Research Centre for Chemical Biology, University of Newcastle, Callaghan, NSW 2308, Australia Monash Medical Centre, Monash Institute of Medical Research, Clayton, VIC 3168, Australia
| |
Collapse
|
31
|
Sutherland JM, Fraser BA, Sobinoff AP, Pye VJ, Davidson TL, Siddall NA, Koopman P, Hime GR, McLaughlin EA. Developmental Expression of Musashi-1 and Musashi-2 RNA-Binding Proteins During Spermatogenesis: Analysis of the Deleterious Effects of Dysregulated Expression1. Biol Reprod 2014; 90:92. [DOI: 10.1095/biolreprod.113.115261] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
32
|
Mai Z, Lei M, Yu B, Du H, Liu J. The effects of cigarette smoke extract on ovulation, oocyte morphology and ovarian gene expression in mice. PLoS One 2014; 9:e95945. [PMID: 24776817 PMCID: PMC4002431 DOI: 10.1371/journal.pone.0095945] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 04/01/2014] [Indexed: 12/14/2022] Open
Abstract
Cigarette smoking can harm fertility, but the existing research has targeted primarily on ovarian follicles, embryos or sex hormone. In this study, we tested cigarette smoke extract on ovulation, oocyte morphology and ovarian gene expression associated with inhibition of oxidative stress using C57BL/6 mice. Mice in the experimental group were administered a cigarette smoke extract (CSE) solution (2 mg/ml) orally daily, while the blank control group was given dimethylsulfoxide (DMSO). A positive control group (menadione) was used that received an intraperitoneal injection of 15 mg/kg menadione in oil solution daily. We found that the CSE group manifested a reduced diameter of zona pellucida-free oocyte (ZP-free OD) and a morphologically misshapen first polar body (PB). Our results suggest that CSE exposure is associated with a shrink size and poor quality of oocytes. Quitting smoking is a wise choice to ensure good fertility.
Collapse
Affiliation(s)
- Zixin Mai
- Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ming Lei
- Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bolan Yu
- Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hongzi Du
- Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianqiao Liu
- Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- * E-mail:
| |
Collapse
|
33
|
|
34
|
Kuang H, Han D, Xie J, Yan Y, Li J, Ge P. Profiling of differentially expressed microRNAs in premature ovarian failure in an animal model. Gynecol Endocrinol 2014; 30:57-61. [PMID: 24188450 DOI: 10.3109/09513590.2013.850659] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Premature ovarian failure (POF) contributes to amenorrhoea, infertility, early onset of menostasia and osteoporosis. This study profiled differentially expressed miRNAs for association with POF development. Ovarian tissue samples from 4-vinylcyclohexene diepoxide (VCD)-induced rat POF and normal rats were profiled for differentially expressed miRNAs using miRNA microarrays. A total of 63 miRNAs were up-regulated and 20 miRNAs were down-regulated in rat POF tissues versus the control tissues. qRT-PCR verified some of these altered miRNAs, i.e. miR-29a and miR-144 were down-regulated in POF tissues, which may target expression of PLA2G4A that is involved in prostaglandin biosynthesis, whereas miR-27b and miR-190 were up-regulated in POF tissues by negative control of PAPPA and CCL2 expression, respectively, both of which have been shown to relate to response to hormone stimulus. Moreover, the up-regulated miR-151 and miR-672 can also target expression of TNFSF10 and FNDC1, which have been shown to positively regulate cell apoptosis. Profiling of differentially expressed miRNAs in POF provided a novel insight into the molecular events involving the role of miRNAs in POF development with specific emphasis upon miR-27b, miR-190, miR-151, miR-672, miR-29a and miR-144.
Collapse
Affiliation(s)
- Haixue Kuang
- The Key Laboratory of Chinese Ministry of Education, Department of Pharmacology
| | | | | | | | | | | |
Collapse
|
35
|
Liu Y, He XQ, Huang X, Ding L, Xu L, Shen YT, Zhang F, Zhu MB, Xu BH, Qi ZQ, Wang HL. Resveratrol protects mouse oocytes from methylglyoxal-induced oxidative damage. PLoS One 2013; 8:e77960. [PMID: 24194906 PMCID: PMC3806792 DOI: 10.1371/journal.pone.0077960] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/05/2013] [Indexed: 11/29/2022] Open
Abstract
Methylglyoxal, a reactive dicarbonyl compound, is mainly formed from glycolysis. Methylglyoxal can lead to the dysfunction of mitochondria, the depletion of cellular anti-oxidation enzymes and the formation of advanced glycation ends. Previous studies showed that the accumulation of methylglyoxal and advanced glycation ends can impair the oocyte maturation and reduce the oocyte quality in aged and diabetic females. In this study, we showed that resveratrol, a kind of phytoalexin found in the skin of grapes, red wine and other botanical extracts, can alleviate the adverse effects caused by methylglyoxal, such as inhibition of oocyte maturation and disruption of spindle assembly. Besides, methylglyoxal-treated oocytes displayed more DNA double strands breaks and this can also be decreased by treatment of resveratrol. Further investigation of these processes revealed that methylglyoxal may affect the oocyte quality by resulting in excessive reactive oxygen species production, aberrant mitochondrial distribution and high level lipid peroxidation, and resveratrol can block these cytotoxic changes. Collectively, our results showed that resveratrol can protect the oocytes from methylglyoxal-induced cytotoxicity and this was mainly through the correction of the abnormity of cellular reactive oxygen species metabolism.
Collapse
Affiliation(s)
- Yu Liu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China
| | - Xiao-Qin He
- Department of Gynaecology and Obstetrics, Zhongshan Hospital, Xiamen University, Xiamen City, Fujian Province, China
| | - Xin Huang
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China
| | - Lu Ding
- Center of Reproductive Medicine, Xiamen Maternity and Child Health Care Hospital, Xiamen City, Fujian Province, China
| | - Lin Xu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China
| | - Yu-Ting Shen
- Department of Gynaecology and Obstetrics, Zhongshan Hospital, Xiamen University, Xiamen City, Fujian Province, China
| | - Fei Zhang
- Department of Gynaecology and Obstetrics, Zhongshan Hospital, Xiamen University, Xiamen City, Fujian Province, China
| | - Mao-Bi Zhu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China
| | - Bai-Hui Xu
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China
| | - Zhong-Quan Qi
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China
- * E-mail: (H-LW); (Z-QQ)
| | - Hai-Long Wang
- Organ Transplantation Institute, Medical College, Xiamen University, Xiamen City, Fujian Province, China
- * E-mail: (H-LW); (Z-QQ)
| |
Collapse
|
36
|
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
|
37
|
Sobinoff AP, Beckett EL, Jarnicki AG, Sutherland JM, McCluskey A, Hansbro PM, McLaughlin EA. Scrambled and fried: cigarette smoke exposure causes antral follicle destruction and oocyte dysfunction through oxidative stress. Toxicol Appl Pharmacol 2013; 271:156-67. [PMID: 23693141 DOI: 10.1016/j.taap.2013.05.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/07/2013] [Accepted: 05/08/2013] [Indexed: 01/09/2023]
Abstract
Cigarette smoke is a reproductive hazard associated with pre-mature reproductive senescence and reduced clinical pregnancy rates in female smokers. Despite an increased awareness of the adverse effects of cigarette smoke exposure on systemic health, many women remain unaware of the adverse effects of cigarette smoke on female fertility. This issue is compounded by our limited understanding of the molecular mechanisms behind cigarette smoke induced infertility. In this study we used a direct nasal exposure mouse model of cigarette smoke-induced chronic obstructive pulmonary disease to characterise mechanisms of cigarette-smoke induced ovotoxicity. Cigarette smoke exposure caused increased levels of primordial follicle depletion, antral follicle oocyte apoptosis and oxidative stress in exposed ovaries, resulting in fewer follicles available for ovulation. Evidence of oxidative stress also persisted in ovulated oocytes which escaped destruction, with increased levels of mitochondrial ROS and lipid peroxidation resulting in reduced fertilisation potential. Microarray analysis of ovarian tissue correlated these insults with a complex mechanism of ovotoxicity involving genes associated with detoxification, inflammation, follicular activation, immune cell mediated apoptosis and membrane organisation. In particular, the phase I detoxifying enzyme cyp2e1 was found to be significantly up-regulated in developing oocytes; an enzyme known to cause molecular bioactivation resulting in oxidative stress. Our results provide a preliminary model of cigarette smoke induced sub-fertility through cyp2e1 bioactivation and oxidative stress, resulting in developing follicle depletion and oocyte dysfunction.
Collapse
Affiliation(s)
- A P Sobinoff
- Reproductive Science Group, School of Environmental & Life Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | | | | | | | | | | | | |
Collapse
|
38
|
Sattarova EA, Sinitsyna OI, Vasyunina EA, Duzhak AB, Kolosova NG, Zharkov DO, Nevinsky GA. Age-dependent guanine oxidation in DNA of different brain regions of Wistar rats and prematurely aging OXYS rats. Biochim Biophys Acta Gen Subj 2013; 1830:3542-52. [PMID: 23403132 DOI: 10.1016/j.bbagen.2013.01.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 01/17/2013] [Accepted: 01/21/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND Oxidative damage to the cell, including the formation of 8-oxoG, has been regarded as a significant factor in carcinogenesis and aging. An inbred prematurely aging rat strain (OXYS) is characterized by high sensitivity to oxidative stress, lipid peroxidation, protein oxidation, DNA rearrangements, and pathological conditions paralleling several human degenerative diseases including learning and memory deterioration. METHODS We have used monoclonal antibodies against a common pre-mutagenic base lesion 8-oxoguanine (8-oxoG) and 8-oxoguanine DNA glycosylase (OGG1) in combination with indirect immunofluorescence microscopy and image analysis to follow the relative amounts and distribution of 8-oxoG and OGG1 in various cells of different brain regions from OXYS and control Wistar rats. RESULTS It was shown that 8-oxoG increased with age in mature neurons, nestin- and glial fibrillary acidic protein (GFAP)-positive cells of hippocampus and frontal cortex in both strains of rats, with OXYS rats always displaying statistically significantly higher levels of oxidative DNA damage than Wistar rats. The relative content of 8-oxoG and OGG1 in nestin- and GFAP-positive cells was higher than in mature neurons in both Wistar and OXYS rats. However, there was no significant interstrain difference in the content of OGG1 for all types of cells and brain regions analyzed, and no difference in the relative content of 8-oxoG between different brain regions. CONCLUSIONS Oxidation of guanine may play an important role in the development of age-associated decrease in memory and learning capability of OXYS rats. GENERAL SIGNIFICANCE The findings are important for validation of the OXYS rat strain as a model of mammalian aging.
Collapse
Affiliation(s)
- Evgeniya A Sattarova
- SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | | | | | | | | | | | | |
Collapse
|
39
|
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
|
40
|
Sobinoff AP, Nixon B, Roman SD, McLaughlin EA. Staying alive: PI3K pathway promotes primordial follicle activation and survival in response to 3MC-induced ovotoxicity. Toxicol Sci 2012; 128:258-71. [PMID: 22505044 DOI: 10.1093/toxsci/kfs137] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
3-Methylcholanthrene (3MC) is a potent ovotoxicant capable of causing premature ovarian failure through primordial follicle depletion. Despite 3MCs ovotoxicity having been established for 30 years, relatively little information exists on the mechanisms. In this study, we examined the effects of 3MC exposure on the immature ovarian follicle population. Microarray analysis revealed a complex mechanism of 3MC-induced ovotoxicity involving a number of cellular processes associated with xenobiotic metabolism, ovarian cancer, cell cycle progression, and cell death. 3MC exposure was also found to induce developing follicle atresia and aberrant primordial follicle activation via the stimulation of PI3K/Akt and mammalian target of rapamycin (mTOR) signaling pathways. Inhibition of PI3K/Akt signaling resulted in the severe depletion of the primordial follicle pool, with further analysis identifying increased Akt1-stimulated Bad phosphoinhibition in 3MC-treated primordial follicles. Our results suggest that the primordial follicle pool enters a "prosurvival" state upon 3MC exposure and that its depletion is due to a vicious cycle of primordial follicle activation in an attempt to replace developing follicles undergoing follicular atresia.
Collapse
Affiliation(s)
- Alexander P Sobinoff
- Reproductive Science Group, School of Environmental & Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia
| | | | | | | |
Collapse
|
41
|
Sobinoff A, Pye V, Nixon B, Roman S, McLaughlin E. Jumping the gun: Smoking constituent BaP causes premature primordial follicle activation and impairs oocyte fusibility through oxidative stress. Toxicol Appl Pharmacol 2012; 260:70-80. [DOI: 10.1016/j.taap.2012.01.028] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 01/30/2012] [Accepted: 01/31/2012] [Indexed: 12/13/2022]
|
42
|
Sobinoff AP, Mahony M, Nixon B, Roman SD, McLaughlin EA. Understanding the Villain: DMBA-Induced Preantral Ovotoxicity Involves Selective Follicular Destruction and Primordial Follicle Activation through PI3K/Akt and mTOR Signaling. Toxicol Sci 2011; 123:563-75. [DOI: 10.1093/toxsci/kfr195] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
43
|
Van Kempen TA, Milner TA, Waters EM. Accelerated ovarian failure: a novel, chemically induced animal model of menopause. Brain Res 2011; 1379:176-87. [PMID: 21211517 PMCID: PMC3078694 DOI: 10.1016/j.brainres.2010.12.064] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 12/17/2010] [Accepted: 12/20/2010] [Indexed: 10/18/2022]
Abstract
Current rodent models of menopause fail to adequately recapitulate the menopause transition. The intact aging model fails to achieve very low estrogen levels, and the ovariectomy model lacks a perimenopause phase. A new rodent model of accelerated ovarian failure (AOF) successfully replicates human perimenopause and postmenopause, including estrous acyclicity and fluctuating, followed by undetectable, estrogen levels, and allows for the dissociation of the effects of hormone levels from the effects of aging. In this model, an ovotoxic chemical, 4-vinylcyclohexene diepoxide (VCD), selective for primary and primordial follicles, is injected intraperitonelly in animals for 15 days. As the mature follicle population is depleted through natural cycling, ovarian failure follows increasing periods of acyclity. Administered at low doses, VCD specifically causes apoptotic cell death of primordial follicles but does not affect other peripheral tissues, including the liver and spleen, nor does it affect brain inflammation markers. In addition to reducing confounds associated with genetic and surgical manipulations, the AOF model maintains the presence of ovarian tissue which importantly parallels to the menopause transition in humans. The VCD injection procedure can be applied to studies using transgenic or knockout mice strains, or in other disease-state models (e.g., ischemia, atherosclerosis, or diabetes). This AOF model of menopause will generate new insights into women's health particularly in determining the critical periods (i.e., a window of opportunity) during perimenopause for restoring ovarian hormones for the most efficacious effect on memory and mood disorders as well as other menopausal symptoms.
Collapse
Affiliation(s)
- Tracey A Van Kempen
- Division of Neurobiology, Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA.
| | | | | |
Collapse
|