1
|
Sangkate M, Yama P, Suriard A, Butmata W, Thammakhantha S, Daoloy N, Taweechaipaisankul A, Lin CJ, Tang PC, Moonmanee T, Jitjumnong J. Effect of eCG on Terminal Follicular Growth and Corpus Luteum Development and Blood Perfusion in Estrous-Synchronized White Lamphun Cattle. Animals (Basel) 2025; 15:867. [PMID: 40150396 PMCID: PMC11939663 DOI: 10.3390/ani15060867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/08/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Equine chorionic gonadotropin (eCG) has been shown to improve reproductive performance in cattle by stimulating terminal follicular growth. This study was designed to assess the effects of 400 IU of eCG on TFG and its consequences for luteal development in indigenous cows undergoing ovulation synchronization. Specifically, we investigated the follicular and luteal dynamics following eCG treatment using structural markers (follicular and luteal diameters and luteal area) and functional markers (P4 profiles and luteal blood perfusion) to evaluate CL development. A total of twelve cows were assigned into two groups: an untreated control group and an eCG-treated group (n = 6 in each). Ultrasonography was used to scan the ovaries and monitor the development of TFG and ovulation from day 0 to ovulation. The diameter in TFG was measured daily, and the terminal follicle (TF) was identified as the preovulatory follicle throughout the study period. Ovulation was defined by the disappearance of the TF. Following ovulation, the progression of CL development in both groups was observed on days 5-6, and the mid-stage CL was identified between days 9 and 12 post-ovulation. The results indicated a faster increase in the terminal follicular diameter (TFD). From day 4 to ovulation, the eCG-treated group showed a significantly greater terminal follicular growth rate (TFGR) compared to that in the untreated control group. This enhanced follicular growth in the eCG-treated group was associated with more robust CL blood perfusion and maturation. The Doppler imaging assessments revealed a significant increase in the blood perfusion within the CL, which corresponded with the accelerated TFG. Pearson's correlation analysis showed strong positive relationships between follicular growth, luteal function, and CL blood perfusion, underscoring the interconnectivity of these processes. In conclusion, our results highlighted the key role that eCG administration has in TFG and CL development and function in White Lamphun cattle.
Collapse
Affiliation(s)
- Molarat Sangkate
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (M.S.); (P.Y.); (A.S.); (W.B.); (S.T.); (N.D.); (T.M.)
| | - Punnawut Yama
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (M.S.); (P.Y.); (A.S.); (W.B.); (S.T.); (N.D.); (T.M.)
- Department of Animal Science and Fisheries, Faculty of Agricultural Science and Technology Lanna, Phitsanulok 65000, Thailand
| | - Atsawadet Suriard
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (M.S.); (P.Y.); (A.S.); (W.B.); (S.T.); (N.D.); (T.M.)
| | - Wichayaporn Butmata
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (M.S.); (P.Y.); (A.S.); (W.B.); (S.T.); (N.D.); (T.M.)
| | - Setthawut Thammakhantha
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (M.S.); (P.Y.); (A.S.); (W.B.); (S.T.); (N.D.); (T.M.)
| | - Noppanit Daoloy
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (M.S.); (P.Y.); (A.S.); (W.B.); (S.T.); (N.D.); (T.M.)
| | | | - Chih-Jen Lin
- Centre for Reproductive Health, Institute for Regeneration and Repair (IRR), University of Edinburgh, Edinburgh EH16 4 UU, UK;
| | - Pin-Chi Tang
- Department of Animal Science, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung 40227, Taiwan;
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
| | - Tossapol Moonmanee
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (M.S.); (P.Y.); (A.S.); (W.B.); (S.T.); (N.D.); (T.M.)
- Functional Feed Innovation Center, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jakree Jitjumnong
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand; (M.S.); (P.Y.); (A.S.); (W.B.); (S.T.); (N.D.); (T.M.)
- Functional Feed Innovation Center, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
2
|
Han Y, Lu P, Yu Y, Gu W, Li C, Lv Y, Qu X, Zhang Y, Xu Q, Yao S, Chen X, Jin Y. miRNA-125a regulates porcine oocyte maturation in vitro by targeting ADAR. Theriogenology 2025; 235:184-193. [PMID: 39842224 DOI: 10.1016/j.theriogenology.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Follicular fluid extracellular vesicles are beneficial for in vitro oocyte maturation and development; however, their effect on the expression profiles of oocyte microRNAs (miRNAs) and the roles of related miRNAs are unknown. In this study, we aimed to investigate miRNA expression in mature oocytes cultured in follicular fluid extracellular vesicles and the effect of miRNA-125a (miR-125a) on oocyte maturation. The expression profiles of the miRNAs were determined by microRNA sequencing, followed by target gene prediction analysis. We transfected miR-125a mimics and an miR-125a inhibitor to evaluate the effect of modulated miRNA-125a on cumulus expansion, oocyte maturation rate, changes in cytoplasmic maturation-related indicators, and changes in the expression of oocyte maturation-related, cumulus expansion-related, and predicted target genes. We found that miR-125a overexpression decreased the levels of cumulus expansion-related, oocyte maturation-related, and predicted target genes, adenosine deaminase RNA specific (ADAR), and lipid droplet number, and it increased the percentage of oocytes with abnormal cortical granule distribution. Inhibiting miR-125a increased the expression levels of oocyte maturation-related and target genes, number of lipid droplets, and endoplasmic reticulum function, and it decreased lipid droplet size. Mitochondrial membrane potential and reactive oxygen species levels were not significantly different between groups. In conclusion, our results suggest that extracellular vesicles may improve oocyte quality by modulating ADAR through regulating miR-125a.
Collapse
Affiliation(s)
- Yue Han
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133000, China
| | - Ping Lu
- Department of Oncology, Yanbian University Affiliated Hospital, Yanji, 133000, China
| | - Yongsheng Yu
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Sciences, Gongzhuling, 136100, China
| | - Weiyu Gu
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133000, China
| | - Chunyu Li
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133000, China
| | - Yanqiu Lv
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133000, China
| | - Xinglin Qu
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133000, China
| | - Yuyang Zhang
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133000, China
| | - Qinglong Xu
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133000, China
| | - Shunfa Yao
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133000, China
| | - Xuan Chen
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133000, China.
| | - Yi Jin
- Department of Animal Science, College of Agriculture, Yanbian University, Yanji, 133000, China.
| |
Collapse
|
3
|
Uttam V, Vohra V, Chhotaray S, Santhosh A, Diwakar V, Patel V, Gahlyan RK. Exome-wide comparative analyses revealed differentiating genomic regions for performance traits in Indian native buffaloes. Anim Biotechnol 2024; 35:2277376. [PMID: 37934017 DOI: 10.1080/10495398.2023.2277376] [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] [Indexed: 11/08/2023]
Abstract
In India, 20 breeds of buffalo have been identified and registered, yet limited studies have been conducted to explore the performance potential of these breeds, especially in the Indian native breeds. This study is a maiden attempt to delineate the important variants and unique genes through exome sequencing for milk yield, milk composition, fertility, and adaptation traits in Indian local breeds of buffalo. In the present study, whole exome sequencing was performed on Chhattisgarhi (n = 3), Chilika (n = 4), Gojri (n = 3), and Murrah (n = 4) buffalo breeds and after stringent quality control, 4333, 6829, 4130, and 4854 InDels were revealed, respectively. Exome-wide FST along 100-kb sliding windows detected 27, 98, 38, and 35 outlier windows in Chhattisgarhi, Chilika, Gojri, and Murrah, respectively. The comparative exome analysis of InDels and subsequent gene ontology revealed unique breed specific genes for milk yield (CAMSAP3), milk composition (CLCN1, NUDT3), fertility (PTGER3) and adaptation (KCNA3, TH) traits. Study provides insight into mechanism of how these breeds have evolved under natural selection, the impact of these events on their respective genomes, and their importance in maintaining purity of these breeds for the traits under study. Additionally, this result will underwrite to the genetic acquaintance of these breeds for breeding application, and in understanding of evolution of these Indian local breeds.
Collapse
Affiliation(s)
- Vishakha Uttam
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Vikas Vohra
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Supriya Chhotaray
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Ameya Santhosh
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Vikas Diwakar
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Vaibhav Patel
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Rajesh Kumar Gahlyan
- Animal Genetics & Breeding Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| |
Collapse
|
4
|
Han Y, Zhang J, Liang W, Lv Y, Luo X, Li C, Qu X, Zhang Y, Gu W, Chen X, Jin Y. Follicular fluid exosome-derived miR-339-5p enhances in vitro maturation of porcine oocytes via targeting SFPQ, a regulator of the ERK1/2 pathway. Theriogenology 2024; 225:107-118. [PMID: 38805993 DOI: 10.1016/j.theriogenology.2024.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024]
Abstract
In this study, we aimed to investigate cytoplasmic maturation and miRNA expression of mature oocytes cultured in porcine follicular fluid exosomes. We also examined the effect of miR-339-5p on oocyte maturation. Twenty eight differentially expressed miRNAs were detected using miRNA-seq. We then transfected cumulus oocyte complexes with miR-339-5p mimics and inhibitor during culture. The results showed that exosomes increased endoplasmic reticulum levels and the amount of lipid droplets, and decreased ROS levels, lipid droplet size, and percentage of oocytes with abnormal cortical granule distribution. Overexpressing miR-339-5p significantly decreased cumulus expansion genes, oocyte maturation-related genes, target gene proline/glutamine-rich splicing factor (SFPQ), ERK1/2 phosphorylation levels, oocyte maturation rate, blastocyst rate, and lipid droplet number, but increased lipid droplet size and the ratio of oocytes with abnormal cortical granule distribution. Inhibiting miR-339-5p reversed the decrease observed during overexpression. Mitochondrial membrane potential and ROS levels did not differ significantly between groups. In summary, exosomes promote oocyte cytoplasmic maturation and miR-339-5p regulating ERK1/2 activity through SFPQ expression, thereby elevating oocyte maturation and blastocyst formation rate in vitro.
Collapse
Affiliation(s)
- Yue Han
- Yanbian University, Jilin, Yanji, 133000, China
| | | | | | - Yanqiu Lv
- Yanbian University, Jilin, Yanji, 133000, China
| | - Xiaotong Luo
- Institute of Animal Biotechnology, Jilin Academy of Agricultural Sciences, Jilin, Gongzhuling, 136100, China
| | - Chunyu Li
- Yanbian University, Jilin, Yanji, 133000, China
| | - Xinglin Qu
- Yanbian University, Jilin, Yanji, 133000, China
| | | | - Weiyu Gu
- Yanbian University, Jilin, Yanji, 133000, China
| | - Xuan Chen
- Yanbian University, Jilin, Yanji, 133000, China.
| | - Yi Jin
- Yanbian University, Jilin, Yanji, 133000, China.
| |
Collapse
|
5
|
Guo Y, Wang S, Wu X, Zhao R, Chang S, Ma C, Song S, Zeng S. Multi-Omics Reveals the Role of Arachidonic Acid Metabolism in the Gut-Follicle Axis for the Antral Follicular Development of Holstein Cows. Int J Mol Sci 2024; 25:9521. [PMID: 39273467 PMCID: PMC11395146 DOI: 10.3390/ijms25179521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
In vitro embryonic technology is crucial for improving farm animal reproduction but is hampered by the poor quality of oocytes and insufficient development potential. This study investigated the relationships among changes in the gut microbiota and metabolism, serum features, and the follicular fluid metabolome atlas. Correlation network maps were constructed to reveal how the metabolites affect follicular development by regulating gene expression in granulosa cells. The superovulation synchronization results showed that the number of follicle diameters from 4 to 8 mm, qualified oocyte number, cleavage, and blastocyst rates were improved in the dairy heifers (DH) compared with the non-lactating multiparous dairy cows (NDC) groups. The gut microbiota was decreased in Rikenellaceae_RC9_gut_group, Alistipes, and Bifidobacterium, but increased in Firmicutes, Cyanobacteria, Fibrobacterota, Desulfobacterota, and Verrucomicrobiota in the NDC group, which was highly associated with phospholipid-related metabolites of gut microbiota and serum. Metabolomic profiling of the gut microbiota, serum, and follicular fluid further demonstrated that the co-metabolites were phosphocholine and linoleic acid. Moreover, the expression of genes related to arachidonic acid metabolism in granulosa cells was significantly correlated with phosphocholine and linoleic acid. The results in granulosa cells showed that the levels of PLCB1 and COX2, participating in arachidonic acid metabolism, were increased in the DH group, which improved the concentrations of PGD2 and PGF2α in the follicular fluid. Finally, the expression levels of apoptosis-related proteins, cytokines, and steroidogenesis-related genes in granulosa cells and the concentrations of steroid hormones in follicular fluid were determinants of follicular development. According to our results, gut microbiota-related phosphocholine and linoleic acid participate in arachidonic acid metabolism in granulosa cells through the gut-follicle axis, which regulates follicular development. These findings hold promise for enhancing follicular development and optimizing oocyte quality in subfertile dairy cows.
Collapse
Affiliation(s)
- Yajun Guo
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shiwei Wang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xuan Wu
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Rong Zhao
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Siyu Chang
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chen Ma
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shuang Song
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shenming Zeng
- State Key Laboratory of Animal Biotech Breeding, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
6
|
Goetten ALF, Barreta MH, Pinto da Silva Y, Bertolin K, Koch J, Rocha CC, Dias Gonçalves PB, Price CA, Antoniazzi AQ, Portela VM. FGF18 impairs blastocyst viability, DNA double-strand breaks and maternal recognition of pregnancy genes. Theriogenology 2024; 225:81-88. [PMID: 38796960 DOI: 10.1016/j.theriogenology.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Embryonic mortality in cattle is high, reaching 10-40 % in vivo and 60-70 % in vitro. Death of embryos involves reduced expression of genes related to embryonic viability, inhibition of DNA repair and increased DNA damage. In follicular granulosa cells, FGF18 from the theca layer increases apoptosis and DNA damage, so we hypothesized that FGF18 may also affect the oocyte and contribute to early embryonic death. The aims of this study were to identify the effects of FGF18 on cumulus expansion, oocyte maturation and embryo development from cleavage to blastocyst stage using a conventional bovine in vitro embryo production system using ovaries of abattoir origin. Addition of FGF18 during in-vitro maturation did not affect FSH-induced cumulus expansion or rates of nuclear maturation. When FGF18 was present in the culture system, rates of cleavage were not affected however, blastocyst and expanded blastocyst development was substantially inhibited (P < 0.05), indicating a delay of blastulation. The number of phosphorylated histone H2AFX foci per nucleus, a marker of DNA damage, was higher in cleavage-stage embryos cultured with FGF18 than in those from control group (P < 0.05). Furthermore, FGF18 decreased accumulation of PTGS2 and IFNT2 mRNA in blastocysts. In conclusion, these novel findings suggest that FGF18 plays a role in the regulation of embryonic death during the early stages of development by impairing DNA double-strand break repair and expression of genes associated with embryo viability and maternal recognition of pregnancy during the progression from oocyte to expanded blastocysts.
Collapse
Affiliation(s)
- André Lucio Fontana Goetten
- Laboratory of Animal Reproduction Physiology, LAFRA, Federal University of Santa Catarina, Curitibanos, SC, Brazil
| | - Marcos Henrique Barreta
- Laboratory of Animal Reproduction Physiology, LAFRA, Federal University of Santa Catarina, Curitibanos, SC, Brazil
| | - Yago Pinto da Silva
- Laboratory of Animal Reproduction Physiology, LAFRA, Federal University of Santa Catarina, Curitibanos, SC, Brazil
| | - Kalyne Bertolin
- Biotechnology and Animal Reproduction Laboratory, BioRep, Federal University of Santa Maria, RS, Brazil
| | - Júlia Koch
- Biotechnology and Animal Reproduction Laboratory, BioRep, Federal University of Santa Maria, RS, Brazil
| | - Cecilia Constantino Rocha
- Laboratory of Animal Reproduction Physiology, LAFRA, Federal University of Santa Catarina, Curitibanos, SC, Brazil
| | - Paulo Bayard Dias Gonçalves
- Biotechnology and Animal Reproduction Laboratory, BioRep, Federal University of Santa Maria, RS, Brazil; Molecular and Integrative Physiology of Reproduction Laboratory, MINT, Federal University of Pampa, Uruguaiana, RS, Brazil
| | - Christopher Alan Price
- Centre de Recherche en Reproduction et Fertilité, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, QC, Canada
| | - Alfredo Quites Antoniazzi
- Biotechnology and Animal Reproduction Laboratory, BioRep, Federal University of Santa Maria, RS, Brazil
| | - Valerio Marques Portela
- Biotechnology and Animal Reproduction Laboratory, BioRep, Federal University of Santa Maria, RS, Brazil.
| |
Collapse
|
7
|
Yu J, Wei Y, Zhang Z, Chen J, Fu R, Ye P, Chen S, Yang J. Metabolomic Analysis of Follicular Fluid in Normal-Weight Patients with Polycystic Ovary Syndrome. Biomedicines 2024; 12:1810. [PMID: 39200274 PMCID: PMC11352029 DOI: 10.3390/biomedicines12081810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
BACKGROUND This study aimed to examine the differential variations in the metabolic composition of follicular fluid (FF) among normal-weight patients with polycystic ovary syndrome (PCOS) and controls and to identify potential biomarkers that may offer insights into the early identification and management of these patients. METHODS We collected FF samples from 45 normal-weight women with PCOS and 36 normal-weight controls without PCOS who were undergoing in vitro fertilization-embryo transfer. An untargeted metabolomic study of collected FF from infertile women was performed using high-performance liquid chromatography-tandem spectrometry (LC-MS). The tendency of the two groups to separate was demonstrated through multivariate analysis. Univariate analysis and variable importance in projection were used to screen out differential metabolites. Metabolic pathway analysis was conducted using the Kyoto Encyclopedia of Genes and Genomes (KEGG), and a diagnostic model was established using the random forest algorithm. RESULTS The metabolomics analysis revealed an increase in the expression of 23 metabolites and a decrease in that of 10 metabolites in the FF of normal-weight women with PCOS. According to the KEGG pathway analysis, these differential metabolites primarily participated in the metabolism of glycerophospholipids and the biosynthesis of steroid hormones. Based on the biomarker combination of the top 10 metabolites, the area under the curve value was 0.805. The concentrations of prostaglandin E2 in the FF of individuals with PCOS exhibited an inverse association with the proportion of high-quality embryos (p < 0.05). CONCLUSIONS Our research identified a distinct metabolic profile of the FF from normal-weight women with PCOS. The results offer a broader comprehension of the pathogenesis and advancement of PCOS, and the detected differential metabolites could be potential biomarkers and targets for the treatment of PCOS.
Collapse
Affiliation(s)
- Jiayue Yu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; (J.Y.); (Y.W.); (J.C.)
| | - Yiqiu Wei
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; (J.Y.); (Y.W.); (J.C.)
| | - Zhourui Zhang
- The Institute for Advanced Studies, Wuhan University, Wuhan 430072, China; (Z.Z.); (R.F.)
| | - Jiao Chen
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; (J.Y.); (Y.W.); (J.C.)
| | - Rongrong Fu
- The Institute for Advanced Studies, Wuhan University, Wuhan 430072, China; (Z.Z.); (R.F.)
| | - Peng Ye
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Suming Chen
- The Institute for Advanced Studies, Wuhan University, Wuhan 430072, China; (Z.Z.); (R.F.)
| | - Jing Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; (J.Y.); (Y.W.); (J.C.)
| |
Collapse
|
8
|
Shafienia H, Hoseini SM, Khalili MA, Heydari L, Sheikhha MH, Montazeri F. The possible regulatory role of miR-514 and miR-642b in cumulus cells on the oocyte maturation in patients with polycystic ovary syndrome. Reprod Biol 2024; 24:100863. [PMID: 38367330 DOI: 10.1016/j.repbio.2024.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/19/2024]
Abstract
Polycystic ovary syndrome is a common endocrine disorder in reproductive-age women. Accordingly, abnormal microenvironment may negatively influence oocyte developmental competence as a result of the altered expression profile of cumulus cells (CCs), mainly the key players of oocyte maturation, such as epidermal growth factor receptor (EGFR) and prostaglandin E receptor-2 (PTGER2). This study aimed to examine the expression levels of miR-514, miR-642b, and their candidate target genes (EGFR and PTGER2, respectively) in CCs of immature and mature oocytes in patients with PCOS. A total of 40 oocytes at germinal vesicle (GV) and 40 oocytes at metaphase II (MII) stages were retrieved from 30 PCOS women. Quantitative real-time PCR was performed to analyze the expression level of miR-514, miR-642b, EGFR, and PTGER2 in cumulus cells (CCS) of each oocyte. The expression level of miRNAs and their candidate target genes were compared between CCs of GV and MII oocytes. Our study suggests an inverse relationship exists between the expression levels of miR-514 and EGFR, and miR-642b and PTGER2. Furthermore, we observed that CCs of GV oocytes had higher levels of EGFR and PTGER2 mRNA and lower levels of miR-514 and miR-642b expression compared to those of MII oocytes. The present study demonstrated that miR-514 and miR-642b can regulate oocyte development by targeting EGFR and PTGER2, respectively. Therefore, examination of these miRNAs in CCs could be promising parameters to predict oocyte competence in PCOS patients.
Collapse
Affiliation(s)
- Hanieh Shafienia
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Seyed Mehdi Hoseini
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Ali Khalili
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Leila Heydari
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Hasan Sheikhha
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran.
| | - Fateme Montazeri
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran.
| |
Collapse
|
9
|
Hu Y, Zhang R, Zhang S, Ji Y, Zhou Q, Leng L, Meng F, Gong F, Lu G, Lin G, Hu L. Transcriptomic profiles reveal the characteristics of oocytes and cumulus cells at GV, MI, and MII in follicles before ovulation. J Ovarian Res 2023; 16:225. [PMID: 37993893 PMCID: PMC10664256 DOI: 10.1186/s13048-023-01291-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/01/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND The oocyte and its surrounding cumulus cells (CCs) exist as an inseparable entity. The maturation of the oocyte relies on communication between the oocyte and the surrounding CCs. However, oocyte evaluation is primarily based on morphological parameters currently, which offer limited insight into the quality and competence of the oocyte. Here, we conducted transcriptomic profiling of oocytes and their CCs from 47 patients undergoing preimplantation genetic testing for aneuploidy (PGT-A). We aimed to investigate the molecular events occurring between oocytes and CCs at different stages of oocyte maturation (germinal vesicle [GV], metaphase I [MI], and metaphase II [MII]). Our goal is to provide new insights into in vitro oocyte maturation (IVM). RESULTS Our findings indicate that oocyte maturation is a complex and dynamic process and that MI oocytes can be further classified into two distinct subtypes: GV-like-MI oocytes and MII-like-MI oocytes. Human oocytes and cumulus cells at three different stages of maturation were analyzed using RNA-seq, which revealed unique transcriptional machinery, stage-specific genes and pathways, and transcription factor networks that displayed developmental stage-specific expression patterns. We have also identified that both lipid and cholesterol metabolism in cumulus cells is active during the late stage of oocyte maturation. Lipids may serve as a more efficient energy source for oocytes and even embryogenesis. CONCLUSIONS Overall, our study provides a relatively comprehensive overview of the transcriptional characteristics and potential interactions between human oocytes and cumulus cells at various stages of maturation before ovulation. This study may offer novel perspectives on IVM and provide a reliable reference data set for understanding the transcriptional regulation of follicular maturation.
Collapse
Affiliation(s)
- Yena Hu
- Institute of Reproductive and Stems Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, China
| | - Ran Zhang
- Institute of Reproductive and Stems Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, China
| | - Shuoping Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410013, Hunan, China
| | - Yaxing Ji
- Institute of Reproductive and Stems Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, China
| | - Qinwei Zhou
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410013, Hunan, China
| | - Lizhi Leng
- Institute of Reproductive and Stems Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410013, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, Changsha, 410013, Hunan, China
| | - Fei Meng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410013, Hunan, China
| | - Fei Gong
- Institute of Reproductive and Stems Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410013, Hunan, China
| | - Guangxiu Lu
- Institute of Reproductive and Stems Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410013, Hunan, China
- National Engineering and Research Center of Human Stem Cells, Changsha, 410013, Hunan, China
| | - Ge Lin
- Institute of Reproductive and Stems Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, China.
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410013, Hunan, China.
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, Changsha, 410013, Hunan, China.
- National Engineering and Research Center of Human Stem Cells, Changsha, 410013, Hunan, China.
| | - Liang Hu
- Institute of Reproductive and Stems Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, China.
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, 410013, Hunan, China.
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, Changsha, 410013, Hunan, China.
- Hunan Normal University School of Medicine, ChangshaHunan, 410013, China.
| |
Collapse
|
10
|
Maugrion E, Shedova EN, Uzbekov R, Teixeira-Gomes AP, Labas V, Tomas D, Banliat C, Singina GN, Uzbekova S. Extracellular Vesicles Contribute to the Difference in Lipid Composition between Ovarian Follicles of Different Size Revealed by Mass Spectrometry Imaging. Metabolites 2023; 13:1001. [PMID: 37755281 PMCID: PMC10538054 DOI: 10.3390/metabo13091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Follicular fluid (FF) ensures a safe environment for oocyte growth and maturation inside the ovarian follicle in mammals. In each cycle, the large dominant follicle (LF) contains the oocyte designated to be ovulated, whereas the small subordinate follicles (SFs) of the same wave will die through atresia. In cows, the oocytes from the SF, being 2 mm in size, are suitable for in vitro reproduction biotechnologies, and their competence in developing an embryo depends on the size of the follicles. FF contains proteins, metabolites, fatty acids, and a multitude of extracellular vesicles (ffEVs) of different origins, which may influence oocyte competence through bidirectional exchanges of specific molecular cargo between follicular cells and enclosed oocytes. FF composition evolves along with follicle growth, and the abundance of different lipids varies between the LF and SF. Here, significant differences in FF lipid content between the LFs and SFs within the same ovary were demonstrated by MALD-TOF mass spectrometry imaging on bovine ovarian sections. We then aimed to enlighten the lipid composition of FF, and MALDI-TOF lipid profiling was performed on cellular, vesicular, and liquid fractions of FF. Differential analyses on the abundance of detected lipid features revealed specific enrichment of phospholipids in different ffEV types, such as microvesicles (MVs) and exosomes (Exo), compared to depleted FF. MALDI-TOF lipid profiling on MVs and Exo from the LF and SF samples (n = 24) revealed that more than 40% of detected features were differentially abundant between the groups of MVs and Exo from the different follicles (p < 0.01, fold change > 2). Glycerophospholipid and sphingolipid features were more abundant in ffEVs from the SFs, whereas different lysophospholipids, including phosphatidylinositols, were more abundant in the LFs. As determined by functional analysis, the specific lipid composition of ffEVs suggested the involvement of vesicular lipids in cell signaling pathways and largely contributed to the differentiation of the dominant and subordinate follicles.
Collapse
Affiliation(s)
- Emilie Maugrion
- CNRS, INRAE, University of Tours, PRC, 37380 Nouzilly, France (A.-P.T.-G.); (V.L.); (D.T.)
- PIXANIM, INRAE, University of Tours, CHU of Tours, 37380 Nouzilly, France
| | | | - Rustem Uzbekov
- Laboratory of Cell Biology and Electron Microscopy, Medical Faculty, University of Tours, 37032 Tours, France
- Faculty of Bioengineering and Bioinformatics, Moscow State University, 119992 Moscow, Russia
| | - Ana-Paula Teixeira-Gomes
- CNRS, INRAE, University of Tours, PRC, 37380 Nouzilly, France (A.-P.T.-G.); (V.L.); (D.T.)
- PIXANIM, INRAE, University of Tours, CHU of Tours, 37380 Nouzilly, France
| | - Valerie Labas
- CNRS, INRAE, University of Tours, PRC, 37380 Nouzilly, France (A.-P.T.-G.); (V.L.); (D.T.)
- PIXANIM, INRAE, University of Tours, CHU of Tours, 37380 Nouzilly, France
| | - Daniel Tomas
- CNRS, INRAE, University of Tours, PRC, 37380 Nouzilly, France (A.-P.T.-G.); (V.L.); (D.T.)
- PIXANIM, INRAE, University of Tours, CHU of Tours, 37380 Nouzilly, France
| | - Charles Banliat
- PIXANIM, INRAE, University of Tours, CHU of Tours, 37380 Nouzilly, France
- Ecole Supérieure d’Agricultures (ESA), 49007 Angers, France
| | - Galina N. Singina
- L.K. Ernst Federal Research Center for Animal Husbandry, 142132 Podolsk, Russia
| | - Svetlana Uzbekova
- CNRS, INRAE, University of Tours, PRC, 37380 Nouzilly, France (A.-P.T.-G.); (V.L.); (D.T.)
| |
Collapse
|
11
|
Kussano NR, Leme LDO, Dode MAN. Protein source in maturation media affects gene expression in cumulus cells and embryo development in cattle. Anim Biotechnol 2021:1-14. [PMID: 34964703 DOI: 10.1080/10495398.2021.2019755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
We aimed to evaluate if protein source (PS) alterations during IVM affect embryo sex/development and gene expression profile in cumulus cells (CCs). Bovine oocytes were matured and cultured in the presence of FBS or BSA. Then, the PS effect during IVM on gene expression (GPC4, VCAN, GHR, PTGS2, and ALCAM) was determined. CC biopsy was removed before and after IVM treatments. After fertilization and cultured, CCs were grouped according to their fate into CCs from immature COCs, CCs from COCs that did or did not result in embryos (according to PS). Results showed that when the culture was performed in FBS presence, blastocyst rate was higher (p < 0.05) than BSA. However, when embryos were cultured with BSA, no effect (p > 0.05) of PS during IVM was observed. PS used during IVM did not affect embryos sex (p > 0.05) but changed VCAN, GHR, PTGS2, and ALCAM genes expression. No differences (p > 0.05) were observed between immature and mature CCs groups in gene expression, regardless of their fate. Only the GHR gene was related to embryo production but just with FBS on IVM. In conclusion, PS can affect embryo development when using the serum on IVM and IVC, influences CCs gene expression, and has to be considered when studying oocyte quality markers.
Collapse
Affiliation(s)
| | | | - Margot Alves Nunes Dode
- Institute of Biology, University of Brasilia, Brasília, Brazil.,Laboratory of Animal Reproduction, Embrapa Genetic Resources and Biotechnology, Brasília, Brazil
| |
Collapse
|
12
|
Uzbekova S, Bertevello PS, Dalbies-Tran R, Elis S, Labas V, Monget P, Teixeira-Gomes AP. Metabolic exchanges between the oocyte and its environment: focus on lipids. Reprod Fertil Dev 2021; 34:1-26. [PMID: 35231385 DOI: 10.1071/rd21249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Finely regulated fatty acid (FA) metabolism within ovarian follicles is crucial to follicular development and influences the quality of the enclosed oocyte, which relies on the surrounding intra-follicular environment for its growth and maturation. A growing number of studies have examined the association between the lipid composition of follicular compartments and oocyte quality. In this review, we focus on lipids, their possible exchanges between compartments within the ovarian follicle and their involvement in different pathways during oocyte final growth and maturation. Lipidomics provides a detailed snapshot of the global lipid profiles and identified lipids, clearly discriminating the cells or fluid from follicles at distinct physiological stages. Follicular fluid appears as a main mediator of lipid exchanges between follicular somatic cells and the oocyte, through vesicle-mediated and non-vesicular transport of esterified and free FA. A variety of expression data allowed the identification of common and cell-type-specific actors of lipid metabolism in theca cells, granulosa cells, cumulus cells and oocytes, including key regulators of FA uptake, FA transport, lipid transformation, lipoprotein synthesis and protein palmitoylation. They act in harmony to accompany follicular development, and maintain intra-follicular homeostasis to allow the oocyte to accumulate energy and membrane lipids for subsequent meiotic divisions and first embryo cleavages.
Collapse
Affiliation(s)
- Svetlana Uzbekova
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France; and LK Ernst Federal Science Centre for Animal Husbandry, Podolsk, Russia
| | | | | | - Sebastien Elis
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France
| | - Valerie Labas
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France; and INRAE, Université de Tours, CHRU Tours, Plate-Forme PIXANIM, F-37380 Nouzilly, France
| | - Philippe Monget
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France
| | - Ana-Paula Teixeira-Gomes
- CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France; and INRAE, Université de Tours, CHRU Tours, Plate-Forme PIXANIM, F-37380 Nouzilly, France
| |
Collapse
|
13
|
Charpigny G, Marquant-Le Guienne B, Richard C, Adenot P, Dubois O, Gélin V, Peynot N, Daniel N, Brochard V, Nuttinck F. PGE2 Supplementation of Oocyte Culture Media Improves the Developmental and Cryotolerance Performance of Bovine Blastocysts Derived From a Serum-Free in vitro Production System, Mirroring the Inner Cell Mass Transcriptome. Front Cell Dev Biol 2021; 9:672948. [PMID: 34164396 PMCID: PMC8215579 DOI: 10.3389/fcell.2021.672948] [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: 02/26/2021] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
The culture media used throughout the in vitro production (IVP) of bovine embryos remain complex. The serum added to culture media in order to improve embryo development negatively impacts the cryotolerance of blastocysts. Periconceptional prostaglandin E2 (PGE2) signaling is known to exert prosurvival effects on in vitro-generated blastocysts. The purpose of the present study was to evaluate the effects on developmental and cryotolerance performance of a serum-free (SF) IVP system that included defined oocyte culture media supplemented or not with PGE2, versus serum-containing (SC) IVP. RNA-sequencing analysis was used to examine the gene expression of ICM derived under the different IVP conditions. We assessed the degree of cryotolerance of grade-I blastocysts during a three-day post-thaw culture by measuring survival and hatching rates, counting trophectoderm and inner cell mass (ICM) blastomere numbers. We also determined the proportion of ICM cells expressing octamer-binding transcription factor 4 protein (OCT4/POU5F1). We showed that grade-I blastocyst development rates under SF + PGE2 conditions were similar to those obtained under SC conditions, although the cleavage rate remained significantly lower. SC IVP conditions induced changes to ICM gene expression relative to several metabolic processes, catabolic activities, cell death and apoptosis. These alterations were associated with significantly higher levels of ICM cell death at day 7 post-fertilization, and lower survival and hatching rates after thawing. SF IVP conditions supplemented or not with PGE2 induced changes to ICM gene expression related to DNA replication, metabolism and double-strand break repair processes, and were associated with significantly larger ICM cell populations after thawing. SF + PGE2 IVP induced changes to ICM gene expression related to epigenetic regulation and were associated with a significantly higher proportion of ICM cells expressing OCT4. For the first time, our study thus offers a comprehensive analysis of the ICM transcriptome regulated by IVP culture conditions in terms of the cellular changes revealed during culture for three days after thawing.
Collapse
Affiliation(s)
- Gilles Charpigny
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | | | - Christophe Richard
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Pierre Adenot
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France.,INRAE, MIMA2, Université Paris-Saclay, Jouy-en-Josas, France
| | - Olivier Dubois
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Valérie Gélin
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Nathalie Peynot
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Nathalie Daniel
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Vincent Brochard
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Fabienne Nuttinck
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France.,Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| |
Collapse
|
14
|
Chen YC, Li JY, Li CJ, Tsui KH, Wang PH, Wen ZH, Lin LT. Luteal Phase Ovarian Stimulation versus Follicular Phase Ovarian Stimulation results in different Human Cumulus cell genes expression: A pilot study. Int J Med Sci 2021; 18:1600-1608. [PMID: 33746576 PMCID: PMC7976567 DOI: 10.7150/ijms.55955] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 01/21/2021] [Indexed: 12/22/2022] Open
Abstract
Background: Luteal-phase ovarian stimulation (LPOS) is an alternative in vitro fertilization (IVF) protocol. However, limited data showed the genes expression of cumulus cells (CCs) in LPOS. Therefore, this study aimed to investigate CC genes expression between LPOS and follicular-phase ovarian stimulation (FPOS) in poor ovarian responders (PORs) undergoing IVF cycles. Methods: This was a prospective non-randomized trial (ClinicalTrials.gov Identifier: NCT03238833). A total of 36 PORs who met the Bologna criteria and underwent IVF cycles were enrolled. Fifteen PORs were allocated to the LPOS group, and 21 PORs were allocated to the FPOS group. The levels of CC genes involved in inflammation (CXCL1, CXCL3, TNF, PTGES), oxidative phosphorylation (NDUFB7, NDUFA4L2, SLC25A27), apoptosis (DAPK3, BCL6B) and metabolism (PCK1, LDHC) were analyzed using real-time quantitative PCR and compared between the two groups. Results: The number of retrieved oocytes, metaphase II oocytes, fertilized oocytes, day-3 embryos and top-quality day-3 embryos, clinical pregnancy rates and live birth rates were similar between the two groups except for significantly high progesterone levels in the LPOS group. The mRNA expression levels of CXCL1 (0.51 vs 1.00, p < 0.001) and PTGES (0.30 vs 1.00, p < 0.01) were significantly lower in the LPOS group than in the FPOS group. The LPOS group had significantly lower mRNA expression of NDUFB7 (0.12 vs 1.00, p < 0.001) and NDUFA4L2 (0.33 vs 1.00, p < 0.01) than the FPOS group. DAPK3 (3.81 vs 1.00, p < 0.05) and BCL6B (2.59 vs 1.00, p < 0.01) mRNA expression was significantly higher in the LPOS group than in the FPOS group. Increased expression of PCK1 (3.13 vs. 1.00, p < 0.001) and decreased expression of LDHC (0.12 vs. 1.00, p < 0.001) were observed in the LPOS group compared to the FPOS group. Conclusions: Our data revealed different CC genes expression involving in inflammation, oxidative phosphorylation, apoptosis and metabolism between LPOS and FPOS in PORs. However, the results are non-conclusive; further large-scale randomized controlled trials are needed to validate the results.
Collapse
Affiliation(s)
- Yu-Chen Chen
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan
| | - Ju-Yueh Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan.,Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei City, Taiwan.,Institute of BioPharmaceutical Sciences, National Sun Yat‑sen University, Kaohsiung City, Taiwan
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei City, Taiwan.,Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei City, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung City, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Li-Te Lin
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan.,Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei City, Taiwan.,Institute of BioPharmaceutical Sciences, National Sun Yat‑sen University, Kaohsiung City, Taiwan
| |
Collapse
|
15
|
Wyse BA, Fuchs Weizman N, Kadish S, Balakier H, Sangaralingam M, Librach CL. Transcriptomics of cumulus cells - a window into oocyte maturation in humans. J Ovarian Res 2020; 13:93. [PMID: 32787963 PMCID: PMC7425158 DOI: 10.1186/s13048-020-00696-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cumulus cells (CC) encapsulate growing oocytes and support their growth and development. Transcriptomic signatures of CC have the potential to serve as valuable non-invasive biomarkers for oocyte competency and potential. The present sibling cumulus-oocyte-complex (COC) cohort study aimed at defining functional variations between oocytes of different maturity exposed to the same stimulation conditions, by assessing the transcriptomic signatures of their corresponding CC. CC were collected from 18 patients with both germinal vesicle and metaphase II oocytes from the same cycle to keep the biological variability between samples to a minimum. RNA sequencing, differential expression, pathway analysis, and leading-edge were performed to highlight functional differences between CC encapsulating oocytes of different maturity. RESULTS Transcriptomic signatures representing CC encapsulating oocytes of different maturity clustered separately on principal component analysis with 1818 genes differentially expressed. CCs encapsulating mature oocytes were more transcriptionally synchronized when compared with CCs encapsulating immature oocytes. Moreover, the transcriptional activity was lower, albeit not absent, in CC encapsulating mature oocytes, with 2407 fewer transcripts detected than in CC encapsulating immature (germinal vesicle - GV) oocytes. Hallmark pathways and ovarian processes that were affected by oocyte maturity included cell cycle regulation, steroid metabolism, apoptosis, extracellular matrix remodeling, and inflammation. CONCLUSIONS Herein we review our findings and discuss how they align with previous literature addressing transcriptomic signatures of oocyte maturation. Our findings support the available literature and enhance it with several genes and pathways, which have not been previously implicated in promoting human oocyte maturation. This study lays the ground for future functional studies that can enhance our understanding of human oocyte maturation.
Collapse
Affiliation(s)
- Brandon A Wyse
- CReATe Fertility Centre, 790 Bay St. Suite 420, Toronto, ON, M5G 1N8, Canada.
| | - Noga Fuchs Weizman
- CReATe Fertility Centre, 790 Bay St. Suite 420, Toronto, ON, M5G 1N8, Canada
| | - Seth Kadish
- CReATe Fertility Centre, 790 Bay St. Suite 420, Toronto, ON, M5G 1N8, Canada
| | - Hanna Balakier
- CReATe Fertility Centre, 790 Bay St. Suite 420, Toronto, ON, M5G 1N8, Canada
| | | | - Clifford L Librach
- CReATe Fertility Centre, 790 Bay St. Suite 420, Toronto, ON, M5G 1N8, Canada
- Department of Obstetrics and Gynecology; Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Physiology; Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Obstetrics and Gynecology, Women's College Hospital, Toronto, Canada
| |
Collapse
|
16
|
Rodrigues SAD, Pontelo TP, Kussano NR, Kawamoto TS, Leme LO, Caixeta FMC, Pfeifer LFM, Franco MM, Dode MAN. Effects of Prostaglandins E2 and F2α on the in vitro maturation of bovine oocytes. Domest Anim Endocrinol 2020; 72:106447. [PMID: 32403000 DOI: 10.1016/j.domaniend.2020.106447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 11/21/2022]
Abstract
We aimed to elucidate the effects of PGE2 and PGF2α on the in vitro maturation (IVM) of bovine oocytes. First, cumulus-oocyte complexes were matured in the media supplemented with or without PGE2, PGF2α, or PGE2 plus PGF2α for the final 24, 12, or 6 h of culture. Then, the cumulus-oocyte complexes were matured in the absence or presence of a PG endoperoxide synthase 2 (PTGS2) enzyme inhibitor (NS398) supplemented with PGE2, PGF2α, or PGE2 plus PGF2α. Finally, the expression of genes associated with PGs activity in cumulus cells (PTGS2, PG E-synthase-1 [PTGES1], and aldo-keto reductase 1 [AKR1B1]) or oocytes (receptors for PGE2 [PTGER2] and PGF2α [PTGFR]) of different competencies was quantified. Supplementation of the IVM medium with PGs did not improve in vitro embryo production or embryo quality (P > 0.05). During maturation, the relative abundance of PTGS2 transcripts increased (P < 0.05) only in the less-competent group, whereas those of PTGES1 increased in the less-competent and in the more-competent groups. Conversely, AKR1B1 expression decreased only in the less-competent group (P < 0.05). Receptors for the PGE2 and PGF2α genes were very low or undetectable in oocytes. In conclusion, PGE2 and PGF2α are not recommended for media supplementation during maturation because they have no effect on embryo development. Although genes related to PGs activity are differentially expressed in cumulus cells of cumulus-oocyte complexes of different competence during maturation, the expression of PGE2 and PGF2α receptor genes was either not detectable or was detected at low levels in oocytes.
Collapse
Affiliation(s)
| | - Thais P Pontelo
- Federal University of Lavras, Veterinary Science, Lavras, MG 32700-000, Brazil
| | - Nayara R Kussano
- University of Brasília, Animal Biology, Brasilia, DF 70910-900, Brazil
| | - Taynan S Kawamoto
- Federal University Uberlândia, Animal Science, Uberlândia, MG 38400-902, Brazil
| | - Ligiane O Leme
- Federal University of Espírito Santo, Animal Science, Vitória, ES 29075-073, Brazil
| | | | | | - Mauricio M Franco
- Federal University Uberlândia, Animal Science, Uberlândia, MG 38400-902, Brazil; Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil
| | - Margot A N Dode
- University of Brasília, Animal Science, Brasilia, DF 70910-900, Brazil; Embrapa Genetic Resources and Biotechnology, Brasília, DF 70770-917, Brazil.
| |
Collapse
|
17
|
Argudo DE, Tenemaza MA, Merchán SL, Balvoa JA, Méndez MS, Soria ME, Galarza LR, Ayala LE, Hernández-Fonseca HJ, Perea MS, Perea FP. Intraovarian influence of bovine corpus luteum on oocyte morphometry and developmental competence, embryo production and cryotolerance. Theriogenology 2020; 155:232-239. [PMID: 32758994 DOI: 10.1016/j.theriogenology.2020.05.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 04/28/2020] [Accepted: 05/30/2020] [Indexed: 12/25/2022]
Abstract
Three experiments were conducted to determine influence of the bovine corpus luteum (CL) on morphometric and functional characteristics of oocytes, and subsequent embryonic development. Cumulus-oocyte complexes were aspirated from two types of cows: 1) with a CL in one ovary (CL+) and without a CL in the contralateral ovary (CL-), 2) and from cows without CL in either ovary (C). Intracellular activity of the enzyme glucose-6-phosphate dehydrogenase (G6PDH), oocyte diameter and thickness of the zona pellucida were determined (Experiment 1). Then, the rate of in vitro oocyte maturation for each ovarian category was evaluated and oocyte diameter and zona pellucida thickness were measured after maturation (Experiment 2). In Experiment 3, in vitro embryo production and cryotolerance were assessed. The oocyte diameter was greater (P < 0.01) and the zona pellucida was thinner in CL+ than in CL- (P > 0.05) or C (P = 0.0131) ovaries. Activity of G6PDH was lower in oocytes from CL+ than CL- (P < 0.01) and C (P = 0.0148) ovaries. Rate of oocyte maturation, oocyte diameter and thickness of the zona pellucida after maturation did not differ among groups. Rate of cleavage was greater in zygotes from CL+ than from CL- or C (P < 0.01); and CL+ ovaries produced more total embryos on day 7 (P < 0.05) and more blastocysts (P < 0.01) than CL- and C ovaries. Rate of expansion and hatching of day-7 vitrified-warmed blastocysts at 24 and 48 h of culture did not differ among groups. In conclusion, oocytes collected from CL+ ovaries were larger and metabolically more prepared to continue maturation than those from ovaries lacking a CL. Also, rates of cleavage and yield of blastocysts were greater for oocytes from CL+ ovaries than from CL- and C ovaries. These findings indicate that a CL influenced oocyte developmental competence and embryonic development, presumably through intraovarian interactions.
Collapse
Affiliation(s)
- Daniel E Argudo
- Unidad Académica de Ciencias Agropecuarias, Universidad Católica de Cuenca, Ecuador
| | - Milton A Tenemaza
- Laboratorio de Biotecnologías de la Reproducción Animal, Facultad de Ciencias Agropecuarias. Universidad de Cuenca, Ecuador
| | - Shirley L Merchán
- Laboratorio de Biotecnologías de la Reproducción Animal, Facultad de Ciencias Agropecuarias. Universidad de Cuenca, Ecuador
| | - José A Balvoa
- Laboratorio de Biotecnologías de la Reproducción Animal, Facultad de Ciencias Agropecuarias. Universidad de Cuenca, Ecuador
| | - Maria S Méndez
- Laboratorio de Biotecnologías de la Reproducción Animal, Facultad de Ciencias Agropecuarias. Universidad de Cuenca, Ecuador
| | - Manuel E Soria
- Laboratorio de Biotecnologías de la Reproducción Animal, Facultad de Ciencias Agropecuarias. Universidad de Cuenca, Ecuador
| | - Luis R Galarza
- Laboratorio de Biotecnologías de la Reproducción Animal, Facultad de Ciencias Agropecuarias. Universidad de Cuenca, Ecuador
| | - Luis E Ayala
- Laboratorio de Biotecnologías de la Reproducción Animal, Facultad de Ciencias Agropecuarias. Universidad de Cuenca, Ecuador
| | | | - Mariana S Perea
- Facultad de Ciencias Veterinarias, Universidad del Zulia, Venezuela
| | - Fernando P Perea
- Departamento de Ciencias Agrarias, Universidad de Los Andes, Trujillo, Venezuela.
| |
Collapse
|
18
|
Boruszewska D, Kowalczyk-Zieba I, Suwik K, Staszkiewicz-Chodor J, Jaworska J, Lukaszuk K, Woclawek-Potocka I. Prostaglandin E 2 affects in vitro maturation of bovine oocytes. Reprod Biol Endocrinol 2020; 18:40. [PMID: 32393337 PMCID: PMC7216604 DOI: 10.1186/s12958-020-00598-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/22/2020] [Indexed: 12/04/2022] Open
Abstract
The role of prostaglandin E2 (PGE2) in the successful resumption of oocyte meiosis and cumulus expansion has been well-documented. However, there remains very little information available on the influence of PGE2 on other processes that occur during oocyte maturation. In this study, we supplemented a maturation medium with PGE2 and monitored oocyte quality markers, glucose metabolism, mitochondrial status, oxidative stress, and apoptosis in the cumulus-oocyte complexes (COCs), using a well-established in vitro model of embryo production in cattle. We found that this increased availability of PGE2 during maturation led to an increase in the expression of genes associated with oocyte competence and improved the quality of blastocysts produced. Prostaglandin E2 also appeared to stimulate glucose uptake and lactate production in the COCs, both influencing the expression of enzymes involved in glycolysis and the hexosamine biosynthetic pathway. We found that PGE2 reduced intracellular reactive oxygen species levels, and simultaneously increased glutathione concentration and stimulated antioxidant gene expression in the oocyte. These results indicate that PGE2 has an important role in the protection of oocytes against oxidative stress. Mitochondrial membrane potential was also improved in PGE2-treated oocytes, and there was a reduction in the occurrence of apoptosis in the COCs. Promotion of an anti-apoptotic balance in transcription of genes involved in apoptosis was present in both oocytes and the cumulus cells. In summary, PGE2 could represent a novel autocrine/paracrine player in the mechanisms that can facilitate successful oocyte maturation and oocyte survival in the cow.
Collapse
Affiliation(s)
- Dorota Boruszewska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10 Str., 10-748, Olsztyn, Poland.
| | - Ilona Kowalczyk-Zieba
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10 Str., 10-748, Olsztyn, Poland
| | - Katarzyna Suwik
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10 Str., 10-748, Olsztyn, Poland
| | - Joanna Staszkiewicz-Chodor
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10 Str., 10-748, Olsztyn, Poland
| | - Joanna Jaworska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10 Str., 10-748, Olsztyn, Poland
| | - Krzysztof Lukaszuk
- Department of Obstetrics and Gynecological Nursing, Faculty of Health Sciences, Medical University of Gdansk, M. Skłodowskiej-Curie 3a Str., 80-210, Gdansk, Poland
- INVICTA Fertility and Reproductive Center, Rajska 10 Str., 80-850, Gdansk, Poland
| | - Izabela Woclawek-Potocka
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10 Str., 10-748, Olsztyn, Poland
| |
Collapse
|
19
|
Granulosa secreted factors improve the developmental competence of cumulus oocyte complexes from small antral follicles in sheep. PLoS One 2020; 15:e0229043. [PMID: 32182244 PMCID: PMC7077809 DOI: 10.1371/journal.pone.0229043] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
Oocyte in vitro maturation can be improved by mimicking the intra-follicular environment. Oocyte, cumulus cells, granulosa cells, and circulating factors act as meiotic regulators in follicles and maintain oocyte in the meiotic phase until oocyte becomes competent and ready to be ovulated. In a randomized experimental design, an ovine model was used to optimize the standard in vitro maturation media by Granulosa secreted factors. At first, the development capacity of oocyte derived from medium (>4 to 6 mm) and small (2 to ≤4 mm) size follicles was determined. Differential gene expression of granulosa secreted factors and their receptors were compared between the cumulus cells of the two groups. Then, the best time and concentration for arresting oocytes at the germinal vesicle stage by natriuretic peptide type C (CNP) were determined by nuclear staining in both groups. Oocyte quality was further confirmed by calcein uptake and gene expression. The developmental competence of cumulus oocyte complexes derived from small size follicles that were cultured in the presence of CNP in combination with amphiregulin (AREG) and prostaglandin E2 (PGE2) for 24 h was determined. Finally, embryo quality was specified by assessing expressions of NANOG, SOX2, CDX2, OCT4, and TET1. The cumulus oocyte complexes derived from small size follicles had a lower capacity to form blastocyst in comparison with cumulus oocyte complexes derived from medium size follicles. Prostaglandin E receptor 2 and prostaglandin-endoperoxide synthase 2 had significantly lower expression in cumulus cells derived from small size follicles in comparison with cumulus cells derived from medium size follicles. Natriuretic peptide type C increased the percentage of cumulus oocyte complexes arresting at the germinal vesicle stage in both oocytes derived from medium and small follicles. Gap junction communication was also improved in the presence of natriuretic peptide type C. In oocytes derived from small size follicles; best blastocyst rates were achieved by sequential exposure of cumulus oocyte complexes in [TCM+CNP (6 h), then cultured in TCM+AREG+PGE2 (18h)] and [TCM+CNP (6 h), then cultured in conventional IVM supplements+AREG+PGE2 (18h)]. Increased SOX2 expression was observed in [TCM+CNP (6 h), then cultured in TCM+AREG+PGE2 (18h)], while decreased OCT4 expression was observed in [TCM+CNP (6 h), then cultured in conventional IVM supplements+AREG+PGE2 (18h)]. It seems that the natriuretic peptide type C modulates meiotic progression, and oocyte development is probably mediated by amphiregulin and prostaglandin E2. These results may provide an alternative IVM method to optimize in vitro embryo production in sheep and subsequently for humans.
Collapse
|
20
|
Piersanti RL, Santos JEP, Sheldon IM, Bromfield JJ. Lipopolysaccharide and tumor necrosis factor-alpha alter gene expression of oocytes and cumulus cells during bovine in vitro maturation. Mol Reprod Dev 2019; 86:1909-1920. [PMID: 31663199 DOI: 10.1002/mrd.23288] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/08/2019] [Indexed: 12/23/2022]
Abstract
Communication between the oocyte and cumulus facilitates oocyte growth, cell cycle regulation, and metabolism. This communication is mediated by direct contact between oocytes and cumulus cells, and soluble secreted molecules. Secreted molecules involved in this process are known inflammatory mediators. Lipopolysaccharide (LPS) is detected in follicular fluid and is associated with reduced fertility, whereas accumulation of inflammatory mediators in follicular fluid, including tumor necrosis factor-α (TNF-α), is associated with female infertility. Maturation of oocytes in the presence of LPS or TNF-α reduces meiotic maturation and the capacity to develop to the blastocyst. Here we evaluated the abundance of 92 candidate genes involved immune function, epigenetic modifications, embryo development, oocyte secreted factors, apoptosis, cell cycle, and cell signaling in bovine cumulus cells or zona-free oocytes after exposure to LPS or TNF-α during in vitro maturation. We hypothesize that LPS or TNF-α will alter the abundance of transcripts in oocytes and cumulus cell in a cell type dependent manner. Exposure to LPS altered abundance of 31 transcripts in oocytes (including ACVR1V, BMP15, DNMT3A) and 12 transcripts in cumulus cells (including AREG, FGF4, PIK3IP1). Exposure to TNF-α altered 1 transcript in oocytes (IGF2) and 4 transcripts in cumulus cells (GJA1, PLD2, PTGER4, STAT1). Cumulus expansion was reduced after exposure to LPS or TNF-α. Exposing COCs to LPS had a marked effect on expression of targeted transcripts in oocytes. We propose that altered oocyte transcript abundance is associated with reduced meiotic maturation and embryo development observed in oocytes cultured in LPS or TNF-α.
Collapse
Affiliation(s)
- Rachel L Piersanti
- Department of Animal Sciences, University of Florida, Gainesville, Florida
| | - José E P Santos
- Department of Animal Sciences, University of Florida, Gainesville, Florida
| | - I Martin Sheldon
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - John J Bromfield
- Department of Animal Sciences, University of Florida, Gainesville, Florida
| |
Collapse
|
21
|
Tetsuka M, Tanakadate M. Activation of HSD11B1 in the bovine cumulus-oocyte complex during IVM and IVF. Endocr Connect 2019; 8:1029-1039. [PMID: 31252401 PMCID: PMC6652248 DOI: 10.1530/ec-19-0188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 06/26/2019] [Indexed: 02/02/2023]
Abstract
The bovine cumulus-oocyte complex (COC) is capable of converting cortisone, an inert glucocorticoid to active cortisol. This mechanism is mediated by 11β-hydroxysteroid oxidoreductase type 1 (HSD11B1), whose expression dramatically increases in the mature COC. In this study, we investigate the time course expression of HSD11B1 and the enzyme activity in the bovine COC undergoing maturation and fertilization in relation to key events taking place in the COC. Bovine COCs were subjected to in vitro maturation (IVM) and fertilization (IVF). The activities of HSD11B1 and HSD11B2, which mediates the opposite reaction, were measured using a radiometric conversion assay. In parallel studies, cumulus expansion, P4 production and the expression of genes associated with ovulation were measured. The reductive activity of HSD11B1 increased in the latter half of IVM and remained high during IVF, whereas the oxidative activity of HSD11B2 remained unchanged over both periods. Consequently, the net glucocorticoid metabolism in the bovine COC shifted from inactivation to activation around the time of ovulation and fertilization. The increase in HSD11B1 expression lagged behind that of P4 increase and cumulus expansion but ahead of the expressions of genes responsible for PGE2 synthesis. The reductive activity of HSD11B1 was well correlated with the cumulus expansion rate. This outcome indicates that the ability of the cumulus to activate glucocorticoids is related to its ability to synthesize hyaluronan. These results also indicate that the activation of HSD11B1 is an integral part of the sequential events taking place at the ovulation and fertilization in the bovine COC.
Collapse
Affiliation(s)
- Masafumi Tetsuka
- Department of Life and Food Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- Correspondence should be addressed to M Tetsuka:
| | - Misato Tanakadate
- Department of Life and Food Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| |
Collapse
|
22
|
Boruszewska D, Grycmacher K, Kowalczyk-Zieba I, Sinderewicz E, Staszkiewicz-Chodor J, Woclawek-Potocka I. Expression of enzymes involved in the synthesis of prostaglandin E 2 in early- and late-cleaved bovine embryos at different stages of preimplantation development. Theriogenology 2019; 133:45-55. [PMID: 31059928 DOI: 10.1016/j.theriogenology.2019.04.032] [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] [Received: 01/09/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022]
Abstract
Prostaglandin (PG) E2 plays a role in numerous aspects of mammalian reproduction, such as oviductal transport of gametes, hatching from the zona pellucida in blastocysts and early embryonic development. Despite the evident role of PGE2 in the regulation of female reproductive processes, in the literature, there is very little information concerning the expression of PGE2 synthesizing enzymes and the exact amount of PGE2 produced by bovine embryos in vitro. In the present study, we aimed to determine the mRNA levels and immunolocalization of the enzymes responsible for PGE2 synthesis (PTGS2, mPGES1, mPGES2 and cPGES) in embryos at the 2-cell, 4-cell, 8-cell, 16-cell, morula, early blastocyst, blastocyst, expanded blastocyst and hatched blastocyst stages, using a well-defined bovine model of oocyte developmental competence based on the time of first cleavage. PTGS2, mPGES2 and cPGES transcripts and proteins were detected in all stages of embryos, whereas the mPGES1 transcript and protein were not detected in embryos from the 2- to 16-cell stage. The results showed different transcription profiles of the enzymes involved in PGE2 synthesis in early- and late-cleaved embryos during the early stages of their in vitro preimplantation development. We also found that all the analysed stages of bovine preimplantation embryos released PGE2, with the highest concentration on Day 7 of culture in both the early- and late-cleaved groups. The present study is the first to demonstrate PGE2 synthesis and production by bovine early- and late-cleaved embryos at different stages of preimplantation development. Bovine embryos can produce PGE2, which may exert paracrine regulation during development. The transcription levels of PGE2 synthases were affected by the embryonic stage of development and quality. Our results indicate that the different transcription profiles of PTGS2, mPGES1, mPGES2 and cPGES, as well as PGE2 concentration, in early-versus late-cleaved embryos are dependent on the quality of the oocytes from which the embryos were obtained, which could reveal the association of PGE2 production during bovine preimplantation development with more advanced stages of embryo development.
Collapse
Affiliation(s)
- Dorota Boruszewska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| | - Katarzyna Grycmacher
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| | - Ilona Kowalczyk-Zieba
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| | - Emilia Sinderewicz
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| | - Joanna Staszkiewicz-Chodor
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| | - Izabela Woclawek-Potocka
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
23
|
Ge ZW, Zhu XL, Wang BC, Hu JL, Sun JJ, Wang S, Chen XJ, Meng SP, Liu L, Cheng ZY. MicroRNA-26b relieves inflammatory response and myocardial remodeling of mice with myocardial infarction by suppression of MAPK pathway through binding to PTGS2. Int J Cardiol 2019; 280:152-159. [PMID: 30679074 DOI: 10.1016/j.ijcard.2018.12.077] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/19/2018] [Accepted: 12/27/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Myocardial infarction (MI) is a common cardiovascular disease caused by myocardial ischemia. Also, microRNA (miRNA) participates in the pathophysiology of many cardiovascular diseases, which can affect stem cell transplantation in the treatment of MI. In this study, our aim is to explore effect of miR-26b on inflammatory response and myocardial remodeling through the MAPK pathway by targeting PTGS2 in mice with MI. METHODS Microarray data analysis was conducted to screen MI-related differentially expressed gens (DEGs). Relationship between miR-26b and PTGS2 was testified. Cardiac function, inflammatory reaction, infarct size, and myocardial fibrosis were observed. The miR-26b expression and mRNA and protein levels of, PTGS2, ERK, JNK and p38 and Bcl-2/Bax were examined. The effect of miR-26b on cell apoptosis was also analyzed. RESULTS MiR-26b was predicted to target PTGS2 further to mediate the MAPK pathway, thus affecting MI. MiR-26b negatively targeted PTGS2. MI mice showed decreased cardiac function, as well as increased inflammatory reaction, myocardial injury, area of fibrosis and myocardial cell apoptosis. After injection of miR-26b agomir or NS-398 (PTGS2 inhibitor), inflammatory response of MI mice was attenuated and myocardial remodeling induced by MI was alleviated. CONCLUSION These findings indicate that miR-26b inhibits PTGS2 to activate the MAPK pathway, so as to reduce inflammatory response and improve myocardial remodeling in mice with MI.
Collapse
Affiliation(s)
- Zhen-Wei Ge
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Xi-Liang Zhu
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Bao-Cai Wang
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Jun-Long Hu
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Jun-Jie Sun
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Sheng Wang
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Xian-Jie Chen
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Shu-Ping Meng
- ICU of Cardiovascular Surgery, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Lin Liu
- Department of Cardiovascular Ultrasound, Henan Provincial People's Hospital, Zhengzhou 450003, PR China
| | - Zhao-Yun Cheng
- Department of Cardiovascular Surgery, Henan Provincial People's Hospital, Zhengzhou 450003, PR China.
| |
Collapse
|
24
|
Lipid Identification and Transcriptional Analysis of Controlling Enzymes in Bovine Ovarian Follicle. Int J Mol Sci 2018; 19:ijms19103261. [PMID: 30347829 PMCID: PMC6214003 DOI: 10.3390/ijms19103261] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 10/16/2018] [Accepted: 10/18/2018] [Indexed: 12/17/2022] Open
Abstract
Ovarian follicle provides a favorable environment for enclosed oocytes, which acquire their competence in supporting embryo development in tight communications with somatic follicular cells and follicular fluid (FF). Although steroidogenesis in theca (TH) and granulosa cells (GC) is largely studied, and the molecular mechanisms of fatty acid (FA) metabolism in cumulus cells (CC) and oocytes are emerging, little data is available regarding lipid metabolism regulation within ovarian follicles. In this study, we investigated lipid composition and the transcriptional regulation of FA metabolism in 3–8 mm ovarian follicles in bovine. Using liquid chromatography and mass spectrometry (MS), 438 and 439 lipids were identified in FF and follicular cells, respectively. From the MALDI-TOF MS lipid fingerprints of FF, TH, GC, CC, and oocytes, and the MS imaging of ovarian sections, we identified 197 peaks and determined more abundant lipids in each compartment. Transcriptomics revealed lipid metabolism-related genes, which were expressed constitutively or more specifically in TH, GC, CC, or oocytes. Coupled with differential lipid composition, these data suggest that the ovarian follicle contains the metabolic machinery that is potentially capable of metabolizing FA from nutrient uptake, degrading and producing lipoproteins, performing de novo lipogenesis, and accumulating lipid reserves, thus assuring oocyte energy supply, membrane synthesis, and lipid-mediated signaling to maintain follicular homeostasis.
Collapse
|
25
|
Gamarra G, Ponsart C, Lacaze S, Nuttinck F, Cordova A, Mermillod P, Marquant-Le Guienne B, Monniaux D, Humblot P, Ponter AA. Oral propylene glycol modifies follicular fluid and gene expression profiles in cumulus-oocyte complexes and embryos in feed-restricted heifers. Reprod Fertil Dev 2018; 30:417-429. [PMID: 28822459 DOI: 10.1071/rd17037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 07/03/2017] [Indexed: 01/30/2023] Open
Abstract
Dietary supplementation with propylene glycol (PG) increases in vitro production of high-quality embryos in feed-restricted heifers. The aim of the present study was to evaluate the effects of PG in feed-restricted heifers on follicular fluid insulin and insulin-like growth factor (IGF) 1 concentrations, expression of IGF system genes in oocytes and cumulus cells and the expression of selected genes in blastocysts. Feed-restricted (R) heifers were drenched with water or PG during induced oestrous cycles (400mL of PG or water/drench, daily drenching at 1600 hours for the first 9 days of the oestrous cycle). Ovum pick-up (OPU) was performed after superovulation to produce in vitro embryos and without superovulation to recover oocytes, cumulus cells and follicular fluid. OPU was also performed in a control group (not feed restricted and no drenching). Follicular fluid IGF1 concentrations were reduced by R, and PG restored IGF1 concentrations to those seen in the control group. In cumulus cells, expression of IGF1, IGF1 receptor (IGF1R) and IGF binding protein 4 (IGFBP4) was decreased in the R group, and fully (IGF1 and IGF1R) or partially (IGFBP4) restored to control levels by PG. Blastocyst perilipin 2 (PLIN2; also known as adipophilin), Bcl-2-associated X protein (BAX), SCL2A1 (facilitated glucose/fructose transporter GLUT1), aquaporin 3 (AQP3), DNA (cytosine-5)-methyltransferase 3A (DNMT3A) and heat shock 70-kDa protein 9 (HSPA9B) expression were decreased in R heifers; PG restored the expression of the last four genes to control levels. In conclusion, these results suggest that, during follicular growth, PG exerts epigenetic regulatory effects on gene expression in blastocyst stage embryos.
Collapse
Affiliation(s)
- G Gamarra
- ALLICE, Département Recherche et Développement, 78350 Jouy en Josas, France
| | - C Ponsart
- ANSES, Animal Health Laboratory, 94706 Maisons-Alfort, France
| | | | - F Nuttinck
- INRA, UMR 1198 Biologie du Développement et Reproduction, 78350 Jouy-en-Josas, France
| | - A Cordova
- INRA, UMR 7247 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
| | - P Mermillod
- INRA, UMR 7247 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
| | | | - D Monniaux
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
| | - P Humblot
- Division of Reproduction, Department of Clinical Sciences, Faculty of Veterinary Medicine and Agricultural Sciences, SLU, PO Box 7054, SE 75007, Uppsala, Sweden
| | - A A Ponter
- INRA, UMR 1198 Biologie du Développement et Reproduction, 78350 Jouy-en-Josas, France
| |
Collapse
|
26
|
Nuttinck F. Oocyte related factors impacting on embryo quality: relevance for in vitro embryo production. Anim Reprod 2018; 15:271-277. [PMID: 34178150 PMCID: PMC8202467 DOI: 10.21451/1984-3143-ar2018-0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The outcome of pregnancy is closely linked to early events that occur during the onset of embryogenesis.
The first stages in embryonic development are mainly governed by the storage of maternal factors
present in the oocyte at the time of fertilisation. In this review, we outline the different
classes of oocyte transcripts that may be involved in activation of the embryonic genome as
well as those associated with epigenetic reprogramming, imprinting maintenance or the control
of transposon mobilisation during preimplantation development. We also report the influence
of cumulus-oocyte crosstalk during the maturation process on the oocyte transcriptome and
how in vitro procedures can affect these interactions.
Collapse
|
27
|
Niringiyumukiza JD, Cai H, Xiang W. Prostaglandin E2 involvement in mammalian female fertility: ovulation, fertilization, embryo development and early implantation. Reprod Biol Endocrinol 2018; 16:43. [PMID: 29716588 PMCID: PMC5928575 DOI: 10.1186/s12958-018-0359-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/20/2018] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Infertility in mammalian females has been a challenge in reproductive medicine. The causes of female infertility include anovulation, ovulated oocyte defects, abnormal fertilization, and insufficient luteal support for embryo development, as well as early implantation. Ovulation induction, in vitro fertilization and luteal support regimens have been performed for decades to increase fertility rates. The identification of proteins and biochemical factors involved in female reproduction is essential to further increase female fertility rates. Evidence has shown that prostaglandins (PGs) might be involved in the female reproductive process, mainly ovulation, fertilization, and implantation. However, only a few studies on individual PGs in female reproduction have been done so far. This review aimed to identify the pivotal role of prostaglandin E2 (PGE2), a predominant PG, in female reproduction to improve fertility, specifically ovulation, fertilization, embryo development and early implantation. RESULTS Prostaglandin E2 (PGE2) was shown to play a relevant role in the ovulatory cascade, including meiotic maturation, cumulus expansion and follicle rupture, through inducing ovulatory genes, such as Areg, Ereg, Has2 and Tnfaip6, as well as increasing intracellular cAMP levels. PGE2 reduces extracellular matrix viscosity and thereby optimizes the conditions for sperm penetration. PGE2 reduces the phagocytic activity of polymorphonuclear neutrophils (PMNs) against sperm. In the presence of PGE2, sperm function and binding capacity to oocytes are enhanced. PGE2 maintains luteal function for embryo development and early implantation. In addition, it induces chemokine expression for trophoblast apposition and adhesion to the decidua for implantation. CONCLUSION It has been shown that PGE2 positively affects different stages of female fertility. Therefore, PGE2 should be taken into consideration when optimizing reproduction in infertile females. We suggest that in clinical practice, the administration of non-steroidal anti-inflammatory drugs, which are PGE2 synthesis inhibitors, should be reasonable and limited in infertile women. Additionally, assessments of PGE2 protein and receptor expression levels should be taken into consideration.
Collapse
Affiliation(s)
- Jean Damascene Niringiyumukiza
- 0000 0004 0368 7223grid.33199.31Family Planning Research Institute/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Hongcai Cai
- 0000 0004 0368 7223grid.33199.31Family Planning Research Institute/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Wenpei Xiang
- 0000 0004 0368 7223grid.33199.31Family Planning Research Institute/Center of Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| |
Collapse
|
28
|
Nuttinck F, Jouneau A, Charpigny G, Hue I, Richard C, Adenot P, Ruffini S, Laffont L, Chebrout M, Duranthon V, Guienne BML. Prosurvival effect of cumulus prostaglandin G/H synthase 2/prostaglandin2 signaling on bovine blastocyst: impact on in vivo posthatching development. Biol Reprod 2017; 96:531-541. [PMID: 28339853 PMCID: PMC5819843 DOI: 10.1095/biolreprod.116.145367] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/24/2017] [Indexed: 12/29/2022] Open
Abstract
Apoptotic activity is a common physiological process which culminates at the blastocyst stage in the preimplantation embryo of many mammals. The degree of embryonic cell death can be influenced by the oocyte microenvironment. However, the prognostic significance of the incidence of apoptosis remains undefined. Prostaglandin E2 (PGE2) derived from prostaglandin G/H synthase-2 (PTGS2) activity is a well-known prosurvival factor that is mainly studied in oncology. PGE2 is the predominant PTGS2-derived prostaglandin present in the oocyte microenvironment during the periconceptional period. Using an in vitro model of bovine embryo production followed by transfer and collection procedures, we investigated the impact of periconceptional PGE2 on the occurrence of spontaneous apoptosis in embryos and on subsequent in vivo posthatching development. Different periconceptional PGE2 environments were obtained using NS-398, a specific inhibitor of PTGS2 activity, and exogenous PGE2. We assessed the level of embryonic cell death in blastocysts at day 8 postfertilization by counting total cell numbers, by the immunohistochemical staining of active caspase-3, and by quantifying terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling signals and apoptosis regulator (BCL-2/BAX) mRNA expression. Morphometric parameters were used to estimate the developmental stage of the embryonic disk and the extent of trophoblast elongation on day 15 conceptuses. Our findings indicate that periconceptional PGE2 signaling durably impacts oocytes, conferring increased resistance to spontaneous apoptosis in blastocysts and promoting embryonic disk development and the elongation process during preimplantation development.
Collapse
Affiliation(s)
| | - Alice Jouneau
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | - Gilles Charpigny
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | - Isabelle Hue
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | | | - Pierre Adenot
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | - Sylvie Ruffini
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | - Ludivine Laffont
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | - Martine Chebrout
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | | | | |
Collapse
|
29
|
Khajeh M, Rahbarghazi R, Nouri M, Darabi M. Potential role of polyunsaturated fatty acids, with particular regard to the signaling pathways of arachidonic acid and its derivatives in the process of maturation of the oocytes: Contemporary review. Biomed Pharmacother 2017; 94:458-467. [PMID: 28779707 DOI: 10.1016/j.biopha.2017.07.140] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 01/20/2023] Open
Abstract
Oocyte meiotic maturation is one of the significant physiological requirements for ovulation and fertility. It is believed that Cyclic Adenosine Monophosphate, protein kinase A and protein kinase C pathways along with eicosanoids, particularly prostaglandin E2, and steroids are the key factors regulating mammalian oocyte maturation. The aim of the current study was to highlight the molecular events triggered by arachidonic acid during oocyte meiotic arrest and resumption at the time of gonadotrophin surge. It should be noted that arachidonic acid release is tightly regulated by Follicle-stimulating and Luteinizing hormones during oocyte development.
Collapse
Affiliation(s)
- Masoumeh Khajeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Single-cell analysis of differences in transcriptomic profiles of oocytes and cumulus cells at GV, MI, MII stages from PCOS patients. Sci Rep 2016; 6:39638. [PMID: 28004769 PMCID: PMC5177934 DOI: 10.1038/srep39638] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/24/2016] [Indexed: 11/09/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common frequent endocrine disorder among women of reproductive age. Although assisted reproductive techniques (ARTs) are used to address subfertility in PCOS women, their effectiveness is not clear. Our aim was to compare transcriptomic profiles of oocytes and cumulus cells (CCs) between women with and without PCOS, and assess the effectiveness of ARTs in treating PCOS patients. We collected oocytes and CCs from 16 patients with and without PCOS patients to categorize them into 6 groups according to oocyte nuclear maturation. Transcriptional gene expression of oocyte and CCs was determined via single-cell RNA sequencing. The ratio of fertilization and cleavage was higher in PCOS patients than in non-PCOS patients undergoing ARTs, and there was no difference in the number of high-quality embryos between the groups. Differentially expressed genes including PPP2R1A, PDGFRA, EGFR, GJA1, PTGS2, TNFAIP6, TGF-β1, CAV1, INHBB et al. were investigated as potential causes of PCOS oocytes and CCs disorder at early stages, but their expression returned to the normal level at the metaphase II (MII) stage via ARTs. In conclusion, ARTs can improve the quality of cumulus-oocyte complex (COC) and increase the ratio of fertilization and cleavage in PCOS women.
Collapse
|
31
|
Kemiläinen H, Adam M, Mäki-Jouppila J, Damdimopoulou P, Damdimopoulos AE, Kere J, Hovatta O, Laajala TD, Aittokallio T, Adamski J, Ryberg H, Ohlsson C, Strauss L, Poutanen M. The Hydroxysteroid (17β) Dehydrogenase Family Gene HSD17B12 Is Involved in the Prostaglandin Synthesis Pathway, the Ovarian Function, and Regulation of Fertility. Endocrinology 2016; 157:3719-3730. [PMID: 27490311 DOI: 10.1210/en.2016-1252] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The hydroxysteroid (17beta) dehydrogenase (HSD17B)12 gene belongs to the hydroxysteroid (17β) dehydrogenase superfamily, and it has been implicated in the conversion of estrone to estradiol as well as in the synthesis of arachidonic acid (AA). AA is a precursor of prostaglandins, which are involved in the regulation of female reproduction, prompting us to study the role of HSD17B12 enzyme in the ovarian function. We found a broad expression of HSD17B12 enzyme in both human and mouse ovaries. The enzyme was localized in the theca interna, corpus luteum, granulosa cells, oocytes, and surface epithelium. Interestingly, haploinsufficiency of the HSD17B12 gene in female mice resulted in subfertility, indicating an important role for HSD17B12 enzyme in the ovarian function. In line with significantly increased length of the diestrous phase, the HSD17B+/- females gave birth less frequently than wild-type females, and the litter size of HSD17B12+/- females was significantly reduced. Interestingly, we observed meiotic spindle formation in immature follicles, suggesting defective meiotic arrest in HSD17B12+/- ovaries. The finding was further supported by transcriptome analysis showing differential expression of several genes related to the meiosis. In addition, polyovular follicles and oocytes trapped inside the corpus luteum were observed, indicating a failure in the oogenesis and ovulation, respectively. Intraovarian concentrations of steroid hormones were normal in HSD17B12+/- females, whereas the levels of AA and its metabolites (6-keto prostaglandin F1alpha, prostaglandin D2, prostaglandin E2, prostaglandin F2α, and thromboxane B2) were decreased. In conclusion, our study demonstrates that HSD17B12 enzyme plays an important role in female fertility through its role in AA metabolism.
Collapse
Affiliation(s)
- Heidi Kemiläinen
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Marion Adam
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Jenni Mäki-Jouppila
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Pauliina Damdimopoulou
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Anastasios E Damdimopoulos
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Juha Kere
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Outi Hovatta
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Teemu D Laajala
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Tero Aittokallio
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Jerzy Adamski
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Henrik Ryberg
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Claes Ohlsson
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Leena Strauss
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| | - Matti Poutanen
- Department of Physiology and Turku Center for Disease Modeling (H.K., M.A., J.M.-J., T.D.L., L.S., M.P.), Institute of Biomedicine, University of Turku, FI-20540 Turku, Finland; Department of Clinical Science, Intervention and Technology (P.D., O.H.), Karolinska Institute, 141 52 Huddinge, Sweden; Swedish Toxicology Sciences Research Center (P.D.), Karolinska Institutet, 141 86 Stockholm, Sweden; Department of Biosciences and Nutrition (A.E.D., J.K.), Karolinska Institutet, 171 77 Stockholm, Sweden; Department of Mathematics and Statistics (T.D.L., T.A.), University of Turku, FI-20014 Turku, Finland; Institute for Molecular Medicine Finland (T.A.), University of Helsinki, FI-00014 Helsinki, Finland; Experimental Genetics (J.A.), Center of Life and Food Sciences, Weihenstephan, 85354 Freising, Germany; Institute of experimental Genetics (J.A.), Helmholtz Zentrum, 81377 München, Germany; Genome Analysis Center (J.A.), German Research Center for Environmental Health, 85764 Neuherberg, Germany; Institute of Neuroscience and Physiology (H.R.), Sahlgrenska Academy, University of Gothenburg, SE-405 30 Gothenburg, Sweden; Institute of Medicine (C.O., M.P.), The Sahlgrenska Academy, University of Gothenburg, SE-413 46 Gothenburg, Sweden
| |
Collapse
|
32
|
Kim SO, Duffy DM. Mapping PTGERs to the Ovulatory Follicle: Regional Responses to the Ovulatory PGE2 Signal. Biol Reprod 2016; 95:33. [PMID: 27307073 PMCID: PMC5029471 DOI: 10.1095/biolreprod.116.140574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/27/2016] [Accepted: 06/03/2016] [Indexed: 12/11/2022] Open
Abstract
Prostaglandin E2 (PGE2) is a key intrafollicular mediator of ovulation in many, if not all, mammalian species. PGE2 acts at follicular cells via four distinct PGE2 receptors (PTGERs). Within the ovulatory follicle, each cell type (e.g., oocyte, cumulus granulosa cell, mural granulosa cell, theca cell, endothelial cell) expresses a different subset of the four PTGERs. Expression of a subset of PTGERs has consequences for the generation of intracellular signals and ultimately the unique functions of follicular cells that respond to PGE2. Just as the ovulatory LH surge regulates PGE2 synthesis, the LH surge also regulates expression of the four PTGERs. The pattern of expression of the four PTGERs among follicular cells before and after the LH surge forms a spatial and temporal map of PGE2 responses. Differential PTGER expression, coupled with activation of cell-specific intracellular signals, may explain how a single paracrine mediator can have pleotropic actions within the ovulatory follicle. Understanding the role of each PTGER in ovulation may point to previously unappreciated opportunities to both promote and prevent fertility.
Collapse
Affiliation(s)
- Soon Ok Kim
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Diane M Duffy
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| |
Collapse
|
33
|
Dau AMP, da Silva EP, da Rosa PRA, Bastiani FT, Gutierrez K, Ilha GF, Comim FV, Gonçalves PBD. Bovine ovarian cells have (pro)renin receptors and prorenin induces resumption of meiosis in vitro. Peptides 2016; 81:1-8. [PMID: 27060674 DOI: 10.1016/j.peptides.2016.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 03/14/2016] [Accepted: 03/23/2016] [Indexed: 01/02/2023]
Abstract
The discovery of a receptor that binds prorenin and renin in human endothelial and mesangial cells highlights the possible effect of renin-independent prorenin in the resumption of meiosis in oocytes that was postulated in the 1980s.This study aimed to identify the (pro)renin receptor in the ovary and to assess the effect of prorenin on meiotic resumption. The (pro)renin receptor protein was detected in bovine cumulus-oocyte complexes, theca cells, granulosa cells, and in the corpus luteum. Abundant (pro)renin receptor messenger ribonucleic acid (mRNA) was detected in the oocytes and cumulus cells, while prorenin mRNA was identified in the cumulus cells only. Prorenin at concentrations of 10(-10), 10(-9), and 10(-8)M incubated with oocytes co-cultured with follicular hemisections for 15h caused the resumption of oocyte meiosis. Aliskiren, which inhibits free renin and receptor-bound renin/prorenin, at concentrations of 10(-7), 10(-5), and 10(-3)M blocked this effect (P<0.05). To determine the involvement of angiotensin II in prorenin-induced meiosis resumption, cumulus-oocyte complexes and follicular hemisections were treated with prorenin and with angiotensin II or saralasin (angiotensin II antagonist). Prorenin induced the resumption of meiosis independently of angiotensin II. Furthermore, cumulus-oocyte complexes cultured with forskolin (200μM) and treated with prorenin and aliskiren did not exhibit a prorenin-induced resumption of meiosis (P<0.05). Only the oocytes' cyclic adenosine monophosphate levels seemed to be regulated by prorenin and/or forskolin treatment after incubation for 6h. To the best of our knowledge, this is the first study to identify the (pro)renin receptor in ovarian cells and to demonstrate the independent role of prorenin in the resumption of oocyte meiosis in cattle.
Collapse
Affiliation(s)
- Andressa Minussi Pereira Dau
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Federal University of Santa Maria, Roraima Avenue 1000, 97105-900, Veterinary Hospital, Santa Maria, RS, Brazil
| | - Eduardo Pradebon da Silva
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Federal University of Santa Maria, Roraima Avenue 1000, 97105-900, Veterinary Hospital, Santa Maria, RS, Brazil
| | - Paulo Roberto Antunes da Rosa
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Federal University of Santa Maria, Roraima Avenue 1000, 97105-900, Veterinary Hospital, Santa Maria, RS, Brazil
| | - Felipe Tusi Bastiani
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Federal University of Santa Maria, Roraima Avenue 1000, 97105-900, Veterinary Hospital, Santa Maria, RS, Brazil
| | - Karina Gutierrez
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Federal University of Santa Maria, Roraima Avenue 1000, 97105-900, Veterinary Hospital, Santa Maria, RS, Brazil
| | - Gustavo Freitas Ilha
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Federal University of Santa Maria, Roraima Avenue 1000, 97105-900, Veterinary Hospital, Santa Maria, RS, Brazil
| | - Fabio Vasconcellos Comim
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Federal University of Santa Maria, Roraima Avenue 1000, 97105-900, Veterinary Hospital, Santa Maria, RS, Brazil; Department of Medical Clinic, Faculty of Medicine, Federal University of Santa Maria, Roraima Avenue 1000, 97105-900, Santa Maria, RS, Brazil
| | - Paulo Bayard Dias Gonçalves
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Federal University of Santa Maria, Roraima Avenue 1000, 97105-900, Veterinary Hospital, Santa Maria, RS, Brazil.
| |
Collapse
|
34
|
Analysis of genome-wide copy number variations in Chinese indigenous and western pig breeds by 60 K SNP genotyping arrays. PLoS One 2014; 9:e106780. [PMID: 25198154 PMCID: PMC4157799 DOI: 10.1371/journal.pone.0106780] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 08/07/2014] [Indexed: 12/28/2022] Open
Abstract
Copy number variations (CNVs) represent a substantial source of structural variants in mammals and contribute to both normal phenotypic variability and disease susceptibility. Although low-resolution CNV maps are produced in many domestic animals, and several reports have been published about the CNVs of porcine genome, the differences between Chinese and western pigs still remain to be elucidated. In this study, we used Porcine SNP60 BeadChip and PennCNV algorithm to perform a genome-wide CNV detection in 302 individuals from six Chinese indigenous breeds (Tongcheng, Laiwu, Luchuan, Bama, Wuzhishan and Ningxiang pigs), three western breeds (Yorkshire, Landrace and Duroc) and one hybrid (Tongcheng×Duroc). A total of 348 CNV Regions (CNVRs) across genome were identified, covering 150.49 Mb of the pig genome or 6.14% of the autosomal genome sequence. In these CNVRs, 213 CNVRs were found to exist only in the six Chinese indigenous breeds, and 60 CNVRs only in the three western breeds. The characters of CNVs in four Chinese normal size breeds (Luchuan, Tongcheng and Laiwu pigs) and two minipig breeds (Bama and Wuzhishan pigs) were also analyzed in this study. Functional annotation suggested that these CNVRs possess a great variety of molecular function and may play important roles in phenotypic and production traits between Chinese and western breeds. Our results are important complementary to the CNV map in pig genome, which provide new information about the diversity of Chinese and western pig breeds, and facilitate further research on porcine genome CNVs.
Collapse
|
35
|
Effect of α-linolenic acid on oocyte maturation and embryo development of prepubertal sheep oocytes. Theriogenology 2014; 82:686-96. [DOI: 10.1016/j.theriogenology.2014.05.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 05/05/2014] [Accepted: 05/27/2014] [Indexed: 11/16/2022]
|
36
|
Rajput SK, Lee K, Zhenhua G, Di L, Folger JK, Smith GW. Embryotropic actions of follistatin: paracrine and autocrine mediators of oocyte competence and embryo developmental progression. Reprod Fertil Dev 2014; 26:37-47. [PMID: 24305175 DOI: 10.1071/rd13282] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Despite several decades since the birth of the first test tube baby and the first calf derived from an in vitro-fertilised embryo, the efficiency of assisted reproductive technologies remains less than ideal. Poor oocyte competence is a major factor limiting the efficiency of in vitro embryo production. Developmental competence obtained during oocyte growth and maturation establishes the foundation for successful fertilisation and preimplantation embryonic development. Regulation of molecular and cellular events during fertilisation and embryo development is mediated, in part, by oocyte-derived factors acquired during oocyte growth and maturation and programmed by factors of follicular somatic cell origin. The available evidence supports an important intrinsic role for oocyte-derived follistatin and JY-1 proteins in mediating embryo developmental progression after fertilisation, and suggests that the paracrine and autocrine actions of oocyte-derived growth differentiation factor 9, bone morphogenetic protein 15 and follicular somatic cell-derived members of the fibroblast growth factor family impact oocyte competence and subsequent embryo developmental progression after fertilisation. An increased understanding of the molecular mechanisms mediating oocyte competence and stage-specific developmental events during early embryogenesis is crucial for further improvements in assisted reproductive technologies.
Collapse
Affiliation(s)
- Sandeep K Rajput
- Laboratory of Mammalian Reproductive Biology and Genomics, Department of Animal Science, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | | | | | |
Collapse
|
37
|
Pradhan A, Olsson PE. Juvenile ovary to testis transition in zebrafish involves inhibition of ptges. Biol Reprod 2014; 91:33. [PMID: 24920039 DOI: 10.1095/biolreprod.114.119016] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The sex differentiation mechanisms in zebrafish (Danio rerio) remains elusive, partly because of the absence of sex chromosomes but also because the process appears to depend on the synchrony of multiple genes and possibly environmental factors. Zebrafish gonadal development is initiated through the development of immature oocytes. Depending on multiple signaling cues, in about half of the individuals, the juvenile ovaries degenerate or undergo apoptosis to initiate testes development while the other half maintains the oogenic pathway. We have previously shown that activation of NFκB and prostaglandin synthase 2 (ptgs2) results in female-biased sex ratios. Prostaglandin synthase and prostaglandins are involved in multiple physiological functions, including cell survival and apoptosis. In the present study, we show that inhibition of ptgs2 by meloxicam results in male-biased sex ratios. On further evaluation, we observed that exposure with the prostaglandin D2 (PGD2) analogue BW-245C induced SRY-box containing gene 9a (sox9a) and resulted in male-biased sex ratios. On the other hand, prostaglandin E2 (PGE2) treatment resulted in female-biased sex ratios and involved activation of NFκB and the β-catenin pathway as well as inhibition of sox9. Exposure to the β-catenin inhibitor PNU-74654 resulted in up-regulation of ptgds and male-biased sex ratios, further confirming the involvement of β-catenin in the female differentiation pathway. In this study, we show that PGD2 and PGE2 can program the gonads to either the testis or the ovary differentiation pathways, indicating that prostaglandins are involved in the regulation of zebrafish gonadal differentiation.
Collapse
Affiliation(s)
- Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden
| |
Collapse
|
38
|
Mo X, Wu G, Yuan D, Jia B, Liu C, Zhu S, Hou Y. Leukemia inhibitory factor enhances bovine oocyte maturation and early embryo development. Mol Reprod Dev 2014; 81:608-18. [PMID: 24687528 DOI: 10.1002/mrd.22327] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 03/23/2014] [Indexed: 11/12/2022]
Abstract
The present study was conducted to examine the effects of leukemia inhibitory factor (LIF) on bovine oocyte maturation and early embryo development in vitro. Results showed that LIF supplementation (25 ng/ml) enhanced nuclear maturation of intact cumulus-oocyte complexes (COCs) compared to the vehicle control. Similar results were observed in denuded oocytes, indicating that LIF directly influences oocyte development. LIF-treated oocytes showed a higher cortical-granule-migration rate and increased expression of CD9, a tetraspanin transmembrane protein essential for fertilization. After in vitro fertilization, oocytes receiving LIF supplementation exhibited a higher cleavage rate and yielded a significantly higher number of blastocysts. To further dissect the molecular mechanism underlying this LIF-induced bovine oocyte maturation phenotype, we examined the involvement of two signaling cascades, mitogen-activated protein kinases (MAPK3/1)- and the signal transducer and activator of transcription 3 (STAT3)-dependent pathways. Western blot results revealed that LIF phosphorylated MAPK3/1 and STAT3. Inhibition of MAPK3/1 activation with MEK inhibitor U0126 only partially blocked LIF-induced nuclear maturation, although it attenuated oocyte cytoplasmic maturation. Inhibition of JAK/STAT3 activation with a specific pharmacological inhibitor completely abolished the LIF-response in bovine oocyte. In summary, these data revealed a novel role for LIF in bovine oocyte maturation subsequent embryonic development.
Collapse
Affiliation(s)
- Xianhong Mo
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
39
|
Alteration of energy metabolism gene expression in cumulus cells affects oocyte maturation via MOS–mitogen-activated protein kinase pathway in dairy cows with an unfavorable “Fertil−” haplotype of one female fertility quantitative trait locus. Theriogenology 2014; 81:599-612. [DOI: 10.1016/j.theriogenology.2013.11.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 11/17/2013] [Accepted: 11/18/2013] [Indexed: 11/24/2022]
|
40
|
Jia B, Wu G, Fu X, Mo X, Du M, Hou Y, Zhu S. trans
-10, cis
-12 conjugated linoleic acid enhances in vitro maturation of porcine oocytes. Mol Reprod Dev 2013; 81:20-30. [DOI: 10.1002/mrd.22273] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/19/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Baoyu Jia
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology; China Agricultural University; Beijing People's Republic of China
| | - Guoquan Wu
- Yunnan Animal Science and Veterinary Institute; Kunming Yunnan People's Republic of China
| | - Xiangwei Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology; China Agricultural University; Beijing People's Republic of China
| | - Xianhong Mo
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences; China Agricultural University; Beijing People's Republic of China
| | - Ming Du
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology; China Agricultural University; Beijing People's Republic of China
| | - Yunpeng Hou
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences; China Agricultural University; Beijing People's Republic of China
| | - Shien Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology; China Agricultural University; Beijing People's Republic of China
| |
Collapse
|
41
|
Role of PTGS2-generated PGE2 during gonadotrophin-induced bovine oocyte maturation and cumulus cell expansion. Reprod Biomed Online 2013; 28:388-400. [PMID: 24447957 DOI: 10.1016/j.rbmo.2013.11.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 10/28/2013] [Accepted: 11/11/2013] [Indexed: 11/20/2022]
Abstract
Prostaglandin E2 (PGE2) is an autocrine/paracrine factor which mediates gonadotrophin (Gn) stimulation of cumulus expansion and oocyte maturation in rodents. Its role in bovine oocyte maturation is less characterized. This study detected PTGS2 (COX2) and PGE synthases (PTGES1, PTGES2 and PTGES3) in bovine cumulus-oocyte complexes (COC). Only PTGS2 and PTGES1 expression changed during maturation. In Gn-free media, no cumulus expansion and ∼45% nuclear maturation was achieved, while Gn-induced maturation showed full cumulus expansion (score 3) and ∼87% maturation. PGE2 supplementation without Gn induced mild cumulus expansion (score 0.5-1) but increased nuclear maturation to levels similar to those obtained with Gn alone. In the presence of Gn, exogenous PGE2 did not affect expansion or nuclear maturation and subsequent embryo development. Treatment with PTGS2 selective inhibitor (NS398), PTGS2-specific siRNA or PTGER2-receptor antagonist (AH6809) resulted in ∼20-25% reduction in nuclear maturation. NS398 and AH6809 did not affect cumulus expansion. Most oocytes not reaching metaphase of second meiosis (MII) following NS398, AH6809 and PTGS2-specific siRNA treatments were at MI. After longer maturation, NS398-treated oocytes had normal MII rate and uncompromised embryo development. PGE2 has a limited role in cumulus expansion in bovine COC but is important for the timing of Gn-induced nuclear maturation. We confirmed that genes involved in the synthesis of prostaglandin E2 (PGE2) are expressed by cumulus-oocyte complexes (or eggs) of cows and that PGE2 is synthesized during oocyte maturation in the presence of gonadotrophin hormones. When we inhibited synthesis of PGE2 or blocked its receptors, oocyte maturation, but not cumulus expansion, was compromised. Further investigation showed that oocyte maturation is delayed but not arrested when PGE2 synthesis is inhibited. On the other hand, addition of exogenous PGE2 induced a high maturation rate and mild cumulus expansion only in the absence of gonadotrophin stimulation, and had no effect in the presence of gonadotrophin.
Collapse
|
42
|
Angiotensin II, progesterone, and prostaglandins are sequential steps in the pathway to bovine oocyte nuclear maturation. Theriogenology 2012; 77:1779-87. [DOI: 10.1016/j.theriogenology.2011.12.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 12/12/2011] [Accepted: 12/15/2011] [Indexed: 11/18/2022]
|
43
|
Hobbs RJ, Howard J, Wildt DE, Comizzoli P. Absence of seasonal changes in FSHR gene expression in the cat cumulus-oocyte complex in vivo and in vitro. Reproduction 2012; 144:111-22. [PMID: 22596062 DOI: 10.1530/rep-12-0098] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Domestic cat oocytes are seasonally sensitive to FSH. Compared with those collected during the breeding season, oocytes from the nonbreeding (NB) season require more FSH during in vitro maturation to achieve comparable developmental competence. This study tested the hypothesis that this seasonal variation was due to altered expression of FSH receptors (FSHR) and/or FSH-induced genes. Relative expression levels of FSHR mRNA and FSH-enhanced gene estrogen receptor β (ESR2) were measured by qPCR in whole ovaries and immature cumulus-oocyte complexes (COCs) isolated from cat ovaries during the natural breeding vs NB seasons. Expression levels of FSH-induced genes prostaglandin-endoperoxide synthase 2 (PTGS2), early growth response protein-1 (EGR1), and epidermal growth factor receptor (EGFR) were examined in mature COCs from both seasons that were a) recovered in vivo or b) matured in vitro with conventional (1 μg/ml) or high (10 μg/ml) FSH concentrations. Overall, FSHR mRNA levels were lower in whole ovaries during the NB compared with breeding season but were similar in immature COCs, whereas ESR2 levels did not differ in either group between intervals. We observed changes in PTGS2, EGR1, and EGFR mRNA expression patterns across maturation in COCs within but not between the two seasons. The lack of seasonal differentiation in FSH-related genes was not consistent with the decreased developmental capacity of oocytes fertilized during the NB season. These findings reveal that the seasonal decrease in cat oocyte sensitivity to FSH occurs both in vivo and in vitro. Furthermore, this decline is unrelated to changes in expression of FSHR mRNA or mRNA of FSH-induced genes in COCs from antral follicles.
Collapse
Affiliation(s)
- Rebecca J Hobbs
- Smithsonian Conservation Biology Institute, Center for Species Survival, National Zoological Park, PO Box 37012, MRC 5502, Washington, District of Columbia 20013, USA.
| | | | | | | |
Collapse
|
44
|
Coyral-Castel S, Brisard D, Touzé JL, Dupont M, Ramé C, Uzbekova S, Dupont J. Analysis of in vivo oocyte maturation, in vitro embryo development and gene expression in cumulus cells of dairy cows and heifers selected for one fertility quantitative trait loci (QTL) located on BTA3. Theriogenology 2012; 77:1822-33.e1. [PMID: 22401835 DOI: 10.1016/j.theriogenology.2011.12.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 12/26/2011] [Accepted: 12/31/2011] [Indexed: 10/28/2022]
Abstract
We have previously shown that Holstein cows selected for their homozygous favorable ("fertil+") or unfavorable ("fertil-") haplotype at one quantitative trait loci (QTL) of female fertility located on chromosome 3 (QTL-F-Fert-BTA3) had a different success rate 35 and 90 days after the first artificial insemination. To determine whether the lower fertility in "fertil-" animals could be related to oocyte quality, we analyzed the embryo development rate in vitro and the oocyte meiotic maturation in vivo in "fertil+" and "fertil-" heifers. In vitro maturation and fertilization of immature oocytes recovered by ovum pick-up from "fertil+" and "fertil-" heifers resulted in similar cleavage and blastocyst rates in the two haplotypes. However the percentage of expanded blastocysts and the number of cells per blastocyst were significantly higher in "fertil+". Oocytes from presumptive preovulatory follicles were analyzed after ovarian stimulation. A similar rate of immature (from prophase to metaphase-I) and mature oocytes (metaphase-II) was obtained in the two haplotypes, whereas a significantly higher percentage of oocytes from metaphase-I to metaphase-II was observed in "fertil+" compared to "fertil-" heifers. Since cumulus cells (CCs) could reflect the developmental competence of oocytes, we analyzed the expression of seven genes included in the QTL-F-Fert-BTA3 using real-time PCR in bovine CCs after in vivo or in vitro maturation, as a model of higher and lower competence, respectively. Transcript levels of TAGLN2, EEF1A1 and PIGM were higher in CCs after in vitro maturation (IVM) compared to in vivo maturation, whereas no difference was observed for IFI16, KIRREL, SPTA1 and PEX19 expression. The expression levels of all these genes in in preovulatory CCs were not significantly different between "fertil+" and "fertil-" heifers. In conclusion, the lower fertility of "fertil-" females could be partially due to a lowest quality of the oocytes and consequently of preimplantation embryo development.
Collapse
Affiliation(s)
- S Coyral-Castel
- INRA, UMR85 Physiologie de la Reproduction et des comportements, F-37380 Nouzilly, France
| | | | | | | | | | | | | |
Collapse
|
45
|
Bilodeau-Goeseels S. Bovine oocyte meiotic inhibition before in vitro maturation and its value to in vitro embryo production: does it improve developmental competence? Reprod Domest Anim 2011; 47:687-93. [PMID: 21988654 DOI: 10.1111/j.1439-0531.2011.01924.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The efficiency of bovine in vitro embryo production has remained low despite extensive effort to understand the effects of culture conditions, media composition and supplementation. As bovine oocytes resume meiosis spontaneously when cultured, it was hypothesized that preventing meiosis in vitro before in vitro maturation (IVM) and in vitro fertilization (IVF) would allow more oocytes to acquire developmental competence. This article reviews some of the factors involved in meiotic arrest as well as the effects of meiotic inhibition before IVM on bovine oocytes developmental competence following IVF. Follicular components and cAMP-elevating agents can delay or inhibit meiosis in various proportions of oocytes; however, few studies have examined their effects on development following IVM and IVF because they are not practical (follicular components) or have a transient effect on meiosis (cAMP-elevating agents). Protein synthesis or phosphorylation inhibition prevented meiosis in high percentages of oocytes; however, these non-specific inhibitions led to lower developmental competence compared with non-arrested oocytes. Maturation promoting factor (MPF) inhibition with specific inhibitors has been examined in several studies. Despite faster maturation following removal from inhibition and some structural damage to the oocytes, MPF inhibition generally led to blastocyst rates similar to control, non-arrested oocytes. Future work will involve evaluating the effects on arrested oocytes of molecules that can improve developmental competence in non-arrested oocytes. It is also anticipated that new IVM systems that take into consideration new knowledge of the mechanisms involved in the control of meiosis will be developed. Moreover, global gene expression analysis studies will also provide clues to the culture conditions required for optimal expression of developmental competence.
Collapse
Affiliation(s)
- S Bilodeau-Goeseels
- Agriculture and Agri-Food Canada, Lethbridge Research Centre, Lethbridge, AB, Canada.
| |
Collapse
|