1
|
Caniçais C, Sobral D, Vasconcelos S, Cunha M, Pinto A, Mesquita Guimarães J, Santos F, Barros A, Dória S, Marques CJ. Transcriptomic analysis and epigenetic regulators in human oocytes at different stages of oocyte meiotic maturation. Dev Biol 2025; 519:55-64. [PMID: 39681207 DOI: 10.1016/j.ydbio.2024.12.004] [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: 11/18/2024] [Accepted: 12/11/2024] [Indexed: 12/18/2024]
Abstract
Human oocytes are highly specialized cells with the capacity to store and regulate mRNAs during oocyte maturation, in preparation for post-fertilization steps. Here we performed single-oocyte transcriptomic analysis of human oocytes in three meitoic maturation stages - Germinal Vesicle (GV; n = 6), Metaphase I (MI; n = 6) and Metaphase II (MII; n = 7). Single-oocyte transcriptomic analysis revealed that the total number of expressed genes progressively decreased from GV to MII stages, with 9660 genes being transcribed in GV, 8734 in MI and 5889 in MII. The same tendency was observed for the number of uniquely expressed genes, with 1328 uniquely expressed genes in GV, 401 in MI and 72 in MII. GO analysis of the uniquely expressed genes showed distinct terms in GV oocytes such as transferase activity, organonitrogen compound metabolic process and ncRNA processing. Analysis of Differentially Expressed Genes (DEGs) between the three maturation stages revealed 1165 DEGs between GV and MII oocytes, with 635 being upregulated and 528 downregulated, 42 DEGs between GV and MI, with 38 being upregulated and 4 downregulated, and no significant changes in gene expression between MI and MII oocytes. Comprehensive analysis of epigenetic regulators showed high expression of several histone-modifying enzymes, namely deacetylases, acetylases, lysine demethylases and methyltransferases, and DNA methylation regulators, namely the maintenance methyltransferase DNMT1 and its co-regulators DPPA3 and UHRF1. Some of these epigenetic regulators were differentially expressed between maturation stages, namely SIRT3, SIRT6, KDM3AP1, KMT2E, DNMT1, DPPA3 and the MEST and RASGRF1 imprinted genes. Our study contributes with important information on the transcriptional landscape of human oocytes in different stages of meiotic maturation, providing important insights into candidate biomarkers of human oocyte quality.
Collapse
Affiliation(s)
- Carla Caniçais
- Genetics Unit, Department of Pathology, Faculty of Medicine University of Porto (FMUP), 4200-319, Portugal; ICBAS- School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal
| | - Daniel Sobral
- Genomics and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016, Lisbon, Portugal
| | - Sara Vasconcelos
- Genetics Unit, Department of Pathology, Faculty of Medicine University of Porto (FMUP), 4200-319, Portugal
| | - Mariana Cunha
- Centre for Reproductive Genetics A Barros (CGRAB), 4100-009, Porto, Portugal
| | - Alice Pinto
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
| | | | - Fátima Santos
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Alberto Barros
- Genetics Unit, Department of Pathology, Faculty of Medicine University of Porto (FMUP), 4200-319, Portugal; Centre for Reproductive Genetics A Barros (CGRAB), 4100-009, Porto, Portugal; PROCRIAR Fertility Clinic, 4100-130, Porto, Portugal
| | - Sofia Dória
- Genetics Unit, Department of Pathology, Faculty of Medicine University of Porto (FMUP), 4200-319, Portugal; ICBAS- School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal
| | - C Joana Marques
- Genetics Unit, Department of Pathology, Faculty of Medicine University of Porto (FMUP), 4200-319, Portugal; CINTESIS@RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
2
|
Saadeldin IM, Ehab S, Alshammari MEF, Abdelazim AM, Assiri AM. The Mammalian Oocyte: A Central Hub for Cellular Reprogramming and Stemness. Stem Cells Cloning 2025; 18:15-34. [PMID: 39991743 PMCID: PMC11846613 DOI: 10.2147/sccaa.s513982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
The mammalian oocyte is pivotal in reproductive biology, acting as a central hub for cellular reprogramming and stemness. It uniquely contributes half of the zygotic nuclear genome and the entirety of the mitochondrial genome, ensuring individual development and health. Oocyte-mediated reprogramming, exemplified by nuclear transfer, resets somatic cell identity to achieve pluripotency and has transformative potential in regenerative medicine. This process is critical for understanding cellular differentiation, improving assisted reproductive technologies, and advancing cloning and stem cell research. During fertilization, the maternal-zygotic transition shifts developmental control from maternal factors to zygotic genome activation, establishing totipotency. Oocytes also harbor reprogramming factors that guide nuclear remodeling, epigenetic modifications, and metabolic reprogramming, enabling early embryogenesis. Structures like mitochondria, lipid droplets, and cytoplasmic lattices contribute to energy production, molecular regulation, and cellular organization. Recent insights into oocyte components, such as ooplasmic nanovesicles and endolysosomal vesicular assemblies (ELVAS), highlight their roles in maintaining cellular homeostasis, protein synthesis, and reprogramming efficiency. By unraveling the reprogramming mechanisms inherent in oocytes, we advance our understanding of cloning, cell differentiation, and stem cell therapy, highlighting their valuable significance in developmental biology and regenerative medicine.
Collapse
Affiliation(s)
- Islam M Saadeldin
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia
| | - Seif Ehab
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | | | - Aaser M Abdelazim
- Department of Medical Laboratories Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, 67714, Saudi Arabia
| | - Abdullah M Assiri
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia
| |
Collapse
|
3
|
Li N, Wang H, Zou S, Yu X, Li J. Perspective in the Mechanisms for Repairing Sperm DNA Damage. Reprod Sci 2025; 32:41-51. [PMID: 39333437 PMCID: PMC11729216 DOI: 10.1007/s43032-024-01714-5] [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: 06/08/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
DNA damage in spermatozoa is a major cause of male infertility. It is also associated with adverse reproductive outcomes (including reduced fertilization rates, embryo quality and pregnancy rates, and higher rates of spontaneous miscarriage). The damage to sperm DNA occurs during the production and maturation of spermatozoa, as well as during their transit through the male reproductive tract. DNA damage repair typically occurs during spermatogenesis, oocytes after fertilization, and early embryonic development stages. The known mechanisms of sperm DNA repair mainly include nucleotide excision repair (NER), base excision repair (BER), mismatch repair (MMR), and double-strand break repair (DSBR). The most severe type of sperm DNA damage is double-strand break, and it will be repaired by DSBR, including homologous recombination (HR), classical non-homologous end joining (cNHEJ), alternative end joining (aEJ), and single-strand annealing (SSA). However, the precise mechanisms of DNA repair in spermatozoa remain incompletely understood. DNA repair-associated proteins are of great value in the repair of sperm DNA. Several repair-related proteins have been identified as playing critical roles in condensing chromatin, regulating transcription, repairing DNA damage, and regulating the cell cycle. It is noteworthy that XRCC4-like factor (XLF) and paralog of XRCC4 and XLF (PAXX) -mediated dimerization promote the processing of populated ends for cNHEJ repair, which suggests that XLF and PAXX have potential value in the mechanism of sperm DNA repair. This review summarizes the classic and potential repair mechanisms of sperm DNA damage, aiming to provide a perspective for further research on DNA damage repair mechanisms.
Collapse
Affiliation(s)
- Nihong Li
- Chengdu Fifth People's Hospital, The Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Hong Wang
- Chengdu Fifth People's Hospital, The Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Siying Zou
- Chengdu Fifth People's Hospital, The Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Xujun Yu
- College of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Junjun Li
- Chengdu Fifth People's Hospital, The Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China.
| |
Collapse
|
4
|
Harris AL, Dinopoulou V, Loutradis D, Drakakis P, Kiessling AA. Microarray evidence that 8-cell human embryos express some hormone family members including oxytocin. J Assist Reprod Genet 2024; 41:323-332. [PMID: 38133877 PMCID: PMC10894797 DOI: 10.1007/s10815-023-03002-8] [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: 04/26/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023] Open
Abstract
OBJECTIVE This study is to discover hormone pathways active in early cleaving human embryos. METHODS A list of 152 hormones and receptors were compiled to query the microarray database of mRNAs in 8-cell human embryos, two lines of human embryonic stem cells plus human fibroblasts before and after induced pluripotency. RESULTS Over half of the 152 hormones and receptors were silent on the arrays of all cell types, and more were detected at high or moderate levels on the 8-cell arrays than on the pluripotent cell or fibroblast arrays. Eight hormone family genes were uniquely detected at least 22-fold higher on the 8-cell arrays than the stem cell arrays: AVPI1, CCK, CORT, FSTL4, GIP, GPHA2, OXT, and PPY suggesting novel roles for these proteins in early development. Oxytocin was detected by pilot immunoassay in culture media collected from Day 3 embryos. Robust detection of CRHR1 and EPOR suggests the 8-cell embryo may be responsive to maternal CRH and EPO. The over-expression of POMC and GHITM suggests POMP peptide products may have undiscovered roles in early development and GHITM may contribute to mitochondrial remodeling. Under-detected on the 8-cell arrays at least tenfold were two key enzymes in steroid biosynthesis, DHCR24 and FDPS. CONCLUSIONS The 8-cell human embryo may be secreting oxytocin, which could stimulate its own progress down the fallopian tube as well as play a role in early neural precursor development. The 8-cell embryo does not synthesize reproductive steroid hormones. As previously reported for growth factor families, the early embryo over-expresses more hormones than hormone receptors.
Collapse
Affiliation(s)
- Amy Lee Harris
- Department of Obstetrics and Gynecology, Harvard Medical School, Massachusetts General Hospital Fertility Center, Boston, MA, USA
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Fairborn, OH, USA
| | - Vasiliki Dinopoulou
- 1St Department of Obstetrics and Gynecology, Alexandra Hospital, Athens University Medical School, Lourou 4-2, 115 28, Athina, Greece
| | - Dimitris Loutradis
- 1St Department of Obstetrics and Gynecology, Alexandra Hospital, Athens University Medical School, Lourou 4-2, 115 28, Athina, Greece
| | - Peter Drakakis
- 1St Department of Obstetrics and Gynecology, Alexandra Hospital, Athens University Medical School, Lourou 4-2, 115 28, Athina, Greece
| | | |
Collapse
|
5
|
Song Y, Zhang N, Zhang Y, Wang J, Lv Q, Zhang J. Single-Cell Transcriptome Analysis Reveals Development-Specific Networks at Distinct Synchronized Antral Follicle Sizes in Sheep Oocytes. Int J Mol Sci 2024; 25:910. [PMID: 38255985 PMCID: PMC10815039 DOI: 10.3390/ijms25020910] [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: 12/13/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The development of the ovarian antral follicle is a complex, highly regulated process. Oocytes orchestrate and coordinate the development of mammalian ovarian follicles, and the rate of follicular development is governed by a developmental program intrinsic to the oocyte. Characterizing oocyte signatures during this dynamic process is critical for understanding oocyte maturation and follicular development. Although the transcriptional signature of sheep oocytes matured in vitro and preovulatory oocytes have been previously described, the transcriptional changes of oocytes in antral follicles have not. Here, we used single-cell transcriptomics (SmartSeq2) to characterize sheep oocytes from small, medium, and large antral follicles. We characterized the transcriptomic landscape of sheep oocytes during antral follicle development, identifying unique features in the transcriptional atlas, stage-specific molecular signatures, oocyte-secreted factors, and transcription factor networks. Notably, we identified the specific expression of 222 genes in the LO, 8 and 6 genes that were stage-specific in the MO and SO, respectively. We also elucidated signaling pathways in each antral follicle size that may reflect oocyte quality and in vitro maturation competency. Additionally, we discovered key biological processes that drive the transition from small to large antral follicles, revealing hub genes involved in follicle recruitment and selection. Thus, our work provides a comprehensive characterization of the single-oocyte transcriptome, filling a gap in the mapping of the molecular landscape of sheep oogenesis. We also provide key insights into the transcriptional regulation of the critical sizes of antral follicular development, which is essential for understanding how the oocyte orchestrates follicular development.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiaxin Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.S.)
| |
Collapse
|
6
|
Ducreux B, Ferreux L, Patrat C, Fauque P. Overview of Gene Expression Dynamics during Human Oogenesis/Folliculogenesis. Int J Mol Sci 2023; 25:33. [PMID: 38203203 PMCID: PMC10778858 DOI: 10.3390/ijms25010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
The oocyte transcriptome follows a tightly controlled dynamic that leads the oocyte to grow and mature. This succession of distinct transcriptional states determines embryonic development prior to embryonic genome activation. However, these oocyte maternal mRNA regulatory events have yet to be decoded in humans. We reanalyzed human single-oocyte RNA-seq datasets previously published in the literature to decrypt the transcriptomic reshuffles ensuring that the oocyte is fully competent. We applied trajectory analysis (pseudotime) and a meta-analysis and uncovered the fundamental transcriptomic requirements of the oocyte at any moment of oogenesis until reaching the metaphase II stage (MII). We identified a bunch of genes showing significant variation in expression from primordial-to-antral follicle oocyte development and characterized their temporal regulation and their biological relevance. We also revealed the selective regulation of specific transcripts during the germinal vesicle-to-MII transition. Transcripts associated with energy production and mitochondrial functions were extensively downregulated, while those associated with cytoplasmic translation, histone modification, meiotic processes, and RNA processes were conserved. From the genes identified in this study, some appeared as sensitive to environmental factors such as maternal age, polycystic ovary syndrome, cryoconservation, and in vitro maturation. In the future, the atlas of transcriptomic changes described in this study will enable more precise identification of the transcripts responsible for follicular growth and oocyte maturation failures.
Collapse
Affiliation(s)
- Bastien Ducreux
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Développement (GAD) INSERM UMR1231, 2 Rue Angélique Ducoudray, F-21000 Dijon, France;
| | - Lucile Ferreux
- Faculty of Medicine, Inserm 1016, Université de Paris Cité, F-75014 Paris, France; (L.F.); (C.P.)
- Department of Reproductive Biology-CECOS, Aphp.Centre-Université Paris Cité, Cochin, F-75014 Paris, France
| | - Catherine Patrat
- Faculty of Medicine, Inserm 1016, Université de Paris Cité, F-75014 Paris, France; (L.F.); (C.P.)
- Department of Reproductive Biology-CECOS, Aphp.Centre-Université Paris Cité, Cochin, F-75014 Paris, France
| | - Patricia Fauque
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Développement (GAD) INSERM UMR1231, 2 Rue Angélique Ducoudray, F-21000 Dijon, France;
- Laboratoire de Biologie de la Reproduction-CECOS, CHU Dijon Bourgogne, 14 Rue Gaffarel, F-21000 Dijon, France
| |
Collapse
|
7
|
Nguyen TTA, Demeestere I. A Journey to Reach the Ovary Using Next-Generation Technologies. Int J Mol Sci 2023; 24:16593. [PMID: 38068916 PMCID: PMC10705884 DOI: 10.3390/ijms242316593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Although effective in terms of the chances of future live birth, the current methods for fertility preservation, such as oocyte, embryo, or ovarian tissue cryopreservation, cannot be offered to all cancer patients in all clinical contexts. Expanding options for fertility preservation is crucial to addressing the need to encompass all situations. One emerging strategy is pharmacoprotection, a non-invasive approach that has the potential to fill existing gaps in fertility preservation. In addition to the identification of the most effective therapeutic agents, the potential for off-target effects remains one of the main limitations of this strategy for clinical application, particularly when healthy ovarian tissue is targeted. This review focuses on the advances in pharmacoprotective approaches and the challenge of targeting the ovaries to deliver these agents. The unique properties of gold nanoparticles (AuNPs) make them an attractive candidate for this purpose. We discuss how AuNPs meet many of the requirements for an ideal drug delivery system, as well as the existing limitations that have hindered the progression of AuNP research into more clinical trials. Additionally, the review highlights microRNA (miRNA) therapy as a next-generation approach to address the issues of fertility preservation and discusses the obstacles that currently impede its clinical availability.
Collapse
Affiliation(s)
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
| |
Collapse
|
8
|
Braga DPAF, Setti A, Morishima C, Provenza RR, Iaconelli A, Borges E. The effect of sperm DNA fragmentation on ICSI outcomes depending on oocyte quality. Andrology 2023; 11:1682-1693. [PMID: 37004191 DOI: 10.1111/andr.13435] [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: 03/22/2022] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND Sperm deoxyribonucleic acid (DNA) fragmentation is commonly encountered in spermatozoa, and the oocyte assumes responsibility for repairing sperm DNA fragmentation during the oocyte-embryo transition. OBJECTIVES This study aimed to investigate whether the effect of sperm DNA fragmentation on intracytoplasmic sperm injection outcomes depends on the incidence of oocyte dimorphisms. MATERIALS AND METHODS For the present cohort, 2942 fertilized oocytes from 525 patients submitted to intracytoplasmic sperm injection cycles were assessed. The present study was conducted in a private in vitro fertilization center affiliated to a university from June 2016 to July 2019. Semen samples were divided into the following two groups depending on the sperm DNA fragmentation index: a low fragmentation index group (<30% sperm DNA fragmentation, n = 1468) and a high fragmentation index group (≥30% sperm DNA fragmentation, n = 486). In addition, mature oocytes were examined before sperm injection, and intracytoplasmic and extracytoplasmic defects were recorded. The effect of the sperm DNA fragmentation index on laboratory and clinical intracytoplasmic sperm injection outcomes (depending on the presence of oocyte defects) was evaluated. RESULTS Significant increases in the rates of fertilization, high-quality embryo, implantation, and pregnancy were noted for cycles with <30% sperm DNA fragmentation than cycles with ≥30% sperm DNA fragmentation (regardless of the presence of oocyte dimorphisms). The presence of dimorphisms significantly impacted laboratory and clinical outcomes. The lowest fertilization and high-quality embryo rates were observed when a high sperm DNA fragmentation index was associated with the presence of dark cytoplasm, vacuoles, resistant membrane, and non-resistant membrane. The lowest implantation and pregnancy rates were observed when a high sperm DNA fragmentation index was associated with the presence of vacuoles, defective perivitelline space, and fragmented polar body. The effect of sperm DNA fragmentation on miscarriage rates was significantly influenced by the presence of centrally located cytoplasmic granulation, a defective perivitelline space and non-resistant membrane. CONCLUSION A high sperm DNA fragmentation index increases the likelihood of miscarriage in intracytoplasmic sperm injection cycles, an effect that may potentially be magnified by the presence of oocyte dysmorphisms.
Collapse
Affiliation(s)
| | | | - Christina Morishima
- Instituto Sapientiae-Centro de Estudos e Pesquisa em Reprodução Assistida, Sao Paulo, Brazil
| | | | | | | |
Collapse
|
9
|
Dučić T, Sanchez-Mata A, Castillo-Sanchez J, Algarra M, Gonzalez-Munoz E. Monitoring oocyte-based human pluripotency acquisition using synchrotron-based FTIR microspectroscopy reveals specific biomolecular trajectories. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 297:122713. [PMID: 37084681 DOI: 10.1016/j.saa.2023.122713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/09/2023] [Accepted: 04/03/2023] [Indexed: 05/03/2023]
Abstract
The reprogramming of human somatic cells to induced pluripotent cells (iPSCs) has become a milestone and a paradigm shift in the field of regenerative medicine and human disease modeling including drug testing and genome editing. However, the molecular processes occurring during reprogramming and affecting the pluripotent state acquired remain largely unknown. Of interest, different pluripotent states have been described depending on the reprogramming factors used and the oocyte has emerged as a valuable source of information for candidate factors. The present study investigates the molecular changes occurring in somatic cells during reprogramming with either canonical (OSK) or oocyte-based (AOX15) combinations using synchrotron-radiation Fourier transform infrared (SR FTIR) spectroscopy. The data acquired by SR FTIR indicates different representation and conformation of biological relevant macromolecules (lipids, nucleic acids, carbohydrates and proteins) depending on the reprogramming combination used and at different stages during the reprogramming process. Association analysis based on cells spectra suggest that pluripotency acquisition trajectories converge at late intermediate stages while they diverge at early stages. Our results suggest that OSK and AOX15 reprogramming operates through differential mechanisms affecting nucleic acids reorganization and day 10 comes out as a candidate hinge point to further study the molecular pathways involved in the reprogramming process. This study indicates that SR FTIR approach contribute unpaired information to distinguish pluripotent states and to decipher pluripotency acquisition roadmaps and landmarks that will enable advanced biomedical applications of iPSCs.
Collapse
Affiliation(s)
- Tanja Dučić
- ALBA Synchrotron Light Source, Carrer de la Llum 2-26, 08290 Cerdanyola del Vallès, Barcelona, Spain.
| | - Alicia Sanchez-Mata
- Biomedical Research Institute and Nanomedicine Platform (IBIMA-BIONAND), C/ Severo Ochoa, 35, Malaga, Spain; Department of Cell Biology, Genetics and Physiology, University of Málaga, 29071 Málaga, Spain
| | - Jesus Castillo-Sanchez
- Biomedical Research Institute and Nanomedicine Platform (IBIMA-BIONAND), C/ Severo Ochoa, 35, Malaga, Spain; Department of Cell Biology, Genetics and Physiology, University of Málaga, 29071 Málaga, Spain
| | - Manuel Algarra
- INAMAT(2) Institute for Advanced Materials and Mathematics, Department of Sciences, Public University of Navarre, Campus de Arrosadia, 31006 Pamplona, Spain
| | - Elena Gonzalez-Munoz
- Biomedical Research Institute and Nanomedicine Platform (IBIMA-BIONAND), C/ Severo Ochoa, 35, Malaga, Spain; Department of Cell Biology, Genetics and Physiology, University of Málaga, 29071 Málaga, Spain.
| |
Collapse
|
10
|
Paulino LRFM, de Assis EIT, Azevedo VAN, Silva BR, da Cunha EV, Silva JRV. Why Is It So Difficult To Have Competent Oocytes from In vitro Cultured Preantral Follicles? Reprod Sci 2022; 29:3321-3334. [PMID: 35084715 DOI: 10.1007/s43032-021-00840-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022]
Abstract
The developmental competence of oocytes is acquired gradually during follicular development, mainly through oocyte accumulation of RNA molecules and proteins that will be used during fertilization and early embryonic development. Several attempts to develop in vitro culture systems to support preantral follicle development up to maturation are reported in the literature, but oocyte competence has not yet been achieved in human and domestic animals. The difficulties to have fertilizable oocytes are related to thousands of mRNAs and proteins that need to be synthesized, long-term duration of follicular development, size of preovulatory follicles, composition of in vitro culture medium, and the need of multi-step culture systems. The development of a culture system that maintains bidirectional communication between the oocyte and granulosa cells and that meets the metabolic demands of each stage of follicle growth is the key to sustain an extended culture period. This review discusses the physiological and molecular mechanisms that determine acquisition of oocyte competence in vitro, like oocyte transcriptional activity, follicle and oocyte sizes, and length and regulation of follicular development in murine, human, and domestic animal species. The state of art of in vitro follicular development and the challenges to have complete follicular development in vitro are also highlighted.
Collapse
Affiliation(s)
- Laís R F M Paulino
- Laboratory of Biotechnology and Physiology of Reproduction (LABIREP), Federal University of Ceara, Av. Comandante Maurocélio Rocha Ponte 100, Sobral, CE, CEP 62041-040, Brazil
| | - Ernando I T de Assis
- Laboratory of Biotechnology and Physiology of Reproduction (LABIREP), Federal University of Ceara, Av. Comandante Maurocélio Rocha Ponte 100, Sobral, CE, CEP 62041-040, Brazil
| | - Venância A N Azevedo
- Laboratory of Biotechnology and Physiology of Reproduction (LABIREP), Federal University of Ceara, Av. Comandante Maurocélio Rocha Ponte 100, Sobral, CE, CEP 62041-040, Brazil
| | - Bianca R Silva
- Laboratory of Biotechnology and Physiology of Reproduction (LABIREP), Federal University of Ceara, Av. Comandante Maurocélio Rocha Ponte 100, Sobral, CE, CEP 62041-040, Brazil
| | - Ellen V da Cunha
- Laboratory of Biotechnology and Physiology of Reproduction (LABIREP), Federal University of Ceara, Av. Comandante Maurocélio Rocha Ponte 100, Sobral, CE, CEP 62041-040, Brazil
| | - José R V Silva
- Laboratory of Biotechnology and Physiology of Reproduction (LABIREP), Federal University of Ceara, Av. Comandante Maurocélio Rocha Ponte 100, Sobral, CE, CEP 62041-040, Brazil.
| |
Collapse
|
11
|
Vondrakova J, Frolikova M, Ded L, Cerny J, Postlerova P, Palenikova V, Simonik O, Nahacka Z, Basus K, Valaskova E, Machan R, Pacey A, Holubcova Z, Koubek P, Ezrova Z, Park S, Liu R, Partha R, Clark N, Neuzil J, Ikawa M, Erickson K, Lam KS, Moore H, Komrskova K. MAIA, Fc receptor-like 3, supersedes JUNO as IZUMO1 receptor during human fertilization. SCIENCE ADVANCES 2022; 8:eabn0047. [PMID: 36070373 PMCID: PMC9451160 DOI: 10.1126/sciadv.abn0047] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 07/23/2022] [Indexed: 05/17/2023]
Abstract
Gamete fusion is a critical event of mammalian fertilization. A random one-bead one-compound combinatorial peptide library represented synthetic human egg mimics and identified a previously unidentified ligand as Fc receptor-like 3, named MAIA after the mythological goddess intertwined with JUNO. This immunoglobulin super family receptor was expressed on human oolemma and played a major role during sperm-egg adhesion and fusion. MAIA forms a highly stable interaction with the known IZUMO1/JUNO sperm-egg complex, permitting specific gamete fusion. The complexity of the MAIA isotype may offer a cryptic sexual selection mechanism to avoid genetic incompatibility and achieve favorable fitness outcomes.
Collapse
Affiliation(s)
- Jana Vondrakova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Michaela Frolikova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Lukas Ded
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Jiri Cerny
- Laboratory of Structural Bioinformatics of Proteins, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Pavla Postlerova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
- Department of Veterinary Sciences, Faculty of Agrobiology, Food, and Natural Resources, Czech University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic
| | - Veronika Palenikova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Ondrej Simonik
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Zuzana Nahacka
- Laboratory of Molecular Therapy, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Krystof Basus
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Eliska Valaskova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Radek Machan
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Allan Pacey
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Sheffield S10 2RX, UK
| | - Zuzana Holubcova
- Reprofit International, Clinic of Reproductive Medicine, Brno, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Koubek
- ProCrea Swiss IVF Center, Prague, Czech Republic
| | - Zuzana Ezrova
- Laboratory of Molecular Therapy, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Soojin Park
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Raghavendran Partha
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nathan Clark
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Jiri Neuzil
- Laboratory of Molecular Therapy, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
- School of Pharmacy and Medical Science, Griffith University, Parklands Avenue, Southport, Qld 4222, Australia
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kent Erickson
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, CA, USA
| | - Harry Moore
- Centre for Stem Cell Biology, University of Sheffield, Sheffield S10 2TN, UK
| | - Katerina Komrskova
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
- Department of Zoology, Faculty of Science, Charles University, Vinicna 7, 128 44 Prague, Czech Republic
| |
Collapse
|
12
|
Arias-Hidalgo C, Juanes-Velasco P, Landeira-Viñuela A, García-Vaquero ML, Montalvillo E, Góngora R, Hernández ÁP, Fuentes M. Single-Cell Proteomics: The Critical Role of Nanotechnology. Int J Mol Sci 2022; 23:6707. [PMID: 35743151 PMCID: PMC9224324 DOI: 10.3390/ijms23126707] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 11/24/2022] Open
Abstract
In single-cell analysis, biological variability can be attributed to individual cells, their specific state, and the ability to respond to external stimuli, which are determined by protein abundance and their relative alterations. Mass spectrometry (MS)-based proteomics (e.g., SCoPE-MS and SCoPE2) can be used as a non-targeted method to detect molecules across hundreds of individual cells. To achieve high-throughput investigation, novel approaches in Single-Cell Proteomics (SCP) are needed to identify and quantify proteins as accurately as possible. Controlling sample preparation prior to LC-MS analysis is critical, as it influences sensitivity, robustness, and reproducibility. Several nanotechnological approaches have been developed for the removal of cellular debris, salts, and detergents, and to facilitate systematic sample processing at the nano- and microfluidic scale. In addition, nanotechnology has enabled high-throughput proteomics analysis, which have required the improvement of software tools, such as DART-ID or DO-MS, which are also fundamental for addressing key biological questions. Single-cell proteomics has many applications in nanomedicine and biomedical research, including advanced cancer immunotherapies or biomarker characterization, among others; and novel methods allow the quantification of more than a thousand proteins while analyzing hundreds of single cells.
Collapse
Affiliation(s)
- Carlota Arias-Hidalgo
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (C.A.-H.); (P.J.-V.); (A.L.-V.); (M.L.G.-V.); (E.M.); (R.G.)
| | - Pablo Juanes-Velasco
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (C.A.-H.); (P.J.-V.); (A.L.-V.); (M.L.G.-V.); (E.M.); (R.G.)
| | - Alicia Landeira-Viñuela
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (C.A.-H.); (P.J.-V.); (A.L.-V.); (M.L.G.-V.); (E.M.); (R.G.)
| | - Marina L. García-Vaquero
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (C.A.-H.); (P.J.-V.); (A.L.-V.); (M.L.G.-V.); (E.M.); (R.G.)
| | - Enrique Montalvillo
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (C.A.-H.); (P.J.-V.); (A.L.-V.); (M.L.G.-V.); (E.M.); (R.G.)
| | - Rafael Góngora
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (C.A.-H.); (P.J.-V.); (A.L.-V.); (M.L.G.-V.); (E.M.); (R.G.)
| | - Ángela-Patricia Hernández
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (C.A.-H.); (P.J.-V.); (A.L.-V.); (M.L.G.-V.); (E.M.); (R.G.)
- Department of Pharmaceutical Sciences: Organic Chemistry, Faculty of Pharmacy, University of Salamanca, CIETUS, IBSAL, 37007 Salamanca, Spain
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (C.A.-H.); (P.J.-V.); (A.L.-V.); (M.L.G.-V.); (E.M.); (R.G.)
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
13
|
Takeuchi H, Yamamoto M, Fukui M, Inoue A, Maezawa T, Nishioka M, Kondo E, Ikeda T, Matsumoto K, Miyamoto K. Single‐cell profiling of transcriptomic changes during
in vitro
maturation of human oocytes. Reprod Med Biol 2022; 21:e12464. [PMID: 35582522 PMCID: PMC9084694 DOI: 10.1002/rmb2.12464] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose In vitro maturation (IVM) of human oocytes offers an invaluable opportunity for infertility treatment. However, in vitro matured oocytes often show lower developmental abilities than their in vivo counterparts, and molecular mechanisms underlying successful maturation remain unclear. In this study, we investigated gene expression profiles of in vitro matured oocytes at the single‐cell level to gain mechanistic insight into IVM of human oocytes. Methods Human oocytes were retrieved by follicular puncture and in vitro matured. In total, 19 oocytes from 11 patients were collected and subjected to single‐cell RNA‐seq analyses. Results Global gene expression profiles were similar among oocytes at the same maturation stage, while a small number of oocytes showed distinct transcriptomes from those at the corresponding maturation stage. Differential gene expression analysis identified hundreds of transcripts that dynamically altered their expression during IVM, and we revealed molecular pathways and upstream regulators that may govern oocyte maturation. Furthermore, oocytes that were delayed in their maturation showed distinct transcriptomes. Finally, we identified genes whose transcripts were enriched in each stage of oocyte maturation. Conclusions Our work uncovers transcriptomic changes during human oocyte IVM and the differential gene expression profile of each oocyte.
Collapse
Affiliation(s)
- Hiroki Takeuchi
- Department of Obstetrics and Gynecology Graduate School of Medicine Mie University Mie Japan
| | - Mari Yamamoto
- Graduate School of Biology‐Oriented Science and Technology Kindai University Wakayama Japan
| | - Megumi Fukui
- Department of Obstetrics and Gynecology Graduate School of Medicine Mie University Mie Japan
| | - Akihiro Inoue
- Graduate School of Biology‐Oriented Science and Technology Kindai University Wakayama Japan
| | - Tadashi Maezawa
- Department of Obstetrics and Gynecology Graduate School of Medicine Mie University Mie Japan
| | - Mikiko Nishioka
- Department of Obstetrics and Gynecology Graduate School of Medicine Mie University Mie Japan
| | - Eiji Kondo
- Department of Obstetrics and Gynecology Graduate School of Medicine Mie University Mie Japan
| | - Tomoaki Ikeda
- Department of Obstetrics and Gynecology Graduate School of Medicine Mie University Mie Japan
| | - Kazuya Matsumoto
- Graduate School of Biology‐Oriented Science and Technology Kindai University Wakayama Japan
| | - Kei Miyamoto
- Graduate School of Biology‐Oriented Science and Technology Kindai University Wakayama Japan
| |
Collapse
|
14
|
Lombardo SD, Wangsaputra IF, Menche J, Stevens A. Network Approaches for Charting the Transcriptomic and Epigenetic Landscape of the Developmental Origins of Health and Disease. Genes (Basel) 2022; 13:764. [PMID: 35627149 PMCID: PMC9141211 DOI: 10.3390/genes13050764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/04/2022] [Accepted: 04/13/2022] [Indexed: 02/04/2023] Open
Abstract
The early developmental phase is of critical importance for human health and disease later in life. To decipher the molecular mechanisms at play, current biomedical research is increasingly relying on large quantities of diverse omics data. The integration and interpretation of the different datasets pose a critical challenge towards the holistic understanding of the complex biological processes that are involved in early development. In this review, we outline the major transcriptomic and epigenetic processes and the respective datasets that are most relevant for studying the periconceptional period. We cover both basic data processing and analysis steps, as well as more advanced data integration methods. A particular focus is given to network-based methods. Finally, we review the medical applications of such integrative analyses.
Collapse
Affiliation(s)
- Salvo Danilo Lombardo
- Max Perutz Labs, Department of Structural and Computational Biology, University of Vienna, 1030 Vienna, Austria;
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Ivan Fernando Wangsaputra
- Maternal and Fetal Health Research Group, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, UK;
| | - Jörg Menche
- Max Perutz Labs, Department of Structural and Computational Biology, University of Vienna, 1030 Vienna, Austria;
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1030 Vienna, Austria
- Faculty of Mathematics, University of Vienna, 1030 Vienna, Austria
| | - Adam Stevens
- Maternal and Fetal Health Research Group, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, UK;
| |
Collapse
|
15
|
Sciorio R, Miranian D, Smith GD. Non-invasive oocyte quality assessment. Biol Reprod 2022; 106:274-290. [PMID: 35136962 DOI: 10.1093/biolre/ioac009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/04/2022] [Accepted: 01/11/2022] [Indexed: 12/27/2022] Open
Abstract
Oocyte quality is perhaps the most important limiting factor in female fertility; however, the current methods of determining oocyte competence are only marginally capable of predicting a successful pregnancy. We aim to review the predictive value of non-invasive techniques for the assessment of human oocytes and their related cells and biofluids that pertain to their developmental competence. Investigation of the proteome, transcriptome, and hormonal makeup of follicular fluid, as well as cumulus-oocyte complexes are currently underway; however, prospective randomized non-selection-controlled trials of the future are needed before determining their prognostic value. The biological significance of polar body morphology and genetics are still unknown and the subject of debate. The predictive utility of zygotic viscoelasticity for embryo development has been demonstrated, but similar studies performed on oocytes have yet to be conducted. Metabolic profiling of culture media using human oocytes are also limited and may require integration of automated, high-throughput targeted metabolomic assessments in real time with microfluidic platforms. Light exposure to oocytes can be detrimental to subsequent development and utilization of time-lapse imaging and morphometrics of oocytes is wanting. Polarized light, Raman microspectroscopy, and coherent anti-Stokes Raman scattering are a few novel imaging tools that may play a more important role in future oocyte assessment. Ultimately, the integration of chemistry, genomics, microfluidics, microscopy, physics, and other biomedical engineering technologies into the basic studies of oocyte biology, and in testing and perfecting practical solutions of oocyte evaluation, are the future for non-invasive assessment of oocytes.
Collapse
Affiliation(s)
- Romualdo Sciorio
- Edinburgh Assisted Conception Programme, EFREC, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Daniel Miranian
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Gary D Smith
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.,Department of Physiology, Urology, and Reproductive Sciences Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
16
|
Newman H, Catt S, Vining B, Vollenhoven B, Horta F. DNA repair and response to sperm DNA damage in oocytes and embryos, and the potential consequences in ART: a systematic review. Mol Hum Reprod 2021; 28:6483093. [PMID: 34954800 DOI: 10.1093/molehr/gaab071] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Sperm DNA damage is considered a predictive factor for the clinical outcomes of patients undergoing ART. Laboratory evidence suggests that zygotes and developing embryos have adopted specific response and repair mechanisms to repair DNA damage of paternal origin. We have conducted a systematic review in accordance with guidelines from Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) to identify and review the maternal mechanisms used to respond and repair sperm DNA damage during early embryonic development, how these mechanisms operate and their potential clinical implications. The literature search was conducted in Ovid MEDLINE and Embase databases until May 2021. Out of 6297 articles initially identified, 36 studies were found to be relevant through cross referencing and were fully extracted. The collective evidence in human and animal models indicate that the early embryo has the capacity to repair DNA damage within sperm by activating maternally driven mechanisms throughout embryonic development. However, this capacity is limited and likely declines with age. The link between age and decreased DNA repair capacity could explain decreased oocyte quality in older women, poor reproductive outcomes in idiopathic cases, and patients who present high sperm DNA damage. Ultimately, further understanding mechanisms underlying the maternal repair of sperm DNA damage could lead to the development of targeted therapies to decrease sperm DNA damage, improved oocyte quality to combat incoming DNA insults or lead to development of methodologies to identify individual spermatozoa without DNA damage.
Collapse
Affiliation(s)
- H Newman
- Education Program in Reproduction & Development, Department of Obstetrics and Gynecology, Monash University, Melbourne, VIC 3168, Australia
| | - S Catt
- Education Program in Reproduction & Development, Department of Obstetrics and Gynecology, Monash University, Melbourne, VIC 3168, Australia
| | - B Vining
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia.,Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3800, Australia
| | - B Vollenhoven
- Education Program in Reproduction & Development, Department of Obstetrics and Gynecology, Monash University, Melbourne, VIC 3168, Australia.,Monash IVF, Melbourne, VIC, 3168, Australia.,Women's and Newborn Program, Monash Health, VIC, 3169, Australia
| | - F Horta
- Education Program in Reproduction & Development, Department of Obstetrics and Gynecology, Monash University, Melbourne, VIC 3168, Australia.,Monash IVF, Melbourne, VIC, 3168, Australia
| |
Collapse
|
17
|
Wang X, Wang L, Dou J, Yu T, Cao P, Fan N, Borjigin U, Nashun B. Distinct role of histone chaperone Asf1a and Asf1b during fertilization and pre-implantation embryonic development in mice. Epigenetics Chromatin 2021; 14:55. [PMID: 34906203 PMCID: PMC8670131 DOI: 10.1186/s13072-021-00430-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Background Asf1 is a well-conserved histone chaperone that regulates multiple cellular processes in different species. Two paralogous genes, Asf1a and Asf1b exist in mammals, but their role during fertilization and early embryogenesis remains to be investigated further. Methods We analyzed the dynamics of histone chaperone Asf1a and Asf1b in oocytes and pre-implantation embryos in mice by immunofluorescence and real-time quantitative PCR, and further investigated the role of Asf1a and Asf1b during fertilization and pre-implantation development by specific Morpholino oligos-mediated knock down approach. Results Immunofluorescence with specific antibodies revealed that both Asf1a and Asf1b were deposited in the nuclei of fully grown oocytes, accumulated abundantly in zygote and 2-cell embryonic nuclei, but turned low at 4-cell stage embryos. In contrast to the weak but definite nuclear deposition of Asf1a, Asf1b disappeared from embryonic nuclei at morula and blastocyst stages. The knockdown of Asf1a and Asf1b by specific Morpholino oligos revealed that Asf1a but not Asf1b was required for the histone H3.3 assembly in paternal pronucleus. However, knockdown of either Asf1a or Asf1b expression decreased developmental potential of pre-implantation embryos. Furthermore, while Asf1a KD severely reduced H3K56 acetylation level and the expression of Oct4 in blastocyst stage embryos, Asf1b KD almost eliminated nuclear accumulation of proliferating cell marker-PCNA in morula stage embryos. These results suggested that histone chaperone Asf1a and Asf1b play distinct roles during fertilization and pre-implantation development in mice. Conclusions Our data suggested that both Asf1a and Asf1b are required for pre-implantation embryonic development. Asf1a regulates H3K56ac levels and Oct4 expression, while Asf1b safeguards pre-implantation embryo development by regulating cell proliferation. We also showed that Asf1a, but not Asf1b, was necessary for the assembly of histone H3.3 in paternal pronuclei after fertilization. Supplementary Information The online version contains supplementary material available at 10.1186/s13072-021-00430-7.
Collapse
Affiliation(s)
- Xuemei Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Yuquan District, Hohhot, 010070, Inner Mongolia, China
| | - Lu Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Yuquan District, Hohhot, 010070, Inner Mongolia, China
| | - Jie Dou
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Yuquan District, Hohhot, 010070, Inner Mongolia, China
| | - Tianjiao Yu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Yuquan District, Hohhot, 010070, Inner Mongolia, China
| | - Pengbo Cao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Yuquan District, Hohhot, 010070, Inner Mongolia, China
| | - Na Fan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Yuquan District, Hohhot, 010070, Inner Mongolia, China
| | - Uyunbilig Borjigin
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Yuquan District, Hohhot, 010070, Inner Mongolia, China
| | - Buhe Nashun
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Yuquan District, Hohhot, 010070, Inner Mongolia, China.
| |
Collapse
|
18
|
Horta F, Ravichandran A, Catt S, Vollenhoven B, Temple-Smith P. Ageing and ovarian stimulation modulate the relative levels of transcript abundance of oocyte DNA repair genes during the germinal vesicle-metaphase II transition in mice. J Assist Reprod Genet 2021; 38:55-69. [PMID: 33067741 PMCID: PMC7822980 DOI: 10.1007/s10815-020-01981-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/13/2020] [Indexed: 10/23/2022] Open
Abstract
PURPOSE Oocyte quality and reproductive outcome are negatively affected by advanced maternal age, ovarian stimulation and method of oocyte maturation during assisted reproduction; however, the mechanisms responsible for these associations are not fully understood. The aim of this study was to compare the effects of ageing, ovarian stimulation and in-vitro maturation on the relative levels of transcript abundance of genes associated with DNA repair during the transition of germinal vesicle (GV) to metaphase II (MII) stages of oocyte development. METHODS The relative levels of transcript abundance of 90 DNA repair-associated genes was compared in GV-stage and MII-stage oocytes from unstimulated and hormone-stimulated ovaries from young (5-8-week-old) and old (42-45-week-old) C57BL6 mice. Ovarian stimulation was conducted using pregnant mare serum gonadotropin (PMSG) or anti-inhibin serum (AIS). DNA damage response was quantified by immunolabeling of the phosphorylated histone variant H2AX (γH2AX). RESULTS The relative transcript abundance in DNA repair genes was significantly lower in MII oocytes compared to GV oocytes in young unstimulated and PMSG stimulated but was higher in AIS-stimulated mice. Interestingly, an increase in the relative level of transcript abundance of DNA repair genes was observed in MII oocytes from older mice in unstimulated, PMSG-stimulated and AIS-stimulated mice. Decreased γH2AX levels were found in both GV oocytes (82.9%) and MII oocytes (37.5%) during ageing in both ovarian stimulation types used (PMSG/AIS; p < 0.05). CONCLUSIONS In conclusion, DNA repair relative levels of transcript abundance are altered by maternal age and the method of ovarian stimulation during the GV-MII transition in oocytes.
Collapse
Affiliation(s)
- Fabrizzio Horta
- Education Program in Reproduction and Development, EPRD, Department of Obstetrics and Gynecology, School of Clinical Science, Monash University, Melbourne, VIC, 3168, Australia.
- Monash IVF, Melbourne, VIC, 3168, Australia.
| | - Aravind Ravichandran
- Education Program in Reproduction and Development, EPRD, Department of Obstetrics and Gynecology, School of Clinical Science, Monash University, Melbourne, VIC, 3168, Australia
| | - Sally Catt
- Education Program in Reproduction and Development, EPRD, Department of Obstetrics and Gynecology, School of Clinical Science, Monash University, Melbourne, VIC, 3168, Australia
| | - Beverley Vollenhoven
- Education Program in Reproduction and Development, EPRD, Department of Obstetrics and Gynecology, School of Clinical Science, Monash University, Melbourne, VIC, 3168, Australia
- Monash IVF, Melbourne, VIC, 3168, Australia
- Women's and Newborn Program, Monash Health, Melbourne, Australia
| | - Peter Temple-Smith
- Education Program in Reproduction and Development, EPRD, Department of Obstetrics and Gynecology, School of Clinical Science, Monash University, Melbourne, VIC, 3168, Australia
| |
Collapse
|
19
|
Naim N, Amrit FRG, McClendon TB, Yanowitz JL, Ghazi A. The molecular tug of war between immunity and fertility: Emergence of conserved signaling pathways and regulatory mechanisms. Bioessays 2020; 42:e2000103. [PMID: 33169418 DOI: 10.1002/bies.202000103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022]
Abstract
Reproduction and immunity are energy intensive, intimately linked processes in most organisms. In women, pregnancy is associated with widespread immunological adaptations that alter immunity to many diseases, whereas, immune dysfunction has emerged as a major cause for infertility in both men and women. Deciphering the molecular bases of this dynamic association is inherently challenging in mammals. This relationship has been traditionally studied in fast-living, invertebrate species, often in the context of resource allocation between life history traits. More recently, these studies have advanced our understanding of the mechanistic underpinnings of the immunity-fertility dialogue. Here, we review the molecular connections between reproduction and immunity from the perspective of human pregnancy to mechanistic discoveries in laboratory organisms. We focus particularly on recent invertebrate studies identifying conserved signaling pathways and transcription factors that regulate resource allocation and shape the balance between reproductive status and immune health.
Collapse
Affiliation(s)
- Nikki Naim
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Francis R G Amrit
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - T Brooke McClendon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Judith L Yanowitz
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Arjumand Ghazi
- Departments of Pediatrics, Developmental Biology and Cell Biology and Physiology, John, G. Rangos Sr. Research Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
Male Factors: the Role of Sperm in Preimplantation Embryo Quality. Reprod Sci 2020; 28:1788-1811. [DOI: 10.1007/s43032-020-00334-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
|
21
|
Kay VR, Rätsep MT, Figueiró-Filho EA, Croy BA. Preeclampsia may influence offspring neuroanatomy and cognitive function: a role for placental growth factor†. Biol Reprod 2020; 101:271-283. [PMID: 31175349 DOI: 10.1093/biolre/ioz095] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/30/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023] Open
Abstract
Preeclampsia (PE) is a common pregnancy complication affecting 3-5% of women. Preeclampsia is diagnosed clinically as new-onset hypertension with associated end organ damage after 20 weeks of gestation. Despite being diagnosed as a maternal syndrome, fetal experience of PE is a developmental insult with lifelong cognitive consequences. These cognitive alterations are associated with distorted neuroanatomy and cerebrovasculature, including a higher risk of stroke. The pathophysiology of a PE pregnancy is complex, with many factors potentially able to affect fetal development. Deficient pro-angiogenic factor expression is one aspect that may impair fetal vascularization, alter brain structure, and affect future cognition. Of the pro-angiogenic growth factors, placental growth factor (PGF) is strongly linked to PE. Concentrations of PGF are inappropriately low in maternal blood both before and during a PE gestation. Fetal concentrations of PGF appear to mirror maternal circulating concentrations. Using Pgf-/- mice that may model effects of PE on offspring, we demonstrated altered central nervous system vascularization, neuroanatomy, and behavior. Overall, we propose that development of the fetal brain is impaired in PE, making the offspring of preeclamptic pregnancies a unique cohort with greater risk of altered cognition and cerebrovasculature. These individuals may benefit from early interventions, either pharmacological or environmental. The early neonatal period may be a promising window for intervention while the developing brain retains plasticity.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Matthew T Rätsep
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
22
|
Tetkova A, Susor A, Kubelka M, Nemcova L, Jansova D, Dvoran M, Del Llano E, Holubcova Z, Kalous J. Follicle-stimulating hormone administration affects amino acid metabolism in mammalian oocytes†. Biol Reprod 2020; 101:719-732. [PMID: 31290535 DOI: 10.1093/biolre/ioz117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/18/2019] [Accepted: 07/04/2019] [Indexed: 12/27/2022] Open
Abstract
Culture media used in assisted reproduction are commonly supplemented with gonadotropin hormones to support the nuclear and cytoplasmic maturation of in vitro matured oocytes. However, the effect of gonadotropins on protein synthesis in oocytes is yet to be fully understood. As published data have previously documented a positive in vitro effect of follicle-stimulating hormone (FSH) on cytoplasmic maturation, we exposed mouse denuded oocytes to FSH in order to evaluate the changes in global protein synthesis. We found that dose-dependent administration of FSH resulted in a decrease of methionine incorporation into de novo synthesized proteins in denuded mouse oocytes and oocytes cultured in cumulus-oocyte complexes. Similarly, FSH influenced methionine incorporation in additional mammalian species including human. Furthermore, we showed the expression of FSH-receptor protein in oocytes. We found that major translational regulators were not affected by FSH treatment; however, the amino acid uptake became impaired. We propose that the effect of FSH treatment on amino acid uptake is influenced by FSH receptor with the effect on oocyte metabolism and physiology.
Collapse
Affiliation(s)
- Anna Tetkova
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic.,Department of Cell Biology, Faculty of Science, Charles University in Prague, Prague 2, Czech Republic
| | - Andrej Susor
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic
| | - Michal Kubelka
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic
| | - Lucie Nemcova
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic
| | - Denisa Jansova
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic
| | - Michal Dvoran
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic.,Department of Cell Biology, Faculty of Science, Charles University in Prague, Prague 2, Czech Republic
| | - Edgar Del Llano
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic.,Department of Cell Biology, Faculty of Science, Charles University in Prague, Prague 2, Czech Republic
| | - Zuzana Holubcova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Reprofit International, Clinic of Reproductive Medicine, Brno, Czech Republic
| | - Jaroslav Kalous
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic
| |
Collapse
|
23
|
The Impact of DNA Methylation Dynamics on the Mutation Rate During Human Germline Development. G3-GENES GENOMES GENETICS 2020; 10:3337-3346. [PMID: 32727923 PMCID: PMC7466984 DOI: 10.1534/g3.120.401511] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
DNA methylation is a dynamic epigenetic modification found in most eukaryotic genomes. It is known to lead to a high CpG to TpG mutation rate. However, the relationship between the methylation dynamics in germline development and the germline mutation rate remains unexplored. In this study, we used whole genome bisulfite sequencing (WGBS) data of cells at 13 stages of human germline development and rare variants from the 1000 Genome Project as proxies for germline mutations to investigate the correlation between dynamic methylation levels and germline mutation rates at different scales. At the single-site level, we found a significant correlation between methylation and the germline point mutation rate at CpG sites during germline developmental stages. Then we explored the mutability of methylation dynamics in all stages. Our results also showed a broad correlation between the regional methylation level and the rate of C > T mutation at CpG sites in all genomic regions, especially in intronic regions; a similar link was also seen at all chromosomal levels. Our findings indicate that the dynamic DNA methylome during human germline development has a broader mutational impact than is commonly assumed.
Collapse
|
24
|
Analysis of Menstrual Blood Stromal Cells Reveals SOX15 Triggers Oocyte-Based Human Cell Reprogramming. iScience 2020; 23:101376. [PMID: 32738616 PMCID: PMC7394916 DOI: 10.1016/j.isci.2020.101376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 05/26/2020] [Accepted: 07/13/2020] [Indexed: 12/14/2022] Open
Abstract
Cell reprogramming has revolutionized cell and regenerative biology field. However, human iPS derivation remains inefficient and variable. A better knowledge of molecular processes and the rationale underlying the importance of somatic cell origin is crucial to uncover reprogramming mechanisms. Here, we analyze the molecular profile of different human somatic cell types. We show menstrual blood-derived stromal cells (MnSCs) have a distinct, reprogramming prone, profile, and we identify SOX15 from their oocyte-related signature as a prominent responsible candidate. SOX15 orchestrates an efficient oocyte-based reprogramming combination when overexpressed with the also oocyte-enriched histone chaperone ASF1A and OCT4 and, through specific mechanism, generates iPSCs with distinguishable pluripotent state that further present higher differentiation capacity than canonical iPSCs. Our work supports the presence of different pluripotency states in reprogramming and the importance of using metaphase-II oocyte and MnSCs information to provide alternative reprogramming combinations and, importantly, to improve and understand pluripotency acquisition. MnSC expression signature reveals SOX15 as a crucial oocyte-enriched reprogramming factor SOX15 orchestrates an efficient oocyte-based reprogramming combination in MnSC Oocyte-based reprogrammed iPSCs (AOX15) show distinct pluripotent state AOX15 iPSCs present higher differentiation capacity than OSKM-iPSCs
Collapse
|
25
|
Li J, Bai L, Liu Z, Wang W. Dual roles of PDE9a in meiotic maturation of zebrafish oocytes. Biochem Biophys Res Commun 2020; 532:40-46. [PMID: 32826058 DOI: 10.1016/j.bbrc.2020.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 01/11/2023]
Abstract
The essential role of cyclic guanosine monophosphate (cGMP) signaling in regulating the oocyte meiotic cell cycle has been established. However, control of the level of cGMP in ovarian follicles is unclear. The cGMP-hydrolyzing phosphodiesterases (PDEs) are important in regulating the cellular cGMP level. We used zebrafish as a model to study the role of a cGMP-hydrolyzing phosphodiesterase-9a (PDE9a) in meiotic maturation of oocytes. Three PDE9a coding genes (PDE9aa, PDE9ab, and PDE9ac) were identified in zebrafish. Both pde9aa and pde9ac are expressed in most adult tissues including the ovary, but pde9ab is only expressed in the ovary, kidney, pituitary, and brain. All three pde9as mRNA exhibited different expression profiles during folliculogenesis. All of them are highly expressed in the oocyte but not in the follicular cell. The expression of both pde9aa and pde9ab, but not pde9ac, in ovarian follicles increases during oocyte maturation either in natural ovulatory cycle or induced by administration of hCG in vivo. We overexpressed pde9aa by injection of capped pde9aa mRNA into the oocytes. The cGMP level was decreased, and oocyte maturation was stimulated. When the activity of PDE9a was blocked by a specific inhibitor, Bay736691, the oocyte maturation was also stimulated. The stimulatory effect could be blocked by a gap junction blocker. However, the spontaneous oocyte maturation of denuded oocytes was not largely affected after treatment with Bay736691. All of the mature oocytes obtained by either treatment of Bay736691 or injection of pde9aa mRNA, could be fertilized in vitro. These results demonstrate the dual roles of PDE9a in oocyte maturation. The basal level of PDE9a is responsible for maintaining the meiotic arrest, and the increased level of PDE9a induced by LH signaling is helpful for stimulating meiotic maturation by hydrolyzing cGMP in oocytes.
Collapse
Affiliation(s)
- Jianzhen Li
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China.
| | - Lin Bai
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Zhiquan Liu
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China
| | - Wenyi Wang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, 730070, China
| |
Collapse
|
26
|
Zhao ZH, Meng TG, Li A, Schatten H, Wang ZB, Sun QY. RNA-Seq transcriptome reveals different molecular responses during human and mouse oocyte maturation and fertilization. BMC Genomics 2020; 21:475. [PMID: 32650721 PMCID: PMC7350670 DOI: 10.1186/s12864-020-06885-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/06/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Female infertility is a worldwide concern and the etiology of infertility has not been thoroughly demonstrated. Although the mouse is a good model system to perform functional studies, the differences between mouse and human also need to be considered. The objective of this study is to elucidate the different molecular mechanisms underlying oocyte maturation and fertilization between human and mouse. RESULTS A comparative transcriptome analysis was performed to identify the differentially expressed genes and associated biological processes between human and mouse oocytes. In total, 8513 common genes, as well as 15,165 and 6126 uniquely expressed genes were detected in human and mouse MII oocytes, respectively. Additionally, the ratios of non-homologous genes in human and mouse MII oocytes were 37 and 8%, respectively. Functional categorization analysis of the human MII non-homologous genes revealed that cAMP-mediated signaling, sister chromatid cohesin, and cell recognition were the major enriched biological processes. Interestingly, we couldn't detect any GO categories in mouse non-homologous genes. CONCLUSIONS This study demonstrates that human and mouse oocytes exhibit significant differences in gene expression profiles during oocyte maturation, which probably deciphers the differential molecular responses to oocyte maturation and fertilization. The significant differences between human and mouse oocytes limit the generalizations from mouse to human oocyte maturation. Knowledge about the limitations of animal models is crucial when exploring a complex process such as human oocyte maturation and fertilization.
Collapse
Affiliation(s)
- Zheng-Hui Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Tie-Gang Meng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Ang Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China. .,University of Chinese Academy of Sciences, Beijing, China.
| | - Qing-Yuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China. .,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
| |
Collapse
|
27
|
Can H, Chanumolu SK, Gonzalez-Muñoz E, Prukudom S, Otu HH, Cibelli JB. Comparative analysis of single-cell transcriptomics in human and Zebrafish oocytes. BMC Genomics 2020; 21:471. [PMID: 32640983 PMCID: PMC7346435 DOI: 10.1186/s12864-020-06860-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
Background Zebrafish is a popular model organism, which is widely used in developmental biology research. Despite its general use, the direct comparison of the zebrafish and human oocyte transcriptomes has not been well studied. It is significant to see if the similarity observed between the two organisms at the gene sequence level is also observed at the expression level in key cell types such as the oocyte. Results We performed single-cell RNA-seq of the zebrafish oocyte and compared it with two studies that have performed single-cell RNA-seq of the human oocyte. We carried out a comparative analysis of genes expressed in the oocyte and genes highly expressed in the oocyte across the three studies. Overall, we found high consistency between the human studies and high concordance in expression for the orthologous genes in the two organisms. According to the Ensembl database, about 60% of the human protein coding genes are orthologous to the zebrafish genes. Our results showed that a higher percentage of the genes that are highly expressed in both organisms show orthology compared to the lower expressed genes. Systems biology analysis of the genes highly expressed in the three studies showed significant overlap of the enriched pathways and GO terms. Moreover, orthologous genes that are commonly overexpressed in both organisms were involved in biological mechanisms that are functionally essential to the oocyte. Conclusions Orthologous genes are concurrently highly expressed in the oocytes of the two organisms and these genes belong to similar functional categories. Our results provide evidence that zebrafish could serve as a valid model organism to study the oocyte with direct implications in human.
Collapse
Affiliation(s)
- Handan Can
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Sree K Chanumolu
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Elena Gonzalez-Muñoz
- LARCEL, Andalusian Laboratory of Cell Reprogramming (LARCel), Andalusian Center for Nanomedicine and Biotechnology-BIONAND, 29590, Málaga, Spain.,Department of Cell Biology, Genetics and Physiology, University of Málaga and; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBNE), 29071, Málaga, Spain
| | - Sukumal Prukudom
- Department of Anatomy, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900, Thailand
| | - Hasan H Otu
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA.
| | - Jose B Cibelli
- Departments of Animal Science and Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
28
|
Martin JH, Aitken RJ, Bromfield EG, Nixon B. DNA damage and repair in the female germline: contributions to ART. Hum Reprod Update 2020; 25:180-201. [PMID: 30541031 DOI: 10.1093/humupd/dmy040] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/27/2018] [Accepted: 11/06/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND DNA integrity and stability are critical determinants of cell viability. This is especially true in the female germline, wherein DNA integrity underpins successful conception, embryonic development, pregnancy and the production of healthy offspring. However, DNA is not inert; rather, it is subject to assault from various environment factors resulting in chemical modification and/or strand breakage. If structural alterations result and are left unrepaired, they have the potential to cause mutations and propagate disease. In this regard, reduced genetic integrity of the female germline ranks among the leading causes of subfertility in humans. With an estimated 10% of couples in developed countries taking recourse to ART to achieve pregnancy, the need for ongoing research into the capacity of the oocyte to detect DNA damage and thereafter initiate cell cycle arrest, apoptosis or DNA repair is increasingly more pressing. OBJECTIVE AND RATIONALE This review documents our current knowledge of the quality control mechanisms utilised by the female germline to prevent and remediate DNA damage during their development from primordial follicles through to the formation of preimplantation embryos. SEARCH METHODS The PubMed database was searched using the keywords: primordial follicle, primary follicle, secondary follicle, tertiary follicle, germinal vesical, MI, MII oocyte, zygote, preimplantation embryo, DNA repair, double-strand break and DNA damage. These keywords were combined with other phrases relevant to the topic. Literature was restricted to peer-reviewed original articles in the English language (published 1979-2018) and references within these articles were also searched. OUTCOMES In this review, we explore the quality control mechanisms utilised by the female germline to prevent, detect and remediate DNA damage. We follow the trajectory of development from the primordial follicle stage through to the preimplantation embryo, highlighting findings likely to have important implications for fertility management, age-related subfertility and premature ovarian failure. In addition, we survey the latest discoveries regarding DNA repair within the metaphase II (MII) oocyte and implicate maternal stores of endogenous DNA repair proteins and mRNA transcripts as a primary means by which they defend their genomic integrity. The collective evidence reviewed herein demonstrates that the MII oocyte can engage in the activation of major DNA damage repair pathway(s), therefore encouraging a reappraisal of the long-held paradigm that oocytes are largely refractory to DNA repair upon reaching this late stage of their development. It is also demonstrated that the zygote can exploit a number of protective strategies to mitigate the risk and/or effect the repair, of DNA damage sustained to either parental germline; affirming that DNA protection is largely a maternally driven trait but that some aspects of repair may rely on a collaborative effort between the male and female germlines. WIDER IMPLICATIONS The present review highlights the vulnerability of the oocyte to DNA damage and presents a number of opportunities for research to bolster the stringency of the oocyte's endogenous defences, with implications extending to improved diagnostics and novel therapeutic applications to alleviate the burden of infertility.
Collapse
Affiliation(s)
- Jacinta H Martin
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, Discipline of Biological Sciences, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Kookaburra Circuit, New Lambton Heights, NSW, Australia
| |
Collapse
|
29
|
A Comparative Analysis of Oocyte Development in Mammals. Cells 2020; 9:cells9041002. [PMID: 32316494 PMCID: PMC7226043 DOI: 10.3390/cells9041002] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022] Open
Abstract
Sexual reproduction requires the fertilization of a female gamete after it has undergone optimal development. Various aspects of oocyte development and many molecular actors in this process are shared among mammals, but phylogeny and experimental data reveal species specificities. In this chapter, we will present these common and distinctive features with a focus on three points: the shaping of the oocyte transcriptome from evolutionarily conserved and rapidly evolving genes, the control of folliculogenesis and ovulation rate by oocyte-secreted Growth and Differentiation Factor 9 and Bone Morphogenetic Protein 15, and the importance of lipid metabolism.
Collapse
|
30
|
Deng K, Feng W, Liu X, Su X, Zuo E, Du S, Huang Y, Shi D, Lu F. Anti-silencing factor 1A is associated with genome stability maintenance of mouse preimplantation embryos†. Biol Reprod 2020; 102:817-827. [PMID: 31916576 DOI: 10.1093/biolre/ioaa001] [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: 05/28/2019] [Revised: 10/07/2019] [Accepted: 01/03/2020] [Indexed: 12/26/2022] Open
Abstract
Genome stability is critical for the normal development of preimplantation embryos, as DNA damages may result in mutation and even embryo lethality. Anti-silencing factor 1A (ASF1A) is a histone chaperone and enriched in the MII oocytes as a maternal factor, which may be associated with the maintenance of genome stability. Thus, this study was undertaken to explore the role of ASF1A in maintaining the genome stability of early mouse embryos. The ASF1A expressed in the preimplantation embryos and displayed a dynamic pattern throughout the early embryonic development. Inhibition of ASF1A expression decreased embryonic development and increased DNA damages. Overexpression of ASF1A improved the developmental potential and decreased DNA damages. When 293T cells that had been integrated with RGS-NHEJ were co-transfected with plasmids of pcDNA3.1-ASF1A, gRNA-NHEJ, and hCas9, less cells expressed eGFP, indicating that non-homologous end joining was reduced by ASF1A. When 293T cells were co-transfected with plasmids of HR-donor, gRNA-HR, hCas9, and pcDNA3.1-ASF1A, more cells expressed eGFP, indicating that homologous recombination (HR) was enhanced by ASF1A. These results indicate that ASF1A may be associated with the genome stability maintenance of early mouse embryos and this action may be mediated by promoting DNA damage repair through HR pathway.
Collapse
Affiliation(s)
- Kai Deng
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China and
| | - Wanyou Feng
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China and
| | - Xiaohua Liu
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China and
| | - Xiaoping Su
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China and
| | - Erwei Zuo
- Center for Animal Genomics, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Shanshan Du
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China and
| | - Yongjun Huang
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China and
| | - Deshun Shi
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China and
| | - Fenghua Lu
- Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning, China and
| |
Collapse
|
31
|
Luteinizing Hormone Action in Human Oocyte Maturation and Quality: Signaling Pathways, Regulation, and Clinical Impact. Reprod Sci 2020; 27:1223-1252. [PMID: 32046451 PMCID: PMC7190682 DOI: 10.1007/s43032-019-00137-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/14/2019] [Indexed: 12/18/2022]
Abstract
The ovarian follicle luteinizing hormone (LH) signaling molecules that regulate oocyte meiotic maturation have recently been identified. The LH signal reduces preovulatory follicle cyclic nucleotide levels which releases oocytes from the first meiotic arrest. In the ovarian follicle, the LH signal reduces cyclic nucleotide levels via the CNP/NPR2 system, the EGF/EGF receptor network, and follicle/oocyte gap junctions. In the oocyte, reduced cyclic nucleotide levels activate the maturation promoting factor (MPF). The activated MPF induces chromosome segregation and completion of the first and second meiotic divisions. The purpose of this paper is to present an overview of the current understanding of human LH signaling regulation of oocyte meiotic maturation by identifying and integrating the human studies on this topic. We found 89 human studies in the literature that identified 24 LH follicle/oocyte signaling proteins. These studies show that human oocyte meiotic maturation is regulated by the same proteins that regulate animal oocyte meiotic maturation. We also found that these LH signaling pathway molecules regulate human oocyte quality and subsequent embryo quality. Remarkably, in vitro maturation (IVM) prematuration culture (PMC) protocols that manipulate the LH signaling pathway improve human oocyte quality of cultured human oocytes. This knowledge has improved clinical human IVM efficiency which may become a routine alternative ART for some infertile patients.
Collapse
|
32
|
Gad A, Nemcova L, Murin M, Kinterova V, Kanka J, Laurincik J, Benc M, Pendovski L, Prochazka R. Global transcriptome analysis of porcine oocytes in correlation with follicle size. Mol Reprod Dev 2019; 87:102-114. [PMID: 31736195 DOI: 10.1002/mrd.23294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/30/2019] [Indexed: 11/09/2022]
Abstract
Although our knowledge regarding oocyte quality and development has improved significantly, the molecular mechanisms that regulate and determine oocyte developmental competence are still unclear. Therefore, the objective of this study was to identify and analyze the transcriptome profiles of porcine oocytes derived from large or small follicles using RNA high-throughput sequencing technology. RNA libraries were constructed from oocytes of large (LO; 3-6 mm) or small (SO; 1.5-1.9 mm) ovarian follicles and then sequenced in an Illumina HiSeq4000. Transcriptome analysis showed a total of 14,557 genes were commonly detected in both oocyte groups. Genes related to the cell cycle, oocyte meiosis, and quality were among the top highly expressed genes in both groups. Differential expression analysis revealed 60 up- and 262 downregulated genes in the LO compared with the SO group. BRCA2, GPLD1, ZP3, ND3, and ND4L were among the highly abundant and highly significant differentially expressed genes (DEGs). The ontological classification of DEGs indicated that protein processing in endoplasmic reticulum was the top enriched pathway. In addition, biological processes related to cell growth and signaling, gene expression regulations, cytoskeleton, and extracellular matrix organization were among the highly enriched processes. In conclusion, this study provides new insights into the global transcriptome changes and the abundance of specific transcripts in porcine oocytes in correlation with follicle size.
Collapse
Affiliation(s)
- Ahmed Gad
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czech Republic.,Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Lucie Nemcova
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czech Republic
| | - Matej Murin
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czech Republic
| | - Veronika Kinterova
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czech Republic
| | - Jiri Kanka
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czech Republic
| | - Jozef Laurincik
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czech Republic.,Department of Zoology and Anthropology, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, Slovak Republic
| | - Michal Benc
- Department of Zoology and Anthropology, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, Slovak Republic.,Biology of Reproduction Department, Institute of Animal Science, Prague, Uhrineves, Czech Republic
| | - Lazo Pendovski
- Department of Functional Morphology, Faculty of Veterinary Medicine, Ss. Cyril and Methodius University in Skopje, Skopje, Republic of Macedonia
| | - Radek Prochazka
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Liběchov, Czech Republic
| |
Collapse
|
33
|
Moura MT, Badaraco J, Sousa RV, Lucci CM, Rumpf R. Improved functional oocyte enucleation by actinomycin D for bovine somatic cell nuclear transfer. Reprod Fertil Dev 2019; 31:1321-1329. [PMID: 30986366 DOI: 10.1071/rd18164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 02/05/2019] [Indexed: 11/23/2022] Open
Abstract
Somatic cell nuclear transfer (SCNT) allows animal cloning but remains technically challenging. This study investigated limitations to functional oocyte enucleation by actinomycin D (AD) as a means of making SCNT easier to perform. Denuding oocytes or inhibiting transcription before AD treatment revealed that the toxicity of this compound during bovine oocyte maturation is mediated by cumulus cells. Exposure of denuded oocytes to higher concentrations of AD (5-20μgmL-1 ) and stepwise reductions of the incubation period (from 14.0 to 0.25h) led to complete inhibition of parthenogenetic development. Bovine SCNT using this improved AD enucleation protocol (NT(AD)) restored cleavage rates compared with rates in the parthenogenetic and SCNT controls (P(CTL) and NT(CTL) respectively). However, NT(AD) was associated with increased caspase-3 activity in cleavage stage embryos and did not recover blastocyst rates. The removal of AD-treated oocyte spindle before reconstruction (NT(AD+SR)) improved embryo development and reduced caspase-3 activity to levels similar to those in the P(CTL) and NT(CTL) groups. Furthermore, mid-term pregnancies were achieved using NT(AD+SR) blastocysts. In conclusion, improvements in AD functional enucleation for bovine SCNT circumvents most cellular roadblocks to early embryonic development and future investigations must focus on restoring blastocyst formation.
Collapse
Affiliation(s)
- Marcelo T Moura
- Laboratório de Reprodução Animal, Embrapa Recursos Genéticos e Biotecnologia, Parque Estação Biológica, Av. W5 Norte (final), CP 02372, CEP 70770-900, Brasília, DF, Brazil; and Departamento de Agronomia e Medicina Veterinária, Universidade de Brasília, Instituto Central de Ciências Sul, Campus Universitário Darci Ribeiro, CEP 70297-400, Brasília, DF, Brazil; and Present address: Laboratório de Biologia Celular, Universidade Federal de São Paulo, Campus Diadema, CEP 09972-270, Diadema, SP, Brazil; and Corresponding author
| | - Jeferson Badaraco
- Laboratório de Reprodução Animal, Embrapa Recursos Genéticos e Biotecnologia, Parque Estação Biológica, Av. W5 Norte (final), CP 02372, CEP 70770-900, Brasília, DF, Brazil
| | - Regivaldo V Sousa
- Laboratório de Reprodução Animal, Embrapa Recursos Genéticos e Biotecnologia, Parque Estação Biológica, Av. W5 Norte (final), CP 02372, CEP 70770-900, Brasília, DF, Brazil
| | - Carolina M Lucci
- Departamento de Agronomia e Medicina Veterinária, Universidade de Brasília, Instituto Central de Ciências Sul, Campus Universitário Darci Ribeiro, CEP 70297-400, Brasília, DF, Brazil
| | - Rodolfo Rumpf
- Laboratório de Reprodução Animal, Embrapa Recursos Genéticos e Biotecnologia, Parque Estação Biológica, Av. W5 Norte (final), CP 02372, CEP 70770-900, Brasília, DF, Brazil; and Departamento de Agronomia e Medicina Veterinária, Universidade de Brasília, Instituto Central de Ciências Sul, Campus Universitário Darci Ribeiro, CEP 70297-400, Brasília, DF, Brazil; and Present address: Geneal Biotecnologia, Rodovia BR-050, Km 184, CEP 38038-050, Uberaba, MG, Brazil
| |
Collapse
|
34
|
Abstract
Proteins and RNA molecules are deposited into the developing egg by the mother. These gene products will drive the first stages of development and are coded by maternal genes. Maternal genes are essential, yet, despite their importance, their evolutionary dynamics is largely unknown. Here I review the current knowledge of maternal gene evolution. The evolutionary origin of maternal genes tends to be more recent than that of zygotic genes. Some studies support the theoretical prediction that maternal genes evolve faster than zygotic genes. However, most studies were done on a limited set of species and genes. I also discuss the way forward to understand the evolution of maternal genes by combining high-throughput genomics and theoretical evolutionary approaches.
Collapse
|
35
|
Adult Pgf -/- mice behaviour and neuroanatomy are altered by neonatal treatment with recombinant placental growth factor. Sci Rep 2019; 9:9285. [PMID: 31243296 PMCID: PMC6594955 DOI: 10.1038/s41598-019-45824-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 06/12/2019] [Indexed: 12/20/2022] Open
Abstract
Offspring of preeclamptic pregnancies have cognitive alterations. Placental growth factor (PGF), is low in preeclampsia; reduced levels may affect brain development. PGF-null mice differ from normal congenic controls in cerebrovasculature, neuroanatomy and behavior. Using brain imaging and behavioral testing, we asked whether developmentally asynchronous (i.e. neonatal) PGF supplementation alters the vascular, neuroanatomic and/or behavioral status of Pgf−/− mice at adulthood. C57BL/6-Pgf−/− pups were treated intraperitoneally on postnatal days 1–10 with vehicle or PGF at 10 pg/g, 70 pg/g or 700 pg/g. These mice underwent behavioral testing and perfusion for MRI and analysis of retinal vasculature. A second cohort of vehicle- or PGF-treated mice was perfused for micro-CT imaging. 10 pg/g PGF-treated mice exhibited less locomotor activity and greater anxiety-like behavior relative to vehicle-treated mice. Depressive-like behavior showed a sex-specific, dose-dependent decrease and was lowest in 700 pg/g PGF-treated females relative to vehicle-treated females. Spatial learning did not differ. MRI revealed smaller volume of three structures in the 10 pg/g group, larger volume of seven structures in the 70 pg/g group and smaller volume of one structure in the 700 pg/g group. No cerebral or retinal vascular differences were detected. Overall, neonatal PGF replacement altered behavior and neuroanatomy of adult Pgf−/− mice.
Collapse
|
36
|
Hamad MF. Quantification of histones and protamines mRNA transcripts in sperms of infertile couples and their impact on sperm's quality and chromatin integrity. Reprod Biol 2019; 19:6-13. [PMID: 30876814 DOI: 10.1016/j.repbio.2019.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/17/2019] [Accepted: 03/02/2019] [Indexed: 11/16/2022]
Abstract
The proper transition of histones to protamine during spermiogenesis is critical for male fertility. This study aimed to quantify the levels of histones and protamines mRNA in sperms of infertile couples and their possible effect on the sperm's quality and chromatin integrity. Spermatozoa from 53 normal and 75 patients were enrolled in this study. Histones and Protamine mRNAs were extracted, reverse-transcribed and applied to real-time quantitative PCR. Chromomycin A3 staining was used to assess protamination and chromatin integrity, and Eosin-Nigrosine-Test and HOS-Test was used to evaluate the sperm's vitality and membrane integrity respectively. Levels of histones H2A and H2B mRNA in patient's sperms were significantly (p < 0.01) higher than that of normal (31.22 ± 2.91, 30.03 ± 2.05 vs. 25.62 ± 1.98, 27.23 ± 3.04, respectively). Protamine PRM2 mRNA in patient's sperms (20.55 ± 2.01) was significantly lower than in normal (21.73 ± 2.64, p < 0.01). The PRM1/PRM2 and H2A/H2B mRNAs ratios were significantly higher (p < 0.01) in patients than normal (1.02 ± 0.10, 1.04 ± 0.07 vs. 0.98 ± 0.06, 0.94 ± 0.08 respectively). Also, the sperm's nuclear histones to protamines transcripts ((H2A + H2B)/(PRM1 + PRM2)) ratios of patients (1.49 ± 0.16) was significantly higher (p < 0.01) than that of normal (1.25 ± 0.15). Histone/protamine transcripts [((H2A + H2B)/(PRM1 + PRM2)) mRNAs ratios] were negatively correlated (p < 0.05) with sperm's count, total count, motility, progressive motility, normal morphology, membrane integrity and positively with chromatin decondensation. The data suggests that histones/protamines mRNAs ratios are important for a sperm's quality and therefore could be used as predictors for male infertility. Also, validation study may be required to confirm the study conclusion.
Collapse
Affiliation(s)
- Mohammed Faiz Hamad
- Department of Basic Sciences, College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Riyadh, Saudi Arabia; IVF & Andrology Laboratory, Department of Obstetrics and Gynecology, Saarland University, Homburg, Saar, Germany.
| |
Collapse
|
37
|
Melo TP, Fortes MRS, Bresolin T, Mota LFM, Albuquerque LG, Carvalheiro R. Multitrait meta-analysis identified genomic regions associated with sexual precocity in tropical beef cattle. J Anim Sci 2018; 96:4087-4099. [PMID: 30053002 DOI: 10.1093/jas/sky289] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/20/2018] [Indexed: 12/31/2022] Open
Abstract
Multitrait meta-analyses are a strategy to produce more accurate genome-wide association studies, especially for complex phenotypes. We carried out a meta-analysis study for traits related to sexual precocity in tropical beef cattle (Nellore and Brahman) aiming to identify important genomic regions affecting these traits. The traits included in the analyses were age at first calving (AFC), early pregnancy (EP), age at first corpus luteum (AGECL), first postpartum anoestrus interval (PPAI), and scrotal circumference (SC). The traits AFC, EP, and SCN were measured in Nellore cattle, while AGECL, PPAI, and SCB were measured in Brahman cattle. Meta-analysis resulted in 108 significant single-nucleotide polymorphisms (SNPs), at an empirical threshold P-value of 1.39 × 10-5 (false discovery rate [FDR] < 0.05). Within 0.5 Mb of the significant SNP, candidate genes were annotated and analyzed for functional enrichment. Most of the closest genes to the SNP with higher significance in each chromosome have been associated with important roles in reproductive function. They are TSC22D2, KLF7, ARHGAP29, 7SK, MAP3K5, TLE3, WDR5, TAF3, TMEM68, PPP1R15B, NR2F2, GALR1, SUFU, and KCNU1. We did not observe any significant SNP in BTA5, BTA12, BTA17, BTA18, BTA19, BTA20, BTA22, BTA23, BTA25, and BTA28. Although the majority of significant SNPs are in BTA14, it was identified significant associations in multiple chromosomes (19 out of 29 autosomes), which is consistent with the postulation that reproductive traits are complex polygenic phenotypes. Five proposed association regions harbor the majority of the significant SNP (76%) and were distributed over four chromosomes (P < 1.39 × 10-5, FDR < 0.05): BTA2 (5.55%) from 95 to 96 Mb, BTA4 (5.55%) from 94.1 to 94.8 Mb, BTA14 (59.26%) from 24 to 25 Mb and 29 to 30 Mb, and BTA21 (5.55%) from 6.7 Mb to 11.4 Mb. These regions harbored key genes related to reproductive function. Moreover, these genes were enriched for functional groups associated with immune response, maternal-fetal tolerance, pregnancy maintenance, embryo development, fertility, and response to stress. Further studies including other breeds and precocity traits could confirm the importance of these regions and identify new candidate regions for sexual precocity in beef cattle.
Collapse
Affiliation(s)
- Thaise P Melo
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP - Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Marina R S Fortes
- The University of Queensland, School of Chemistry and Molecular Biosciences, St Lucia, Queensland, Australia.,The University of Queensland, Queensland Alliance for Agriculture and Food Innovation, St Lucia, Queensland, Australia
| | - Tiago Bresolin
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP - Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Lucio F M Mota
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP - Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil
| | - Lucia G Albuquerque
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP - Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil.,National Council for Scientific and Technological Development (CNPq), Brasília, Distrito Federal, Brazil
| | - Roberto Carvalheiro
- Department of Animal Science, School of Agricultural and Veterinarian Sciences, FCAV/ UNESP - Sao Paulo State University, Jaboticabal, Sao Paulo, Brazil.,National Council for Scientific and Technological Development (CNPq), Brasília, Distrito Federal, Brazil
| |
Collapse
|
38
|
Hoshino Y. Updating the markers for oocyte quality evaluation: intracellular temperature as a new index. Reprod Med Biol 2018; 17:434-441. [PMID: 30377396 PMCID: PMC6194278 DOI: 10.1002/rmb2.12245] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/07/2018] [Accepted: 08/09/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The developmental competence of an embryo is principally dictated by the oocyte. Usually, oocyte selection is based on morphological properties; however, all morphological criteria that are currently used for the grading and screening of oocytes are not able to eliminate the subjectivity. Despite recent studies of the molecular factors related to oocyte quality, it is technically difficult to develop an index based on these factors, and new indices that reflect intracellular conditions are necessary. METHODS Morphological and molecular factors influencing developmental competence were comprehensively reviewed, and intracellular temperature was evaluated as a new marker of oocyte quality. MAIN FINDINGS The intracellular temperature of mature oocytes was high in fresh oocytes and decreased with time after polar body release. Under the same conditions, the intracellular temperature and its distribution differed among oocytes, suggesting that temperature represents the state of each oocyte. CONCLUSION Intracellular temperature is advantageous as an objective and quantitative indicator of oocyte quality. Further studies should evaluate the link between temperature and cellular phenomena to establish its use as an indicator of quality.
Collapse
Affiliation(s)
- Yumi Hoshino
- Graduate School of Biosphere ScienceHiroshima UniversityHiroshimaJapan
| |
Collapse
|
39
|
Kay VR, Rätsep MT, Cahill LS, Hickman AF, Zavan B, Newport ME, Ellegood J, Laliberte CL, Reynolds JN, Carmeliet P, Tayade C, Sled JG, Croy BA. Effects of placental growth factor deficiency on behavior, neuroanatomy, and cerebrovasculature of mice. Physiol Genomics 2018; 50:862-875. [PMID: 30118404 DOI: 10.1152/physiolgenomics.00076.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Preeclampsia, a hypertensive syndrome occurring in 3-5% of human pregnancies, has lifelong health consequences for fetuses. Cognitive ability throughout life is altered, and adult stroke risk is increased. One potential etiological factor for altered brain development is low concentrations of proangiogenic placental growth factor (PGF). Impaired PGF production may promote an antiangiogenic fetal environment during neural and cerebrovascular development. We previously reported delayed vascularization of the hindbrain, altered retinal vascular organization, and less connectivity in the circle of Willis in Pgf-/- mice. We hypothesized Pgf-/- mice would have impaired cognition and altered brain neuroanatomy in addition to compromised cerebrovasculature. Cognitive behavior was assessed in adult Pgf-/- and Pgf+/+ mice by four paradigms followed by postmortem high-resolution MRI of neuroanatomy. X-ray microcomputed tomography imaging investigated the three-dimensional cerebrovascular geometry in another cohort. Pgf-/- mice exhibited poorer spatial memory, less depressive-like behavior, and superior recognition of novel objects. Significantly smaller volumes of 10 structures were detected in the Pgf-/- compared with Pgf+/+ brain. Pgf-/- brain had more total blood vessel segments in the small-diameter range. Lack of PGF altered cognitive functions, brain neuroanatomy, and cerebrovasculature in mice. Pgf-/- mice may be a preclinical model for the offspring effects of low-PGF preeclampsia gestation.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Matthew T Rätsep
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | - Andrew F Hickman
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Bruno Zavan
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada.,Federal University of Alfenas (UNIFAL), Alfenas, Minas Gerais , Brazil
| | - Margaret E Newport
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | | | - James N Reynolds
- Centre for Neuroscience Studies, Queen's University , Kingston, Ontario , Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB - Vesalius Research Center, University of Leuven, Department of Oncology , Leuven , Belgium
| | - Chandrakant Tayade
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada.,Department of Medical Biophysics, University of Toronto , Ontario , Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| |
Collapse
|
40
|
Simões R, Rodrigues Santos A. Factors and molecules that could impact cell differentiation in the embryo generated by nuclear transfer. Organogenesis 2018; 13:156-178. [PMID: 29020571 DOI: 10.1080/15476278.2017.1389367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Somatic cell nuclear transfer is a technique to create an embryo using an enucleated oocyte and a donor nucleus. Nucleus of somatic cells must be reprogrammed in order to participate in normal development within an enucleated egg. Reprogramming refers to the erasing and remodeling of cellular epigenetic marks to a lower differentiation state. Somatic nuclei must be reprogrammed by factors in the oocyte cytoplasm to a rather totipotent state since the reconstructed embryo must initiate embryo development from the one cell stage to term. In embryos reconstructed by nuclear transfer, the donor genetic material must respond to the cytoplasmic environment of the cytoplast and recapitulate this normal developmental process. Enucleation is critically important for cloning efficiency because may affect the ultrastructure of the remaining cytoplast, thus resulting in a decline or destruction of its cellular compartments. Nonetheless, the effects of in vitro culturing are yet to be fully understood. In vitro oocyte maturation can affect the abundance of specific transcripts and are likely to deplete the developmental competence. The epigenetic modifications established during cellular differentiation are a major factor determining this low efficiency as they act as epigenetic barriers restricting reprogramming of somatic nuclei. In this review we discuss some factors that could impact cell differentiation in embryo generated by nuclear transfer.
Collapse
Affiliation(s)
- Renata Simões
- a Centro de Ciências Naturais e Humanas, Universidade Federal do ABC , SP , Brazil
| | | |
Collapse
|
41
|
Gonzalez-Munoz E, Cibelli JB. Somatic Cell Reprogramming Informed by the Oocyte. Stem Cells Dev 2018; 27:871-887. [DOI: 10.1089/scd.2018.0066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Elena Gonzalez-Munoz
- LARCEL, Andalusian Laboratory of Cell Reprogramming (LARCel), Andalusian Center for Nanomedicine and Biotechnology-BIONAND, Málaga, Spain
- Department of Cell Biology, Genetics and Physiology, University of Málaga, Málaga, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Málaga, Spain
| | - Jose B. Cibelli
- LARCEL, Andalusian Laboratory of Cell Reprogramming (LARCel), Andalusian Center for Nanomedicine and Biotechnology-BIONAND, Málaga, Spain
- Department of Animal Science, Michigan State University, East Lansing, MI
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI
| |
Collapse
|
42
|
Martin JH, Bromfield EG, Aitken RJ, Lord T, Nixon B. Double Strand Break DNA Repair occurs via Non-Homologous End-Joining in Mouse MII Oocytes. Sci Rep 2018; 8:9685. [PMID: 29946146 PMCID: PMC6018751 DOI: 10.1038/s41598-018-27892-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/07/2018] [Indexed: 12/12/2022] Open
Abstract
The unique biology of the oocyte means that accepted paradigms for DNA repair and protection are not of direct relevance to the female gamete. Instead, preservation of the integrity of the maternal genome depends on endogenous protein stores and/or mRNA transcripts accumulated during oogenesis. The aim of this study was to determine whether mature (MII) oocytes have the capacity to detect DNA damage and subsequently mount effective repair. For this purpose, DNA double strand breaks (DSB) were elicited using the topoisomerase II inhibitor, etoposide (ETP). ETP challenge led to a rapid and significant increase in DSB (P = 0.0002) and the consequential incidence of metaphase plate abnormalities (P = 0.0031). Despite this, ETP-treated MII oocytes retained their ability to participate in in vitro fertilisation, though displayed reduced developmental competence beyond the 2-cell stage (P = 0.02). To account for these findings, we analysed the efficacy of DSB resolution, revealing a significant reduction in DSB lesions 4 h post-ETP treatment. Notably, this response was completely abrogated by pharmacological inhibition of key elements (DNA-PKcs and DNA ligase IV) of the canonical non-homologous end joining DNA repair pathway, thus providing the first evidence implicating this reparative cascade in the protection of the maternal genome.
Collapse
Affiliation(s)
- Jacinta H Martin
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia. .,Preganancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Preganancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Preganancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Tessa Lord
- School of Molecular Biosciences, Centre for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Preganancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| |
Collapse
|
43
|
Lu Y, Reddy R, Ferrer Buitrago M, Vander Jeught M, Neupane J, De Vos WH, Van den Abbeel E, Lierman S, De Sutter P, Heindryckx B. Strontium fails to induce Ca 2+ release and activation in human oocytes despite the presence of functional TRPV3 channels. Hum Reprod Open 2018; 2018:hoy005. [PMID: 30895246 PMCID: PMC6276696 DOI: 10.1093/hropen/hoy005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/31/2018] [Accepted: 03/15/2018] [Indexed: 01/27/2023] Open
Abstract
STUDY QUESTION Are the transient receptor potential cation channels vanilloid 3 (TRPV3) present and able to mediate strontium (Sr2+) induced artificial activation in human oocytes? SUMMARY ANSWER Sr2+ did not induce Ca2+ rises or provoke activation in human oocytes, however, mRNA for the TRPV3 channel was present in metaphase II (MII) human oocytes after IVM and TRPV3 agonists induced Ca2+ rises and oocyte activation, demonstrating the channels were functional. WHAT IS KNOWN ALREADY Selective activation of TRPV3 by agonists induces Ca2+ entry and promotes mouse oocyte activation, and the absence of TRPV3 channels in mouse oocytes prevents Sr2+ mediated artificial activation. Sr2+ is sometimes used to overcome fertilization failure after ICSI in the clinic, but its efficiency is still controversial and the mechanism(s) of how it mediates the Ca2+ flux has not been studied yet in human. STUDY DESIGN, SIZE, DURATION The protein distribution (n = 10) and mRNA expression level (n = 19) of the TRPV3 channels was investigated in human MII oocytes after IVM. The Sr2+ (10 mM) and TRPV3 agonists (200 μM 2-aminoethoxydiphenyl borate [2-APB] and 200 μM carvacrol)-induced Ca2+ response was analyzed in human (n = 15, n = 16 and n = 16, respectively) and mouse oocytes (n = 15, n = 19 and n = 26, respectively). The subsequent embryonic developmental potential following the parthenogenetic activation using these three agents was recorded in human (n = 10, n = 9 and n = 9, respectively) and mouse (n = 20 per agent) oocytes, by determining pronucleus, or 2-cell and blastocyst formation rates. PARTICIPANTS/MATERIALS, SETTING, METHODS MII oocytes from B6D2F1 mice (6–10 weeks old) as well as human IVM oocytes and IVO oocytes (from patients aged 25–38 years old) with aggregates of smooth endoplasmic reticulum clusters were used. The expression of TRPV3 channels was determined by immunofluorescence staining with confocal microscopy and RT-PCR, and the temporal evolution of intracellular Ca2+ concentration was measured by time-lapse imaging after exposure to Sr2+ and TRPV3 agonists (2-APB and carvacrol). Artificial activation efficiency was assessed using these agents. MAIN RESULTS AND THE ROLE OF CHANCE Sr2+ did not promote Ca2+ oscillations or provoke activation in human oocytes. Transcripts of TRPV3 channels were present in IVM MII human oocytes. TRPV3 protein was expressed and distributed throughout the ooplasm of human oocytes, rather than particularly concentrated in plasma membrane as observed in mouse MII oocytes. Both agonists of TRPV3 (2-APB and carvacrol), promoted a single Ca2+ transient and activated a comparable percentage of more than half of the exposed human oocytes (P > 0.05). The agonist 2-APB was also efficient in activating mouse oocytes, however, significantly fewer mouse oocytes responded to carvacrol than 2-APB in both the Ca2+ analysis and activation test (P < 0.001). LIMITATIONS REASONS FOR CAUTION The availability of fresh IVO matured oocytes in human was limited. Data from TRPV3 knockout model are not included. WIDER IMPLICATIONS OF THE FINDINGS The benefit of clinical application using Sr2+ to overcome fertilization failure after ICSI requires further validation. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by FWO-Vlaanderen, China Scholarship Council and Special Research Fund from Ghent University (Bijzonder Onderzoeksfonds, BOF). No competing interests are declared.
Collapse
Affiliation(s)
- Y Lu
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.,Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Renmin South Road 20, 610041 Chengdu, China
| | - R Reddy
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - M Ferrer Buitrago
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - M Vander Jeught
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - J Neupane
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - W H De Vos
- Cell Systems and Imaging research Group (CSI), Department of Molecular Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium.,Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerpen, Belgium
| | - E Van den Abbeel
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - S Lierman
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - P De Sutter
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - B Heindryckx
- Ghent-Fertility and Stem Cell Team (G-FaST), Department for Reproductive Medicine, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| |
Collapse
|
44
|
Barragán M, Pons J, Ferrer-Vaquer A, Cornet-Bartolomé D, Schweitzer A, Hubbard J, Auer H, Rodolosse A, Vassena R. The transcriptome of human oocytes is related to age and ovarian reserve. Mol Hum Reprod 2018; 23:535-548. [PMID: 28586423 DOI: 10.1093/molehr/gax033] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 06/03/2017] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION How does the human oocyte transcriptome change with age and ovarian reserve? SUMMARY ANSWER Specific sets of human oocyte messenger RNAs (mRNAs) and non-coding RNAs (ncRNAs) are affected independently by age and ovarian reserve. WHAT IS KNOWN ALREADY Although it is well established that the ovarian reserve diminishes with increasing age, and that a woman's age is correlated with lower oocyte quality, the interplay of a diminished reserve and age on oocyte developmental competence is not clear. After maturation, oocytes are mostly transcriptionally quiescent, and developmental competence prior to embryonic genome activationrelies on maternal RNA and proteins. STUDY DESIGN, SIZE, DURATION A total of 36 vitrified/warmed MII oocytes from 30 women undergoing oocyte donation were included in this study, processed and analyzed individually. PARTICIPANTS/MATERIALS, SETTING, METHODS Total RNA from each oocyte was independently isolated, amplified, labeled, and hybridized on HTA 2.0 arrays (Affymetrix). Data were analyzed using TAC software, in four groups, each including nine oocytes, according to the woman's age and antral follicular count (AFC) (mean ± SD): Young with High AFC (YH; age 21 ± 1 years and 24 ± 3 follicles); Old with High AFC (OH; age 32 ± 2 years and 29 ± 7 follicles); Young with Low AFC (YL; age 24 ± 2 years and 8 ± 2 follicles); Old with Low AFC (OL; age 34 ± 1 years and 7 ± 1 follicles). qPCR was performed to validate arrays. MAIN RESULTS AND THE ROLE OF CHANCE We identified a set of 30 differentially expressed mRNAs when comparing oocytes from women with different ages and AFC. In addition, 168 non-coding RNAs (ncRNAs) were differentially expressed in relation to age and/or AFC. Few mRNAs have been identified as differentially expressed transcripts, and among ncRNAs, a set of Piwi-interacting RNAs clusters (piRNAs-c) and precursor microRNAs (pre-miRNAs) were identified as increased in high AFC and old groups, respectively. Our results indicate that age and ovarian reserve are associated with specific ncRNA profiles, suggesting that oocyte quality might be mediated by ncRNA pathways. LARGE SCALE DATA Data can be found via GEO accession number GSE87201. LIMITATIONS, REASONS FOR CAUTION The oldest woman included in the study was 35 years old, thus our results cannot readily be extrapolated to women older than 35 or infertile women. WIDER IMPLICATIONS OF THE FINDINGS We show, for the first time, that several non-coding RNAs, usually regulating DNA transcription, are differentially expressed in relation to age and/or ovarian reserve. Interestingly, the mRNA transcriptome of in vivo matured oocytes remains remarkably stable across ages and ovarian reserve, suggesting the possibility that changes in the non-coding transcriptome might regulate some post-transcriptional/translational mechanisms which might, in turn, affect oocyte developmental competence. STUDY FUNDING AND COMPETING INTEREST(S) This work was supported by intramural funding of Clinica EUGIN and by the Secretary for Universities and Research of the Ministry of Economy and Knowledge of the Government of Catalonia. J.H. and A.S. are employees of Affymetrix, otherwise there are no competing interests.
Collapse
Affiliation(s)
- M Barragán
- Clínica EUGIN, Travessera de les Corts 322, 08029 Barcelona, Spain
| | - J Pons
- Functional Genomics Core, Institute for Research in Biomedicine (IRB) Barcelona, Parc Científic de Barcelona, Baldiri Reixac 10, 08028 Barcelona, Spain
| | - A Ferrer-Vaquer
- Clínica EUGIN, Travessera de les Corts 322, 08029 Barcelona, Spain
| | | | - A Schweitzer
- Thermo Fisher Scientific, 3450 Central Expressway, Santa Clara, CA 95051, USA
| | - J Hubbard
- Thermo Fisher Scientific, 3450 Central Expressway, Santa Clara, CA 95051, USA
| | - H Auer
- Functional GenOmics Consulting, Bellavista 53, 08753 Pallejà, Spain
| | - A Rodolosse
- Functional Genomics Core, Institute for Research in Biomedicine (IRB) Barcelona, Parc Científic de Barcelona, Baldiri Reixac 10, 08028 Barcelona, Spain
| | - R Vassena
- Clínica EUGIN, Travessera de les Corts 322, 08029 Barcelona, Spain
| |
Collapse
|
45
|
Zhu B, Pardeshi L, Chen Y, Ge W. Transcriptomic Analysis for Differentially Expressed Genes in Ovarian Follicle Activation in the Zebrafish. Front Endocrinol (Lausanne) 2018; 9:593. [PMID: 30364302 PMCID: PMC6193065 DOI: 10.3389/fendo.2018.00593] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/18/2018] [Indexed: 12/30/2022] Open
Abstract
In teleosts, the onset of puberty in females is marked by the appearance of the first wave of pre-vitellogenic (PV) follicles from the pool of primary growth (PG) follicles (follicle activation) in the ovary during sexual maturation. To understand the mechanisms underlying follicle activation and therefore puberty onset, we undertook this transcriptomic study to investigate gene expression profiles in the event. Our analysis revealed a total of 2,027 up-regulated and 859 down-regulated genes during the PG-PV transition. Gene Ontology (GO) analysis showed that in addition to basic cellular functions such as gene transcription, cell differentiation, and cell migration, other biological processes such as steroidogenesis, cell signaling and angiogenesis were also enriched in up-regulated genes; by comparison, some processes were down-regulated including piRNA metabolism, gene silencing and proteolysis. Further Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis identified a variety of signaling pathways that might play pivotal roles in PG-PV transition, including MAPK, TGF-β, Hedgehog, FoxO, VEGF, Jak-STAT, and phosphatidylinositol signaling pathways. Other pathways of particular interest included endocytosis and glycosaminoglycan biosynthesis. We also analyzed expression changes of genes expressed in different compartments viz. oocytes and follicle cells. Interestingly, most oocyte-specific genes remained unchanged in expression during follicle activation whereas a great number of genes specifically expressed in the follicle cells showed significant changes in expression. Overall, this study reported a comprehensive analysis for genes, biological processes and pathways involved in follicle activation, which also marks female puberty onset in the zebrafish when occurring for the first time in sexual maturation.
Collapse
Affiliation(s)
- Bo Zhu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, China
| | - Lakhansing Pardeshi
- Genomics and Bioinformatics Core, Faculty of Health Sciences, University of Macau, Taipa, China
| | - Yingying Chen
- Genomics and Bioinformatics Core, Faculty of Health Sciences, University of Macau, Taipa, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, China
- *Correspondence: Wei Ge ;
| |
Collapse
|
46
|
Abstract
The establishment of DNA methylation patterns in oocytes is a highly dynamic process marking gene-regulatory events during fertilization, embryonic development, and adulthood. However, after epigenetic reprogramming in primordial germ cells, how and when DNA methylation is re-established in developing human oocytes remains to be characterized. Here, using single-cell whole-genome bisulfite sequencing, we describe DNA methylation patterns in three different maturation stages of human oocytes. We found that while broad-scale patterns of CpG methylation have been largely established by the immature germinal vesicle stage, localized changes continue into later development. Non-CpG methylation, on the other hand, undergoes a large-scale, generalized remodeling through the final stage of maturation, with the net overall result being the accumulation of methylation as oocytes mature. The role of the genome-wide, non-CpG methylation remodeling in the final stage of oocyte maturation deserves further investigation.
Collapse
|
47
|
Martin JH, Bromfield EG, Aitken RJ, Nixon B. Biochemical alterations in the oocyte in support of early embryonic development. Cell Mol Life Sci 2017; 74:469-485. [PMID: 27604868 PMCID: PMC11107538 DOI: 10.1007/s00018-016-2356-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/28/2016] [Accepted: 09/01/2016] [Indexed: 01/01/2023]
Abstract
Notwithstanding the enormous reproductive potential encapsulated within a mature mammalian oocyte, these cells present only a limited window for fertilization before defaulting to an apoptotic cascade known as post-ovulatory oocyte aging. The only cell with the capacity to rescue this potential is the fertilizing spermatozoon. Indeed, the union of these cells sets in train a remarkable series of events that endows the oocyte with the capacity to divide and differentiate into the trillions of cells that comprise a new individual. Traditional paradigms hold that, beyond the initial stimulation of fluctuating calcium (Ca2+) required for oocyte activation, the fertilizing spermatozoon plays limited additional roles in the early embryo. While this model has now been drawn into question in view of the recent discovery that spermatozoa deliver developmentally important classes of small noncoding RNAs and other epigenetic modulators to oocytes during fertilization, it is nevertheless apparent that the primary responsibility for oocyte activation rests with a modest store of maternally derived proteins and mRNA accumulated during oogenesis. It is, therefore, not surprising that widespread post-translational modifications, in particular phosphorylation, hold a central role in endowing these proteins with sufficient functional diversity to initiate embryonic development. Indeed, proteins targeted for such modifications have been linked to oocyte activation, recruitment of maternal mRNAs, DNA repair and resumption of the cell cycle. This review, therefore, seeks to explore the intimate relationship between Ca2+ release and the suite of molecular modifications that sweep through the oocyte to ensure the successful union of the parental germlines and ensure embryogenic fidelity.
Collapse
Affiliation(s)
- Jacinta H Martin
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.
| | - Elizabeth G Bromfield
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - R John Aitken
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Brett Nixon
- Discipline of Biological Sciences and Priority Research Center for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| |
Collapse
|
48
|
Zhenhua G, Rajput SK, Folger JK, Di L, Knott JG, Smith GW. Pre- and Peri-/Post-Compaction Follistatin Treatment Increases In Vitro Production of Cattle Embryos. PLoS One 2017; 12:e0170808. [PMID: 28122009 PMCID: PMC5266319 DOI: 10.1371/journal.pone.0170808] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/11/2017] [Indexed: 12/23/2022] Open
Abstract
Our previous studies demonstrated that maternal (oocyte derived) follistatin (FST) expression is positively associated with bovine oocyte competence and exogenous follistatin treatment during the pre-compaction period of development (d 1–3 post insemination) is stimulatory to bovine early embryogenesis in vitro [blastocyst rates and cell numbers/allocation to trophectoderm (TE)]. In the present study, bovine embryos were treated with exogenous follistatin during d 1–3, d 4–7 and d 1–7 post insemination to test the hypothesis that embryotropic effects of exogenous follistatin are specific to the pre-compaction period (d 1–3) of early embryogenesis. Follistatin treatment during d 4–7 (peri-/post-compaction period) of embryo culture increased proportion of embryos reaching blastocyst and expanded blastocyst stage and total cell numbers compared to controls, but blastocyst rates and total cell numbers were lower than observed following d 1–3 (pre-compaction) follistatin treatment. Follistatin supplementation during d 1–7 of embryo culture increased development to blastocyst and expanded blastocyst stages and blastocyst total cell numbers compared to d 1–3 and d 4–7 follistatin treatment and untreated controls. A similar increase in blastocyst CDX2 mRNA and protein (TE cell marker) was observed in response to d 1–3, d 4–7 and d 1–7 follistatin treatment. However, an elevation in blastocyst BMP4 protein (TE cell regulator) was observed in response to d 1–3 and d 1–7, but not d 4–7 (peri-/post-compaction) follistatin treatment. In summary, our study revealed the potential utility of follistatin treatment for increasing the success rate of in vitro embryo production in cattle. Such results also expand our understanding of the embryotropic actions of follistatin and demonstrate that follistatin actions on blastocyst development and cell allocation to the TE layer are not specific to the pre-compaction period.
Collapse
Affiliation(s)
- Guo Zhenhua
- Animal Husbandry Research Institute of Heilongjiang Academy of Agricultural Sciences (HAAS), Harbin, P.R. China
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, United States of America
| | - Sandeep K. Rajput
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, United States of America
- Department of Animal Science, Michigan State University, East Lansing, MI, United States of America
| | - Joseph K. Folger
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, United States of America
- Department of Animal Science, Michigan State University, East Lansing, MI, United States of America
| | - Liu Di
- Animal Husbandry Research Institute of Heilongjiang Academy of Agricultural Sciences (HAAS), Harbin, P.R. China
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, United States of America
| | - Jason G. Knott
- Department of Animal Science, Michigan State University, East Lansing, MI, United States of America
- Developmental Epigenetics Laboratory, Michigan State University, East Lansing, MI, United States of America
| | - George W. Smith
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI, United States of America
- Department of Animal Science, Michigan State University, East Lansing, MI, United States of America
- * E-mail:
| |
Collapse
|
49
|
Rätsep MT, Hickman AF, Croy BA. The Elsevier trophoblast research award lecture: Impacts of placental growth factor and preeclampsia on brain development, behaviour, and cognition. Placenta 2016; 48 Suppl 1:S40-S46. [PMID: 26880207 DOI: 10.1016/j.placenta.2016.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 01/19/2016] [Accepted: 02/03/2016] [Indexed: 12/17/2022]
Abstract
Preeclampsia (PE) is a significant gestational disorder affecting 3-5% of all human pregnancies. In many PE pregnancies, maternal plasma is deficient in placental growth factor (PGF), a placentally-produced angiokine. Beyond immediate fetal risks associated with acute termination of the pregnancy, offspring of PE pregnancies (PE-F1) have higher long-term risks for hypertension, stroke, and cognitive impairment compared to F1s from uncomplicated pregnancies. At present, mechanisms that explain PE-F1 gains in postpartum risks are poorly understood. Our laboratory found that mice genetically-deleted for Pgf have altered fetal and adult brain vascular development. This is accompanied by sexually dimorphic alterations in anatomic structure in the adult Pgf-/- brain and impaired cognitive functions. We hypothesize that cerebrovascular and neurological aberrations occur in fetuses exposed to the progressive development of PE and that these brain changes impair cognitive functioning, enhance risk for stroke, elevate severity of stroke, and lead to worse stroke outcomes. These brain and placental outcomes may be linked to down-regulated PGF gene expression in early pre-implantation embryos, prior to gastrulation. This review explores our hypothesis that there are mechanistic links between low PGF detection in maternal plasma prodromal to PE, PE, and altered brain vascular, structural, and functional development amongst PE-F1s. We also include a summary of preliminary outcomes from a pilot study of 7-10 year old children that is the first to report magnetic resonance imaging, magnetic resonance angiography, and functional brain region assessment by eye movement control studies in PE-F1s.
Collapse
Affiliation(s)
- Matthew T Rätsep
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| | - Andrew F Hickman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
50
|
Glinsky GV. Single cell genomics reveals activation signatures of endogenous SCAR's networks in aneuploid human embryos and clinically intractable malignant tumors. Cancer Lett 2016; 381:176-93. [DOI: 10.1016/j.canlet.2016.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 08/01/2016] [Indexed: 10/21/2022]
|