301
|
Sciorio R, El Hajj N. Epigenetic Risks of Medically Assisted Reproduction. J Clin Med 2022; 11:jcm11082151. [PMID: 35456243 PMCID: PMC9027760 DOI: 10.3390/jcm11082151] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 12/14/2022] Open
Abstract
Since the birth of Louise Joy Brown, the first baby conceived via in vitro fertilization, more than 9 million children have been born worldwide using assisted reproductive technologies (ART). In vivo fertilization takes place in the maternal oviduct, where the unique physiological conditions guarantee the healthy development of the embryo. During early embryogenesis, a major wave of epigenetic reprogramming takes place that is crucial for the correct development of the embryo. Epigenetic reprogramming is susceptible to environmental changes and non-physiological conditions such as those applied during in vitro culture, including shift in pH and temperature, oxygen tension, controlled ovarian stimulation, intracytoplasmic sperm injection, as well as preimplantation embryo manipulations for genetic testing. In the last decade, concerns were raised of a possible link between ART and increased incidence of imprinting disorders, as well as epigenetic alterations in the germ cells of infertile parents that are transmitted to the offspring following ART. The aim of this review was to present evidence from the literature regarding epigenetic errors linked to assisted reproduction treatments and their consequences on the conceived children. Furthermore, we provide an overview of disease risk associated with epigenetic or imprinting alterations in children born via ART.
Collapse
Affiliation(s)
- Romualdo Sciorio
- Edinburgh Assisted Conception Programme, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, UK
- Correspondence:
| | - Nady El Hajj
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar;
| |
Collapse
|
302
|
Brooks KE, Daughtry BL, Davis B, Yan MY, Fei SS, Shepherd S, Carbone L, Chavez SL. Molecular contribution to embryonic aneuploidy and karyotypic complexity in initial cleavage divisions of mammalian development. Development 2022; 149:dev198341. [PMID: 35311995 PMCID: PMC9058497 DOI: 10.1242/dev.198341] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/04/2022] [Indexed: 01/05/2023]
Abstract
Embryonic aneuploidy is highly complex, often leading to developmental arrest, implantation failure or spontaneous miscarriage in both natural and assisted reproduction. Despite our knowledge of mitotic mis-segregation in somatic cells, the molecular pathways regulating chromosome fidelity during the error-prone cleavage-stage of mammalian embryogenesis remain largely undefined. Using bovine embryos and live-cell fluorescent imaging, we observed frequent micro-/multi-nucleation of mis-segregated chromosomes in initial mitotic divisions that underwent unilateral inheritance, re-fused with the primary nucleus or formed a chromatin bridge with neighboring cells. A correlation between a lack of syngamy, multipolar divisions and asymmetric genome partitioning was also revealed, and single-cell DNA-seq showed propagation of primarily non-reciprocal mitotic errors. Depletion of the mitotic checkpoint protein BUB1B (also known as BUBR1) resulted in similarly abnormal nuclear structures and cell divisions, as well as chaotic aneuploidy and dysregulation of the kinase-substrate network that mediates mitotic progression, all before zygotic genome activation. This demonstrates that embryonic micronuclei sustain multiple fates, provides an explanation for blastomeres with uniparental origins, and substantiates defective checkpoints and likely other maternally derived factors as major contributors to the karyotypic complexity afflicting mammalian preimplantation development.
Collapse
Affiliation(s)
- Kelsey E. Brooks
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Brittany L. Daughtry
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Brett Davis
- Bioinformatics and Biostatistics Unit, Oregon National Primate Research Center, Beaverton, OR 97006, USA
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Melissa Y. Yan
- Bioinformatics and Biostatistics Unit, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Suzanne S. Fei
- Bioinformatics and Biostatistics Unit, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Selma Shepherd
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Lucia Carbone
- Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA
- Division of Genetics, Oregon National Primate Research Center, Beaverton, OR 97006, USA
- Department of Medical Informatics and Clinical Epidemiology, Division of Bioinformatics and Computational Biomedicine, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Shawn L. Chavez
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR 97006, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
303
|
Poly(I:C) exposure during in vitro fertilization disrupts first cleavage of mouse embryos and subsequent blastocyst development. J Reprod Immunol 2022; 151:103635. [DOI: 10.1016/j.jri.2022.103635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/19/2022] [Accepted: 04/27/2022] [Indexed: 11/18/2022]
|
304
|
Feng W, Zhang D, Fu L, Hu J, Gao S, Song X, Cui L. Temporary Increased LDL-C in Offspring with Extreme Elevation of Maternal Preconception Estradiol: A Retrospective Cohort Study. Clin Epidemiol 2022; 14:453-462. [PMID: 35418784 PMCID: PMC9001024 DOI: 10.2147/clep.s358999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/30/2022] [Indexed: 12/04/2022] Open
Abstract
Objective To investigate the effect of maternal estradiol (E2) elevation on long-term metabolic manifestations in the offspring. Study Design and Setting This was a retrospective cohort study. Overall, 3690 children conceived by in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) between July 2014 and December 2017 were recruited and divided into four groups categorized by maternal E2 quartiles (Q1, <2420; Q2, 2420–3839; Q3, 3839–5599; and Q4, ≥5599 pg/mL). The metabolic profiles were measured during childhood. Linear mixed models were used to evaluate the association between maternal E2 elevation and metabolic phenotypes of the offspring. Results Lipoprotein cholesterol (LDL-C) was significantly higher in the highest quartile group than in the lowest quartile group during infancy (adjusted mean difference [95% confidence interval, CI]): 0.11 [0.02, 0.20], P = 0.005), but the difference disappeared in the later childhood phase. In children born after fresh embryo transfer, LDL-C showed an increasing trend with the increase in maternal E2 level (adjusted mean difference [95% CI]: Q2 vs Q1, −0.01 [−0.11, 0.08], Q3 vs Q1, 0.06 [−0.04, 0.15], Q4 vs Q1, 0.10 [0, 0.20]). Other metabolic variables were comparable across increasing quartiles of maternal E2 levels. Conclusion This study demonstrates a temporary increase in LDL-C levels in infants with higher levels of maternal preconception E2 levels. However, the long-term safety of hyperestrogens after ovarian stimulation in the next generation is favorable. The mechanism underlying the transiently increased metabolic dysfunction risk in infants conceived by IVF/ICSI requires investigation in future studies.
Collapse
Affiliation(s)
- Wanbing Feng
- Center for Reproductive Medicine, Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, People’s Republic of China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Di Zhang
- Center for Reproductive Medicine, Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, People’s Republic of China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Linlin Fu
- Center for Reproductive Medicine, Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, People’s Republic of China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Jingmei Hu
- Center for Reproductive Medicine, Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, People’s Republic of China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Shanshan Gao
- Center for Reproductive Medicine, Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, People’s Republic of China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Xiaocui Song
- Center for Reproductive Medicine, Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Linlin Cui
- Center for Reproductive Medicine, Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, People’s Republic of China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, People’s Republic of China
- Correspondence: Linlin Cui; Xiaocui Song, Center for Reproductive Medicine, Department of Reproductive Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 157, Jingliu Road, Shizhong District, Jinan, Shandong, People’s Republic of China, 250001, Tel +86 531 85651598, Fax +86 531 87068226, Email ;
| |
Collapse
|
305
|
Fan XY, Guo L, Chen LN, Yin S, Wen J, Li S, Ma JY, Jing T, Jiang MX, Sun XH, Chen M, Wang F, Wang ZB, Zhang CF, Wang XH, Ge ZJ, Hu C, Zeng L, Shen W, Sun QY, Ou XH, Luo SM. Reduction of mtDNA heteroplasmy in mitochondrial replacement therapy by inducing forced mitophagy. Nat Biomed Eng 2022; 6:339-350. [PMID: 35437313 DOI: 10.1038/s41551-022-00881-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/10/2022] [Indexed: 11/08/2022]
Abstract
Mitochondrial replacement therapy (MRT) has been used to prevent maternal transmission of disease-causing mutations in mitochondrial DNA (mtDNA). However, because MRT requires nuclear transfer, it carries the risk of mtDNA carryover and hence of the reversion of mtDNA to pathogenic levels owing to selective replication and genetic drift. Here we show in HeLa cells, mouse embryos and human embryos that mtDNA heteroplasmy can be reduced by pre-labelling the mitochondrial outer membrane of a donor zygote via microinjection with an mRNA coding for a transmembrane peptide fused to an autophagy receptor, to induce the degradation of the labelled mitochondria via forced mitophagy. Forced mitophagy reduced mtDNA carryover in newly reconstructed embryos after MRT, and had negligible effects on the growth curve, reproduction, exercise capacity and other behavioural characteristics of the offspring mice. The induction of forced mitophagy to degrade undesired donor mtDNA may increase the clinical feasibility of MRT and could be extended to other nuclear transfer techniques.
Collapse
Affiliation(s)
- Xiao-Yan Fan
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
| | - Lei Guo
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Lei-Ning Chen
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Shen Yin
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
| | - Jiarong Wen
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Sen Li
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Jun-Yu Ma
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Tao Jing
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Man-Xi Jiang
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiao-Hong Sun
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Meilan Chen
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Feng Wang
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chang-Fa Zhang
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xing-Hua Wang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
| | - Zhao-Jia Ge
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
| | - Chun Hu
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Lizhang Zeng
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Xiang-Hong Ou
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.
| | - Shi-Ming Luo
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, China.
| |
Collapse
|
306
|
Li XY, Pan JX, Zhu H, Ding GL, Huang HF. Environmental epigenetic interaction of gametes and early embryos. Biol Reprod 2022; 107:196-204. [PMID: 35323884 DOI: 10.1093/biolre/ioac051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 11/14/2022] Open
Abstract
In recent years, the developmental origins of diseases have been increasingly recognized and accepted. As such, it has been suggested that most adulthood chronic diseases such as diabetes, obesity, cardiovascular disease, and even tumors may develop at a very early stage. In addition to intrauterine environmental exposure, germ cells carry an important inheritance role as the primary link between the two generations. Adverse external influences during differentiation and development can cause damage to germ cells, which may then increase the risk of chronic disease development later in life. Here, we further elucidate and clarify the concept of gamete and embryo origins of adult diseases by focusing on the environmental insults on germ cells, from differentiation to maturation and fertilization.
Collapse
Affiliation(s)
- Xin-Yuan Li
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences
| | - Jie-Xue Pan
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences
| | - Hong Zhu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences
| | - Guo-Lian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - He-Feng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.,Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.,The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Hangzhou, China
| |
Collapse
|
307
|
Xiong Y, Yu C, Zhang Q. Ubiquitin-Proteasome System-Regulated Protein Degradation in Spermatogenesis. Cells 2022; 11:1058. [PMID: 35326509 PMCID: PMC8947704 DOI: 10.3390/cells11061058] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/14/2022] [Accepted: 03/18/2022] [Indexed: 12/12/2022] Open
Abstract
Spermatogenesis is a prolonged and highly ordered physiological process that produces haploid male germ cells through more than 40 steps and experiences dramatic morphological and cellular transformations. The ubiquitin proteasome system (UPS) plays central roles in the precise control of protein homeostasis to ensure the effectiveness of certain protein groups at a given stage and the inactivation of them after this stage. Many UPS components have been demonstrated to regulate the progression of spermatogenesis at different levels. Especially in recent years, novel testis-specific proteasome isoforms have been identified to be essential and unique for spermatogenesis. In this review, we set out to discuss our current knowledge in functions of diverse USP components in mammalian spermatogenesis through: (1) the composition of proteasome isoforms at each stage of spermatogenesis; (2) the specificity of each proteasome isoform and the associated degradation events; (3) the E3 ubiquitin ligases mediating protein ubiquitination in male germ cells; and (4) the deubiquitinases involved in spermatogenesis and male fertility. Exploring the functions of UPS machineries in spermatogenesis provides a global picture of the proteome dynamics during male germ cell production and shed light on the etiology and pathogenesis of human male infertility.
Collapse
Affiliation(s)
- Yi Xiong
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, 718 East Haizhou Rd, Haining 314400, China;
| | - Chao Yu
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Assisted Reproduction Unit, Department of Obstetrics and Gynecology, School of Medicine, Zhejiang University, Sir Run Run Shaw Hospital, 3 East Qing Chun Rd, Hangzhou 310020, China;
- College of Life Sciences, Zhejiang University, 866 Yuhangtang Rd, Hangzhou 310058, China
| | - Qianting Zhang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, 718 East Haizhou Rd, Haining 314400, China;
- Department of Dermatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
| |
Collapse
|
308
|
Di Nisio V, Antonouli S, Damdimopoulou P, Salumets A, Cecconi S. In vivo and in vitro postovulatory aging: when time works against oocyte quality? J Assist Reprod Genet 2022; 39:905-918. [PMID: 35312936 PMCID: PMC9050976 DOI: 10.1007/s10815-022-02418-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/27/2022] [Indexed: 12/26/2022] Open
Abstract
In mammalian species an optimal fertilization window during which successful fertilization occurs. In the majority of mammals estrus marks ovulation time and coincident with mating, thereby allowing the synchronized meeting in the fallopian tubes, between freshly ejaculated sperm and freshly ovulated oocytes. Conversely, women do not show natural visual signs of ovulation such that fertilization can occur hours later involving an aged oocyte and freshly ejaculated spermatozoa. During this time, the oocyte undergoes a rapid degradation known as “postovulatory aging” (POA). POA may become particularly important in the human-assisted reproductive technologies, as the fertilization of retrieved mature oocytes can be delayed due to increased laboratory workload or because of unforeseeable circumstances, like the delayed availability of semen samples. This paper is an updated review of the consequences of POA, either in vivo or in vitro, on oocyte quality with particular attention to modifications caused by POA on oocyte nuclear, cytoplasmic, genomic, and epigenetic maturation, and embryo development.
Collapse
Affiliation(s)
- Valentina Di Nisio
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, 14186, Huddinge, Stockholm, Sweden.
| | - Sevastiani Antonouli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, 14186, Huddinge, Stockholm, Sweden
| | - Andres Salumets
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, 14186, Huddinge, Stockholm, Sweden.,Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia.,Competence Centre On Health Technologies, 50411, Tartu, Estonia
| | - Sandra Cecconi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy.
| | | |
Collapse
|
309
|
Analysis of Clinical Outcomes of Different Fertilization Methods in Patients with ≤3 Eggs Retrieved. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:9467568. [PMID: 35340232 PMCID: PMC8942641 DOI: 10.1155/2022/9467568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 12/03/2022]
Abstract
Objective To explore the intracytoplasmic sperm injection (ICSI) and in vitro fertilization (IVF) method on the clinical outcomes of infertile women with ≤3 eggs retrieved. Study Design. We retrospectively analyzed a cohort of female patients who received IVF/ICSI to assist pregnancy with retrieved eggs ≤3. The general conditions, i.e., two pronuclei (2PN) fertilization rate, abnormal fertilization rate, high-quality embryo rate, cycle cancellation rate, pregnancy rate of fresh embryo transfer, cumulative pregnancy rate, and miscarriage were compared between the two groups. Results When the number of retrieved eggs was one, the fertilization rate of 2PN was higher and the cycle cancellation rate was lower in the ICSI group than in the IVF group (P < 0.05). The pregnancy rates of fresh embryo transfer, frozen-thawed embryo transfer, and the cumulative pregnancy rate were all higher in the ICSI group than in the IVF group (P < 0.05). When the number of retrieved eggs was two, the pregnancy rate of frozen-thawed embryo transfer and cumulative pregnancy rate in the ICSI group were higher than those in the IVF group (P < 0.05). When the number of retrieved eggs was three, the fertilization rate of 2PN and the pregnancy rate of frozen-thawed embryo transfer were higher in the ICSI group than those in the IVF group (all (P < 0.05)). Conclusions For patients with one egg retrieved, ICSI fertilization can reduce abnormal fertilization rate and cycle cancellation rate and improve cumulative pregnancy rate significantly enhancing patients' benefits. However, increasing the number of eggs retrieved decreases the advantages of ICSI fertilization.
Collapse
|
310
|
Li Q, Zhao T, He H, Robert N, Ding T, Hu X, Zhang T, Pan Y, Cui Y, Yu S. Ascorbic acid protects the toxic effects of aflatoxin B 1 on yak oocyte maturation. Anim Sci J 2022; 93:e13702. [PMID: 35257449 DOI: 10.1111/asj.13702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/26/2022] [Accepted: 02/03/2022] [Indexed: 11/29/2022]
Abstract
High-quality oocytes are a prerequisite for successful fertilization. Mammals feeding on aflatoxin-contaminated feed can cause reproductive toxicity, including follicular atresia, poor oocyte development and maturation, and aberrant epigenetic modifications of oocytes. In addition, the important role of ascorbic acid (AA) in reproductive biology has been confirmed, and AA is widely used as an antioxidant in cell culture. However, the toxic effects of aflatoxin B1 (AFB1 ) on yak oocytes and whether AA has protective effects remain unknown. In this study, we found that exposure to AFB1 impedes meiotic maturation of oocytes, promotes apoptosis by triggering high levels of reactive oxygen species (ROS), and disrupts mitochondrial distribution and actin integrity, resulting in a decrease in the fertilization ability and parthenogenetic development ability of oocytes. In addition, these injuries changed the DNA methylation transferase transcription level of mature oocytes. After adding 50 μg/ml AA, the indices recovered to levels close to those of the control group. The results showed that AA could protect yak oocytes from the toxic effects of AFB1 and improve the quality of oocytes.
Collapse
Affiliation(s)
- Qin Li
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tian Zhao
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Honghong He
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Niayale Robert
- Laboratory of Animal Anatomy & Tissue Embryology, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tianyi Ding
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xuequan Hu
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tongxiang Zhang
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yangyang Pan
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yan Cui
- Laboratory of Animal Anatomy & Tissue Embryology, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Sijiu Yu
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
311
|
Almeida JI, Tenreiro MF, Martinez-Santamaria L, Guerrero-Aspizua S, Gisbert JP, Alves PM, Serra M, Baptista PM. Hallmarks of the human intestinal microbiome on liver maturation and function. J Hepatol 2022; 76:694-725. [PMID: 34715263 DOI: 10.1016/j.jhep.2021.10.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/17/2021] [Indexed: 12/18/2022]
Abstract
As one of the most metabolically complex systems in the body, the liver ensures multi-organ homeostasis and ultimately sustains life. Nevertheless, during early postnatal development, the liver is highly immature and takes about 2 years to acquire and develop almost all of its functions. Different events occurring at the environmental and cellular levels are thought to mediate hepatic maturation and function postnatally. The crosstalk between the liver, the gut and its microbiome has been well appreciated in the context of liver disease, but recent evidence suggests that the latter could also be critical for hepatic function under physiological conditions. The gut-liver crosstalk is thought to be mediated by a rich repertoire of microbial metabolites that can participate in a myriad of biological processes in hepatic sinusoids, from energy metabolism to tissue regeneration. Studies on germ-free animals have revealed the gut microbiome as a critical contributor in early hepatic programming, and this influence extends throughout life, mediating liver function and body homeostasis. In this seminar, we describe the microbial molecules that have a known effect on the liver and discuss how the gut microbiome and the liver evolve throughout life. We also provide insights on current and future strategies to target the gut microbiome in the context of hepatology research.
Collapse
Affiliation(s)
- Joana I Almeida
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Miguel F Tenreiro
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Lucía Martinez-Santamaria
- Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, ISCIII), Madrid, Spain; Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| | - Sara Guerrero-Aspizua
- Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, ISCIII), Madrid, Spain
| | - Javier P Gisbert
- Gastroenterology Department. Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid (UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Paula M Alves
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Margarida Serra
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Pedro M Baptista
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Fundación ARAID, Zaragoza, Spain.
| |
Collapse
|
312
|
Han H, Xiao H, Wu Z, Liu L, Chen M, Gu H, Wang H, Chen L. The miR-98-3p/JAG1/Notch1 axis mediates the multigenerational inheritance of osteopenia caused by maternal dexamethasone exposure in female rat offspring. Exp Mol Med 2022; 54:298-308. [PMID: 35332257 PMCID: PMC8979986 DOI: 10.1038/s12276-022-00743-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/28/2021] [Accepted: 12/23/2021] [Indexed: 11/09/2022] Open
Abstract
As a synthetic glucocorticoid, dexamethasone is widely used to treat potential premature delivery and related diseases. Our previous studies have shown that prenatal dexamethasone exposure (PDE) can cause bone dysplasia and susceptibility to osteoporosis in female rat offspring. However, whether the effect of PDE on bone development can be extended to the third generation (F3 generation) and its multigenerational mechanism of inheritance have not been reported. In this study, we found that PDE delayed fetal bone development and reduced adult bone mass in female rat offspring of the F1 generation, and this effect of low bone mass caused by PDE even continued to the F2 and F3 generations. Furthermore, we found that PDE increases the expression of miR-98-3p but decreases JAG1/Notch1 signaling in the bone tissue of female fetal rats. Moreover, the expression changes of miR-98-3p/JAG1/Notch1 caused by PDE continued from the F1 to F3 adult offspring. Furthermore, the expression levels of miR-98-3p in oocytes of the F1 and F2 generations were increased. We also confirmed that dexamethasone upregulates the expression of miR-98-3p in vitro and shows targeted inhibition of JAG1/Notch1 signaling, leading to poor osteogenic differentiation of bone marrow mesenchymal stem cells. In conclusion, maternal dexamethasone exposure caused low bone mass in female rat offspring with a multigenerational inheritance effect, the mechanism of which is related to the inhibition of JAG1/Notch1 signaling caused by the continuous upregulation of miR-98-3p expression in bone tissues transmitted by F2 and F3 oocytes.
Collapse
Affiliation(s)
- Hui Han
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hao Xiao
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Zhixin Wu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Liang Liu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ming Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hanwen Gu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.,Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
313
|
Chen W, Yang H, Yan Q, Zhou X, Tan Z, Wang Z. Effects of maternal feed intake restriction on the blood parameters, fatty acid profile and lipogenetic genes expression of perirenal fat in offspring kids. Anim Reprod Sci 2022; 238:106955. [DOI: 10.1016/j.anireprosci.2022.106955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/16/2022] [Accepted: 02/20/2022] [Indexed: 11/17/2022]
|
314
|
Skakkebæk NE, Lindahl-Jacobsen R, Levine H, Andersson AM, Jørgensen N, Main KM, Lidegaard Ø, Priskorn L, Holmboe SA, Bräuner EV, Almstrup K, Franca LR, Znaor A, Kortenkamp A, Hart RJ, Juul A. Environmental factors in declining human fertility. Nat Rev Endocrinol 2022; 18:139-157. [PMID: 34912078 DOI: 10.1038/s41574-021-00598-8] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 12/14/2022]
Abstract
A severe decline in child births has occurred over the past half century, which will lead to considerable population declines, particularly in industrialized regions. A crucial question is whether this decline can be explained by economic and behavioural factors alone, as suggested by demographic reports, or to what degree biological factors are also involved. Here, we discuss data suggesting that human reproductive health is deteriorating in industrialized regions. Widespread infertility and the need for assisted reproduction due to poor semen quality and/or oocyte failure are now major health issues. Other indicators of declining reproductive health include a worldwide increasing incidence in testicular cancer among young men and alterations in twinning frequency. There is also evidence of a parallel decline in rates of legal abortions, revealing a deterioration in total conception rates. Subtle alterations in fertility rates were already visible around 1900, and most industrialized regions now have rates below levels required to sustain their populations. We hypothesize that these reproductive health problems are partially linked to increasing human exposures to chemicals originating directly or indirectly from fossil fuels. If the current infertility epidemic is indeed linked to such exposures, decisive regulatory action underpinned by unconventional, interdisciplinary research collaborations will be needed to reverse the trends.
Collapse
Affiliation(s)
- Niels E Skakkebæk
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | | | - Hagai Levine
- School of Public Health, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anna-Maria Andersson
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Niels Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Øjvind Lidegaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lærke Priskorn
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Stine A Holmboe
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Elvira V Bräuner
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Almstrup
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Luiz R Franca
- Department of Morphology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ariana Znaor
- Cancer Surveillance Branch, International Agency for Research on Cancer, Lyon, France
| | - Andreas Kortenkamp
- Division of Environmental Sciences, Brunel University London, Uxbridge, UK
| | - Roger J Hart
- Division of Obstetrics and Gynaecology, University of Western Australia, Perth, Western Australia, Australia
- Fertility Specialists of Western Australia, Bethesda Hospital, Claremont, Western Australia, Australia
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
315
|
Bebbere D, Coticchio G, Borini A, Ledda S. Oocyte aging: looking beyond chromosome segregation errors. J Assist Reprod Genet 2022; 39:793-800. [PMID: 35212880 PMCID: PMC9051005 DOI: 10.1007/s10815-022-02441-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/18/2022] [Indexed: 11/30/2022] Open
Abstract
The age-associated decline in female fertility is largely ascribable to a decrease in oocyte quality. This phenomenon is multifaceted and influenced by numerous interconnected maternal and environmental factors. An increase in the rate of meiotic errors is the major cause of the decline in oocyte developmental competence. However, abnormalities in the ooplasm accumulating with age - including altered metabolism, organelle dysfunction, and aberrant gene regulation - progressively undermine oocyte quality. Stockpiling of maternal macromolecules during folliculogenesis is crucial, as oocyte competence to achieve maturation, fertilization, and the earliest phases of embryo development occur in absence of transcription. At the same time, crucial remodeling of oocyte epigenetics during oogenesis is potentially exposed to interfering factors, such as assisted reproduction technologies (ARTs) or environmental changes, whose impact may be enhanced by reproductive aging. As the effects of maternal aging on molecular mechanisms governing the function of the human oocyte remain poorly understood, studies in animal models are essential to deepen current understanding, with translational implications for human ARTs. The present mini review aims at offering an updated and consistent view of cytoplasmic alterations occurring in oocytes during aging, focusing particularly on gene and epigenetic regulation. Appreciation of these mechanisms could inspire solutions to mitigate/control the phenomenon, and thus benefit modern ARTs.
Collapse
Affiliation(s)
- Daniela Bebbere
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy.
| | | | | | - Sergio Ledda
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| |
Collapse
|
316
|
Innocenti F, Fiorentino G, Cimadomo D, Soscia D, Garagna S, Rienzi L, Ubaldi FM, Zuccotti M. Maternal effect factors that contribute to oocytes developmental competence: an update. J Assist Reprod Genet 2022; 39:861-871. [PMID: 35165782 PMCID: PMC9051001 DOI: 10.1007/s10815-022-02434-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/09/2022] [Indexed: 11/30/2022] Open
Abstract
Oocyte developmental competence is defined as the capacity of the female gamete to be fertilized and sustain development to the blastocyst stage. Epigenetic reprogramming, a correct cell division pattern, and an efficient DNA damage response are all critical events that, before embryonic genome activation, are governed by maternally inherited factors such as maternal-effect gene (MEG) products. Although these molecules are stored inside the oocyte until ovulation and exert their main role during fertilization and preimplantation development, some of them are already functioning during folliculogenesis and oocyte meiosis resumption. This mini review summarizes the crucial roles played by MEGs during oocyte maturation, fertilization, and preimplantation development with a direct/indirect effect on the acquisition or maintenance of oocyte competence. Our aim is to inspire future research on a topic with potential clinical perspectives for the prediction and treatment of female infertility.
Collapse
Affiliation(s)
- Federica Innocenti
- GeneraLife IVF, Clinica Valle Giulia, via G. de Notaris, 2b, 00197, Rome, Italy
| | - Giulia Fiorentino
- Laboratory of Developmental Biology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy.,Center for Health Technologies, University of Pavia, Pavia, Italy
| | - Danilo Cimadomo
- GeneraLife IVF, Clinica Valle Giulia, via G. de Notaris, 2b, 00197, Rome, Italy.
| | - Daria Soscia
- GeneraLife IVF, Clinica Valle Giulia, via G. de Notaris, 2b, 00197, Rome, Italy
| | - Silvia Garagna
- Laboratory of Developmental Biology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy.,Center for Health Technologies, University of Pavia, Pavia, Italy
| | - Laura Rienzi
- GeneraLife IVF, Clinica Valle Giulia, via G. de Notaris, 2b, 00197, Rome, Italy
| | | | - Maurizio Zuccotti
- Laboratory of Developmental Biology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy.,Center for Health Technologies, University of Pavia, Pavia, Italy
| | | |
Collapse
|
317
|
Arand J, Chiang HR, Martin D, Snyder MP, Sage J, Reijo Pera RA, Wossidlo M. Tet enzymes are essential for early embryogenesis and completion of embryonic genome activation. EMBO Rep 2022; 23:e53968. [PMID: 34866320 PMCID: PMC8811641 DOI: 10.15252/embr.202153968] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022] Open
Abstract
Mammalian development begins in transcriptional silence followed by a period of widespread activation of thousands of genes. DNA methylation reprogramming is integral to embryogenesis and linked to Tet enzymes, but their function in early development is not well understood. Here, we generate combined deficiencies of all three Tet enzymes in mouse oocytes using a morpholino-guided knockdown approach and study the impact of acute Tet enzyme deficiencies on preimplantation development. Tet1-3 deficient embryos arrest at the 2-cell stage with the most severe phenotype linked to Tet2. Individual Tet enzymes display non-redundant roles in the consecutive oxidation of 5-methylcytosine to 5-carboxylcytosine. Gene expression analysis uncovers that Tet enzymes are required for completion of embryonic genome activation (EGA) and fine-tuned expression of transposable elements and chimeric transcripts. Whole-genome bisulfite sequencing reveals minor changes of global DNA methylation in Tet-deficient 2-cell embryos, suggesting an important role of non-catalytic functions of Tet enzymes in early embryogenesis. Our results demonstrate that Tet enzymes are key components of the clock that regulates the timing and extent of EGA in mammalian embryos.
Collapse
Affiliation(s)
- Julia Arand
- Center of Anatomy and Cell BiologyDepartment of Cell and Developmental BiologyMedical University of ViennaViennaAustria
- Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordCAUSA
- Department of PediatricsStanford UniversityStanfordCAUSA
- Department of GeneticsStanford UniversityStanfordCAUSA
| | - H Rosaria Chiang
- Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordCAUSA
- Department of GeneticsStanford UniversityStanfordCAUSA
- Department of Obstetrics & GynecologyStanford UniversityStanfordCAUSA
| | - David Martin
- Center of Anatomy and Cell BiologyDepartment of Cell and Developmental BiologyMedical University of ViennaViennaAustria
| | | | - Julien Sage
- Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordCAUSA
- Department of PediatricsStanford UniversityStanfordCAUSA
- Department of GeneticsStanford UniversityStanfordCAUSA
| | - Renee A Reijo Pera
- Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordCAUSA
- Department of Obstetrics & GynecologyStanford UniversityStanfordCAUSA
- Present address:
McLaughlin Research InstituteGreat FallsMTUSA
| | - Mark Wossidlo
- Center of Anatomy and Cell BiologyDepartment of Cell and Developmental BiologyMedical University of ViennaViennaAustria
- Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordCAUSA
- Department of GeneticsStanford UniversityStanfordCAUSA
- Department of Obstetrics & GynecologyStanford UniversityStanfordCAUSA
| |
Collapse
|
318
|
Yang Y, Zhang X, Lei Y, Chang G, Zou Y, Yu S, Wu H, Rong H, Lei Z, Xu C. The effects of H22 tumor on the quality of oocytes and the development of early embryos from host mice: A single-cell RNA sequencing approach. Theriogenology 2022; 179:45-59. [PMID: 34826707 DOI: 10.1016/j.theriogenology.2021.11.006] [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: 06/19/2021] [Revised: 10/30/2021] [Accepted: 11/13/2021] [Indexed: 10/19/2022]
Abstract
The association between cancer and female reproduction remains largely unknown. Here we investigated the quality of oocytes and the developmental potential of zygotes using H22 tumor-bearing mice model. The results showed that the number of oocytes was decreased in tumor-bearing mice compared with the control mice, and accompanied scattered chromosomes was observed. Further study revealed an abnormal epigenetic reprogramming occurred in the zygotes from the H22 tumor-bearing mice, as exemplified by the aberrant 5hmC/5mC modifications in the pronuclei. Finally, single-cell RNA sequencing was performed on the oocytes collected from the H22 tumor-bearing mice. Our data showed that 45 of the 202 differentially expressed genes in tumor-bearing group were closely associated with oocyte quality. Protein interaction analysis indicated that the potential interaction among these 45 genes. Collectively, our study uncovered that the quality of oocytes and early embryonic development were affected by H22 tumor bearing via the altered expression patterns of genes related with reproduction, providing new insights into the reproductive capability of female cancer patients.
Collapse
Affiliation(s)
- Yanhong Yang
- The First Affiliated Hospital (School of Clinical Medicine), Guangdong Pharmaceutical University, Nong-Lin-Xia Road 19#, Yue-Xiu District, Guangzhou, 510080, PR China
| | - Xueying Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, PR China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China
| | - Yuting Lei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, PR China
| | - Gang Chang
- Department of Biochemistry and Molecular Biology, Shenzhen University Health Science Center, Shenzhen, 518060, PR China
| | - Yan Zou
- The Reproductive Medical Center of Nanning Second People's Hospital, Nanning, 530031, PR China
| | - Siping Yu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, PR China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China
| | - Huijuan Wu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, PR China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China
| | - Hedong Rong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, PR China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, 510006, PR China
| | - Zili Lei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou, 510006, PR China.
| | - Changlong Xu
- The Reproductive Medical Center of Nanning Second People's Hospital, Nanning, 530031, PR China.
| |
Collapse
|
319
|
Moghadam ARE, Moghadam MT, Hemadi M, Saki G. Oocyte quality and aging. JBRA Assist Reprod 2022; 26:105-122. [PMID: 34338482 PMCID: PMC8769179 DOI: 10.5935/1518-0557.20210026] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 04/15/2021] [Indexed: 11/20/2022] Open
Abstract
It is well known that female reproduction ability decreases during the forth decade of life due to age-related changes in oocyte quality and quantity; although the number of women trying to conceive has today increased remarkably between the ages of 36 to 44. The causes of reproductive aging and physiological aspects of this phenomenon are still elusive. With increase in the women's age, during Assisted Reproductive Technologies (ART) we have perceived a significant decline in the number and quality of retrieved oocytes, as well as in ovarian follicle reserves. This is because of increased aneuploidy due to factors such as spindle apparatus disruption; oxidative stress and mitochondrial damage. The aim of this review paper is to study data on the potential role of the aging process impacting oocyte quality and female reproductive ability. We present the current evidence that show the decreased oocyte quality with age, related to reductions in female reproductive outcome. The aging process is complicated and it is caused by many factors that control cellular and organism life span. Although the factors responsible for reduced oocyte quality remain unknown, the present review focuses on the potential role of ovarian follicle environment, oocyte structure and its organelles. To find a way to optimize oocyte quality and ameliorate clinical outcomes for women with aging-related causes of infertility.
Collapse
Affiliation(s)
- Ali Reza Eftekhari Moghadam
- Cellular & Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Anatomical Science, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahin Taheri Moghadam
- Department of Anatomical Science, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Hemadi
- Department of Anatomical Science, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ghasem Saki
- Department of Anatomical Science, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
320
|
Kuroda K, Katagiri Y, Ishihara O. Optimal individualization of patient-oriented ovarian stimulation in Japanese assisted reproductive technology clinics, a review for unique setting with advanced-age patients. J Obstet Gynaecol Res 2022; 48:521-532. [PMID: 35026870 DOI: 10.1111/jog.15150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/13/2021] [Accepted: 01/03/2022] [Indexed: 11/28/2022]
Abstract
Ovarian stimulation is a key issue in assisted reproductive technology (ART) treatment. ART practice in Japan is unique with various types of ovarian stimulation protocols, which may contribute to lower pregnancy rates compared to other countries. This review aims to clarify optimal individualized ovarian stimulation for improving the pregnancy rate per one oocyte retrieval cycle in Japan. We performed a literature review to describe ovarian stimulation, classification of infertile women depending on ovarian reserve and response, and Japanese ART data and discussed optimal conventional and mild ovarian stimulation protocols in Japan. According to Japanese ART registry data, the live birth rate of 30-35-year-old women was 32%-37% per ET cycle; therefore, four to five embryos are calculatedly needed when aiming a cumulative live birth rate of ≥80%. Mild stimulation aimed at collecting 5-10 oocytes can be alternative choice as an optimal ovarian stimulation protocol in young women. In 40-year-old women, the live birth rate is 18.8%, resulting in eight or more embryos as necessary. Conventional stimulation must be required in women with advanced age. In poor responders, however, mild stimulation may be sufficient for maximumly extracting their ovarian function. In Japan, mild ovarian stimulation can be selected in patients with a good prognosis and poor responders; however, conventional ovarian stimulation is necessary for women in advanced age.
Collapse
Affiliation(s)
- Keiji Kuroda
- Center for Reproductive Medicine and Implantation Research, Sugiyama Clinic Shinjuku, Tokyo, Japan.,Department of Obstetrics and Gynecology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yukiko Katagiri
- Department of Obstetrics and Gynecology, Faculty of Medicine, Toho University, Tokyo, Japan
| | - Osamu Ishihara
- Department of Obstetrics and Gynecology, Saitama Medical University, Saitama, Japan
| |
Collapse
|
321
|
Cait J, Cait A, Scott RW, Winder CB, Mason GJ. Conventional laboratory housing increases morbidity and mortality in research rodents: results of a meta-analysis. BMC Biol 2022; 20:15. [PMID: 35022024 PMCID: PMC8756709 DOI: 10.1186/s12915-021-01184-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/07/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Over 120 million mice and rats are used annually in research, conventionally housed in shoebox-sized cages that restrict natural behaviours (e.g. nesting and burrowing). This can reduce physical fitness, impair thermoregulation and reduce welfare (e.g. inducing abnormal stereotypic behaviours). In humans, chronic stress has biological costs, increasing disease risks and potentially shortening life. Using a pre-registered protocol ( https://atrium.lib.uoguelph.ca/xmlui/handle/10214/17955 ), this meta-analysis therefore tested the hypothesis that, compared to rodents in 'enriched' housing that better meets their needs, conventional housing increases stress-related morbidity and all-cause mortality. RESULTS Comprehensive searches (via Ovid, CABI, Web of Science, Proquest and SCOPUS on May 24 2020) yielded 10,094 publications. Screening for inclusion criteria (published in English, using mice or rats and providing 'enrichments' in long-term housing) yielded 214 studies (within 165 articles, using 6495 animals: 59.1% mice; 68.2% male; 31.8% isolation-housed), and data on all-cause mortality plus five experimentally induced stress-sensitive diseases: anxiety, cancer, cardiovascular disease, depression and stroke. The Systematic Review Center for Laboratory animal Experimentation (SYRCLE) tool assessed individual studies' risks of bias. Random-effects meta-analyses supported the hypothesis: conventional housing significantly exacerbated disease severity with medium to large effect sizes: cancer (SMD = 0.71, 95% CI = 0.54-0.88); cardiovascular disease (SMD = 0.72, 95% CI = 0.35-1.09); stroke (SMD = 0.87, 95% CI = 0.59-1.15); signs of anxiety (SMD = 0.91, 95% CI = 0.56-1.25); signs of depression (SMD = 1.24, 95% CI = 0.98-1.49). It also increased mortality rates (hazard ratio = 1.48, 95% CI = 1.25-1.74; relative median survival = 0.91, 95% CI = 0.89-0.94). Meta-regressions indicated that such housing effects were ubiquitous across species and sexes, but could not identify the most impactful improvements to conventional housing. Data variability (assessed via coefficient of variation) was also not increased by 'enriched' housing. CONCLUSIONS Conventional housing appears sufficiently distressing to compromise rodent health, raising ethical concerns. Results also add to previous work to show that research rodents are typically CRAMPED (cold, rotund, abnormal, male-biased, poorly surviving, enclosed and distressed), raising questions about the validity and generalisability of the data they generate. This research was funded by NSERC, Canada.
Collapse
Affiliation(s)
- Jessica Cait
- Department of Integrative Biology, College of Biological Science, University of Guelph, Guelph, Ontario, Canada
| | - Alissa Cait
- Department of Translational Immunology, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - R Wilder Scott
- School of Biomedical Engineering, Faculty of Medicine and Applied Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Charlotte B Winder
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Georgia J Mason
- Department of Integrative Biology, College of Biological Science, University of Guelph, Guelph, Ontario, Canada.
| |
Collapse
|
322
|
Wu D, Shi H, Yu Y, Yu T, Zhai J. Comparison of the Effectiveness of Various Medicines in the Prevention of Ovarian Hyperstimulation Syndrome: A Network Meta-Analysis of Randomized Controlled Trials. Front Endocrinol (Lausanne) 2022; 13:808517. [PMID: 35154015 PMCID: PMC8825486 DOI: 10.3389/fendo.2022.808517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Previous studies have described the effects of different drugs in preventing ovarian hyperstimulation syndrome (OHSS). However, the efficacies of those drugs in preventing OHSS remain inconclusive. METHODS We searched the PubMed, Web of Science, Embase, and Cochrane Central Register of Controlled Trials (CENTRAL) databases. A network meta-analysis of randomized controlled trials (RCTs) was performed up to August 2021. We investigated the following drugs in our study: aspirin, albumin, metformin, calcium, cabergoline, quinagolide, letrozole, hydroxyethyl starch (HES), and glucocorticoids. The primary outcome was the incidence rate of moderate-to-severe OHSS, with the results presented as risk ratios (RRs) with 95% confidence intervals (CIs). RESULTS The incidence of moderate-to-severe OHSS was significantly reduced by calcium administration (risk ratios [RR] 0.14, 95% confidence interval [CI]: 0.04, 0.46) (grade: high), HES (RR 0.25, 95% CI 0.07, 0.73) (grade: high), and cabergoline (RR 0.43, 95% CI 0.24, 0.71) (grade: moderate). The surface under the cumulative ranking curve (SUCRA) indicated that calcium (SUCRA, 92.4%) was the most effective intervention for preventing moderate-to-severe OHSS. These drugs were safe and did not affect clinical pregnancy, miscarriage, or live birth rates. CONCLUSION Calcium, HES, and cabergoline could effectively and safely prevent moderate-to-severe OHSS, with calcium as the most effective intervention.
Collapse
Affiliation(s)
- Di Wu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hao Shi
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiping Yu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ting Yu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Zhai
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Jun Zhai,
| |
Collapse
|
323
|
SATO K. Multiple roles of endocytosis and autophagy in intracellular remodeling during oocyte-to-embryo transition. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2022; 98:207-221. [PMID: 35545527 PMCID: PMC9130481 DOI: 10.2183/pjab.98.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/15/2022] [Indexed: 06/15/2023]
Abstract
Fertilization is the starting point for creating new progeny. At this time, the highly differentiated oocyte and sperm fuse to form one zygote, which is then converted into a pluripotent early embryo. Recent studies have shown that the lysosomal degradation system via autophagy and endocytosis plays important roles in the remodeling of intracellular components during oocyte-to-embryo transition. For example, in Caenorhabditis elegans, zygotes show high endocytic activity, and some populations of maternal membrane proteins are selectively internalized and delivered to lysosomes for degradation. Furthermore, fertilization triggers selective autophagy of sperm-derived paternal mitochondria, which establishes maternal inheritance of mitochondrial DNA. In addition, it has been shown that autophagy via liquid-liquid phase separation results in the selective degradation of some germ granule components, which are distributed to somatic cells of early embryos. This review outlines the physiological functions of the lysosomal degradation system and its molecular mechanisms in C. elegans and mouse embryos.
Collapse
Affiliation(s)
- Ken SATO
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| |
Collapse
|
324
|
Qi L, Liu YP, Wang SM, Shi H, Chen XL, Wang NN, Su YC. Abnormal BMI in Male and/or Female Partners Are Deleterious for Embryonic Development and Pregnancy Outcome During ART Process: A Retrospective Study. Front Endocrinol (Lausanne) 2022; 13:856667. [PMID: 35528007 PMCID: PMC9068983 DOI: 10.3389/fendo.2022.856667] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE To investigate the effect of BMI in male and/or female partners on embryo development and clinical pregnancy outcome during ART. METHODS Data of 11,130 cycles between January 2018 and December 2020 were retrospectively analyzed. They were divided into Group A, B, C, and D based on couples' BMI values, also were divided into Group I, II, III and IV in IVF cycles and Group i, ii, iii, and iv in ICSI cycles. After grouping, inter-group indicators comparisons and logistic regression analysis were performed. RESULTS In IVF cycles, CPR in Group I and Group III were higher than Group IV. In Group III, it was higher than Group II. The AR in Group IV was higher, but the LBR was lower than Group I, Group II, and Group III. Logistic regression analysis results suggested that AR in Group IV was higher than that in Group I in IVF cycles, whereas LBR was lower.In ICSI cycles, high-quality embryo rate in Group i and Group ii were both higher than that in Group iii and Group iv. The CPR in Group i was higher than Group ii and Group iv, and in Group iii was higher than Group ii and Group iv. The AR in Group i was lower than Group iii and Group iv, and AR in Group ii was lower than Group iv. LBR, in Group I it was higher than Group ii, Group iii, and Group iv. Logistic regression analysis results suggested CPR in Group ii was significantly lower than that in Group i. AR in Group iii was considerably higher than that in Group i. LBR in Group ii and Group iv were significantly lower than that in Group i. CONCLUSION Female higher BMI was not conducive to the formation of high-quality embryos in ICSI cycle. Female and/or male BMI affected AR and LBR more than CPR not only in IVF cycles, but also in ICSI cycles.
Collapse
|
325
|
Repalle D, Saritha KV, Bhandari S, Chittora M, Choudhary J. Role of Female Age in Regulating the Effect of Sperm DNA Fragmentation on the Live Birth Rates in Intracytoplasmic Sperm Injection Cycles with Own and Donor Oocytes. J Hum Reprod Sci 2022; 15:64-71. [PMID: 35494199 PMCID: PMC9053350 DOI: 10.4103/jhrs.jhrs_150_21] [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: 10/23/2021] [Revised: 02/27/2022] [Accepted: 02/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background Sperm DNA integrity assessment has been progressively used as an unfettered measure of sperm as it proffers more prognostic and diagnostic information than routine semen analysis. The contentious effect of sperm DNA fragmentation (SDF) on clinical outcomes can be attributed to female factors such as age, oocyte quality and ovarian reserve. Aims The study is mainly aimed to know the influence of SDF on the live birth rates in intracytoplasmic sperm injection (ICSI) cycles with own and donor oocytes. Second, to know the role of female age in regulating the effect of SDF on the live birth rates in ICSI cycles with own and donor oocytes. Setting and Design A prospective cohort study was done at our tertiary care centre attached to the reproductive medicine unit in medical college. Materials and Methods The study included 356 patients who underwent first ICSI cycles either with own or donor-oocytes along with day 5 fresh embryo transfers only. The main outcome measures were live birth rates and miscarriage rates. Statistical Analysis Used Chi-squared test was used to compare the categorical variables between the groups. The receiver operating characteristic curve was developed to correlate the female age with the live birth rate. Results A significant decrease in the live birth rates (42.85% vs. 26.15%, P = 0.023) and an increase in the miscarriage rates (12.30% vs. 34.61%, P = 0.013) were observed in the high-SDF group ICSI cycles of own-oocyte patients. However, there was no significant difference in the live birth rates and miscarriage rates in the low- and high-SDF groups of donor oocyte ICSI cycle patients (P > 0.05). The own-oocyte ICSI cycle patients were further stratified based on the female age. In the female age group ≤30 years there was no significant difference in the live birth and miscarriage rates (P > 0.05) similar to donor oocyte ICSI cycles. Whereas, there was a significant difference in the live birth rates in the females of age >30 years (13.79% vs. 34.37%, P = 0.040). Conclusion In conclusion, high-SDF has a negative influence on the live birth rates and a positive influence on the miscarriage rates in patients with own-oocyte ICSI cycles. A similar influence was not observed in patients with donor-oocyte ICSI cycles and in young female patients (age ≤30 years) with own-oocyte ICSI cycles.
Collapse
Affiliation(s)
- Deepthi Repalle
- Department of Reproductive Medicine and Surgery, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh, India
| | - K. V. Saritha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh, India
| | - Shilpa Bhandari
- Department of Reproductive Medicine and Surgery, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Megha Chittora
- Department of Reproductive Medicine and Surgery, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| | - Jitendra Choudhary
- Department of Reproductive Medicine and Surgery, Sri Aurobindo Institute of Medical Sciences, Indore, Madhya Pradesh, India
| |
Collapse
|
326
|
Panera N, Mandato C, Crudele A, Bertrando S, Vajro P, Alisi A. Genetics, epigenetics and transgenerational transmission of obesity in children. Front Endocrinol (Lausanne) 2022; 13:1006008. [PMID: 36452324 PMCID: PMC9704419 DOI: 10.3389/fendo.2022.1006008] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022] Open
Abstract
Sedentary lifestyle and consumption of high-calorie foods have caused a relentless increase of overweight and obesity prevalence at all ages. Its presently epidemic proportion is disquieting due to the tight relationship of obesity with metabolic syndrome and several other comorbidities which do call for urgent workarounds. The usual ineffectiveness of present therapies and failure of prevention campaigns triggered overtime a number of research studies which have unveiled some relevant aspects of obesity genetic and epigenetic inheritable profiles. These findings are revealing extremely precious mainly to serve as a likely extra arrow to allow the clinician's bow to achieve still hitherto unmet preventive goals. Evidence now exists that maternal obesity/overnutrition during pregnancy and lactation convincingly appears associated with several disorders in the offspring independently of the transmission of a purely genetic predisposition. Even the pre-conception direct exposure of either father or mother gametes to environmental factors can reprogram the epigenetic architecture of cells. Such phenomena lie behind the transfer of the obesity susceptibility to future generations through a mechanism of epigenetic inheritance. Moreover, a growing number of studies suggests that several environmental factors such as maternal malnutrition, hypoxia, and exposure to excess hormones and endocrine disruptors during pregnancy and the early postnatal period may play critical roles in programming childhood adipose tissue and obesity. A deeper understanding of how inherited genetics and epigenetics may generate an obesogenic environment at pediatric age might strengthen our knowledge about pathogenetic mechanisms and improve the clinical management of patients. Therefore, in this narrative review, we attempt to provide a general overview of the contribution of heritable genetic and epigenetic patterns to the obesity susceptibility in children, placing a particular emphasis on the mother-child dyad.
Collapse
Affiliation(s)
- Nadia Panera
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Claudia Mandato
- Pediatrics Section, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Salermo, Italy
- *Correspondence: Anna Alisi, ; Claudia Mandato,
| | - Annalisa Crudele
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Sara Bertrando
- Pediatrics Clinic, San Giovanni di Dio e Ruggi d’Aragona University Hospital, Salerno, Italy
| | - Pietro Vajro
- Pediatrics Section, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Salermo, Italy
| | - Anna Alisi
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- *Correspondence: Anna Alisi, ; Claudia Mandato,
| |
Collapse
|
327
|
Liu M, Zhao X, Peng Y, Zheng J, Guo K, Fan Y, Jiang L, Yang A, Cui N, Hao G, Wang W. Outcomes After a Single Ovarian Stimulation Cycle in Women of Advanced Reproductive Age: A Retrospective Analysis. Front Endocrinol (Lausanne) 2022; 13:792159. [PMID: 35237234 PMCID: PMC8882593 DOI: 10.3389/fendo.2022.792159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 01/11/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Previous studies showed that age is the most important factor that determines the outcome after embryo transfer (ET), with either in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI), regardless whether fresh or frozen cycles. The average cumulative live birth rate (CLBR) following a single ovarian stimulation cycle in women of advanced reproductive age (≥38 years of age) has been reported to be 22.6-34.1%. The purpose of this study is to compare the CLBR after a single ovarian stimulation cycle in women of different advanced reproductive age bracket (38/39, 40/41, 42/43 years of age or older), and to explore the factors (e.g., age, type of infertility, body mass index (BMI), ovarian stimulation protocols) associated with CLBR. METHODS This retrospective analysis included all women of advanced reproductive age (38 years or older) undergoing IVF or ICSI at authors' institute during a period from January 1, 2016 to December 31, 2018. The study protocol was approved by the Ethics Committee of the Second Hospital of Hebei Medical University (No. 2021-P045). Subjects with underlying diseases were excluded from analysis. The last follow-up was conducted in December 2020, with minimal 2-year follow-up. RESULTS The final analysis included 826 women (40.00 ± 2.10 years of age at the time of ovarian stimulation; n = 633 and 193 for IVF-ET and ICSI-ET, respectively). The number of women in each age bracket was: 424 for 38/39 y, 226 for 40/41 y, 118 for 42/43 y, and 58 for ≥44 y. The number of transferable embryos was 2 (interquartile range: 2,4) for 38/39 y, 2 (2,3) for 40/41 y, 2 (2,3) for 42/43 y, and 2 (1.75,3) for ≥44 y. The rate of fresh embryo transfer was comparable (62.03-72.58%) among the 4 age brackets. The average CLBR following a single cycle was 26.27% in the overall study population, 32.31% for 38/39 y, 26.99% for 40/41 y, 14.4% for 42/43 y, and 3.44% for ≥44 y (P <0.001). In multivariate regression, CLBR was independently associated with younger age (OR for each year: 1.538, 95%CI: 1.193, 1.984) and higher number of transferable embryos (OR for each embryo: 1.495, 95%CI: 1.297, 1.722). CLBR differed significantly in the 38/39 group (P = 0.014), with higher rate in women receiving the Gonadotropin-releasing hormone agonist (GnRH-a) long or GnRH-a ultra-long protocols. CONCLUSIONS Forty-two years of age seemed to be a critical cutoff to achieve reasonable level of CLBR after a single ovarian stimulation cycle in women of advanced reproductive age.
Collapse
Affiliation(s)
- Mengdi Liu
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Cangzhou People's Hospital, Cangzhou, China
| | - Xusheng Zhao
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Handan Women and Children's Hospital, Handan, China
| | - Yuanyuan Peng
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiahua Zheng
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Kaixuan Guo
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yanli Fan
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Jiang
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Aimin Yang
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Na Cui
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guimin Hao
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wei Wang
- The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
328
|
Schatten H. Centrosomes in Reproduction. THE CENTROSOME AND ITS FUNCTIONS AND DYSFUNCTIONS 2022; 235:55-73. [DOI: 10.1007/978-3-031-20848-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
329
|
Kiyozumi D, Ikawa M. Proteolysis in Reproduction: Lessons From Gene-Modified Organism Studies. Front Endocrinol (Lausanne) 2022; 13:876370. [PMID: 35600599 PMCID: PMC9114714 DOI: 10.3389/fendo.2022.876370] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/28/2022] [Indexed: 12/17/2022] Open
Abstract
The physiological roles of proteolysis are not limited to degrading unnecessary proteins. Proteolysis plays pivotal roles in various biological processes through cleaving peptide bonds to activate and inactivate proteins including enzymes, transcription factors, and receptors. As a wide range of cellular processes is regulated by proteolysis, abnormalities or dysregulation of such proteolytic processes therefore often cause diseases. Recent genetic studies have clarified the inclusion of proteases and protease inhibitors in various reproductive processes such as development of gonads, generation and activation of gametes, and physical interaction between gametes in various species including yeast, animals, and plants. Such studies not only clarify proteolysis-related factors but the biological processes regulated by proteolysis for successful reproduction. Here the physiological roles of proteases and proteolysis in reproduction will be reviewed based on findings using gene-modified organisms.
Collapse
Affiliation(s)
- Daiji Kiyozumi
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Kawaguchi, Japan
| |
Collapse
|
330
|
Wang M, Zeng L, Su P, Ma L, Zhang M, Zhang YZ. Autophagy: a multifaceted player in the fate of sperm. Hum Reprod Update 2021; 28:200-231. [PMID: 34967891 PMCID: PMC8889000 DOI: 10.1093/humupd/dmab043] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/11/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Autophagy is an intracellular catabolic process of degrading and recycling proteins and organelles to modulate various physiological and pathological events, including cell differentiation and development. Emerging data indicate that autophagy is closely associated with male reproduction, especially the biosynthetic and catabolic processes of sperm. Throughout the fate of sperm, a series of highly specialized cellular events occur, involving pre-testicular, testicular and post-testicular events. Nonetheless, the most fundamental question of whether autophagy plays a protective or harmful role in male reproduction, especially in sperm, remains unclear. OBJECTIVE AND RATIONALE We summarize the functional roles of autophagy in the pre-testicular (hypothalamic–pituitary–testis (HPG) axis), testicular (spermatocytogenesis, spermatidogenesis, spermiogenesis, spermiation) and post-testicular (sperm maturation and fertilization) processes according to the timeline of sperm fate. Additionally, critical mechanisms of the action and clinical impacts of autophagy on sperm are identified, laying the foundation for the treatment of male infertility. SEARCH METHODS In this narrative review, the PubMed database was used to search peer-reviewed publications for summarizing the functional roles of autophagy in the fate of sperm using the following terms: ‘autophagy’, ‘sperm’, ‘hypothalamic–pituitary–testis axis’, ‘spermatogenesis’, ‘spermatocytogenesis’, ‘spermatidogenesis’, ‘spermiogenesis’, ‘spermiation’, ‘sperm maturation’, ‘fertilization’, ‘capacitation’ and ‘acrosome’ in combination with autophagy-related proteins. We also performed a bibliographic search for the clinical impact of the autophagy process using the keywords of autophagy inhibitors such as ‘bafilomycin A1’, ‘chloroquine’, ‘hydroxychloroquine’, ‘3-Methyl Adenine (3-MA)’, ‘lucanthone’, ‘wortmannin’ and autophagy activators such as ‘rapamycin’, ‘perifosine’, ‘metformin’ in combination with ‘disease’, ‘treatment’, ‘therapy’, ‘male infertility’ and equivalent terms. In addition, reference lists of primary and review articles were reviewed for additional relevant publications. All relevant publications until August 2021 were critically evaluated and discussed on the basis of relevance, quality and timelines. OUTCOMES (i) In pre-testicular processes, autophagy-related genes are involved in the regulation of the HPG axis; and (ii) in testicular processes, mTORC1, the main gate to autophagy, is crucial for spermatogonia stem cell (SCCs) proliferation, differentiation, meiotic progression, inactivation of sex chromosomes and spermiogenesis. During spermatidogenesis, autophagy maintains haploid round spermatid chromatoid body homeostasis for differentiation. During spermiogenesis, autophagy participates in acrosome biogenesis, flagella assembly, head shaping and the removal of cytoplasm from elongating spermatid. After spermatogenesis, through PDLIM1, autophagy orchestrates apical ectoplasmic specialization and basal ectoplasmic specialization to handle cytoskeleton assembly, governing spermatid movement and release during spermiation. In post-testicular processes, there is no direct evidence that autophagy participates in the process of capacitation. However, autophagy modulates the acrosome reaction, paternal mitochondria elimination and clearance of membranous organelles during fertilization. WIDER IMPLICATIONS Deciphering the roles of autophagy in the entire fate of sperm will provide valuable insights into therapies for diseases, especially male infertility.
Collapse
Affiliation(s)
- Mei Wang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China.,Harvard Reproductive Endocrine Science Center and Reproductive Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, P.R. China
| | - Ling Zeng
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Ping Su
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Ling Ma
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China.,Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, P.R. China
| | - Ming Zhang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China.,Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, P.R. China
| | - Yuan Zhen Zhang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China.,Clinical Medicine Research Center of Prenatal Diagnosis and Birth Health in Hubei Province, Wuhan, Hubei, P.R. China
| |
Collapse
|
331
|
Li C, Cao M, Zhou X. Role of epigenetics in parturition and preterm birth. Biol Rev Camb Philos Soc 2021; 97:851-873. [PMID: 34939297 DOI: 10.1111/brv.12825] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 12/19/2022]
Abstract
Preterm birth occurs worldwide and is associated with high morbidity, mortality, and economic cost. Although several risk factors associated with parturition and preterm birth have been identified, mechanisms underlying this syndrome remain unclear, thereby limiting the implementation of interventions for prevention and management. Known triggers of preterm birth include conditions related to inflammatory and immunological pathways, as well as genetics and maternal history. Importantly, epigenetics, which is the study of heritable phenotypic changes that occur without alterations in the DNA sequence, may play a role in linking social and environmental risk factors for preterm birth. Epigenetic approaches to the study of preterm birth, including analyses of the effects of microRNAs, long non-coding RNAs, DNA methylation, and histone modification, have contributed to an improved understanding of the molecular bases of both term and preterm birth. Additionally, epigenetic modifications have been linked to factors already associated with preterm birth, including obesity and smoking. The prevention and management of preterm birth remains a challenge worldwide. Although epigenetic analysis provides valuable insights into the causes and risk factors associated with this syndrome, further studies are necessary to determine whether epigenetic approaches can be used routinely for the diagnosis, prevention, and management of preterm birth.
Collapse
Affiliation(s)
- Chunjin Li
- College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, China
| | - Maosheng Cao
- College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, China
| | - Xu Zhou
- College of Animal Sciences, Jilin University, Changchun, Jilin, 130062, China
| |
Collapse
|
332
|
Cui J, Wu F, Yang X, Liu S, Han S, Chen B. Effects of ammonia on hypothalamic-pituitary-ovarian axis in female rabbits. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 227:112922. [PMID: 34700170 DOI: 10.1016/j.ecoenv.2021.112922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND As one of the most harmful gases in the livestock house, ammonia is recognized as an environmental stressor by Environmental Protection Agency (United States). The study aimed to explore the effect of ammonia on hypothalamic-pituitary-ovarian (HPO) axis of rabbits. A total of ninety two-month-old female IRA rabbits were randomly divided into three groups, and were kept in animal environment control rooms for four weeks at college of animal science and technology, Hebei Agricultural University (Baoding, China). The rabbits in the control group were kept under ammonia concentration of < 3 ppm. The two treatment groups were kept under ammonia concentration of 30 ppm and 50 ppm. Hypothalamus, pituitary, and ovary were collected for hematoxylin and eosin (HE) staining, immunohistochemistry, terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling (TUNEL) and quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Serum was collected for enzyme-linked immunosorbent assay (ELISA). RESULTS Histopathological examination revealed that exposed to excess ammonia damaged the morphology and structure of hypothalamus, pituitary, and ovary. TUNEL assay revealed that apoptosis rate increased in hypothalamus, pituitary, and ovary. The protein expression levels of Bcl-2associated X protein (Bax) and Caspase-9 increased, while B-cell lymphoma-2 (Bcl-2) decreased, resulting in apoptosis. Moreover, the concentration of gonadotropin-releasing hormone (GnRH), follicle-stimulating hormone (FSH), luteinizing hormone (LH), estradiol (E2), and progesterone (PROG) reduced in plasma. The mRNA expression of FSH and LH in pituitary and follicle-stimulating hormone receptor (FSHR), E2, PROG in ovary as well as decreased, indicated hormone secretion disorder. CONCLUSIONS The results indicated that ammonia exposure damaged hypothalamus, pituitary, and ovary, caused hormone secretion disorder and apoptosis.
Collapse
Affiliation(s)
- Jia Cui
- Department of Animal Science and Technology, Hebei Agricultural University, Baoding, China.
| | - Fengyang Wu
- Department of Animal Science and Technology, Hebei Agricultural University, Baoding, China.
| | - Xinyu Yang
- Department of Animal Science and Technology, Hebei Agricultural University, Baoding, China.
| | - Shudong Liu
- Department of Animal Science and Technology, Hebei Agricultural University, Baoding, China.
| | - Shuaijuan Han
- Department of Animal Science and Technology, Hebei Agricultural University, Baoding, China.
| | - Baojiang Chen
- Department of Animal Science and Technology, Hebei Agricultural University, Baoding, China.
| |
Collapse
|
333
|
Dynamics of Known Long Non-Coding RNAs during the Maternal-to-Zygotic Transition in Rabbit. Animals (Basel) 2021; 11:ani11123592. [PMID: 34944367 PMCID: PMC8698111 DOI: 10.3390/ani11123592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/05/2021] [Accepted: 12/14/2021] [Indexed: 01/06/2023] Open
Abstract
The control of pre-implantation development in mammals undergoes a maternal-to-zygotic transition (MZT) after fertilization. The transition involves maternal clearance and zygotic genome activation remodeling the terminal differentiated gamete to confer totipotency. In the study, we first determined the profile of long non-coding RNAs (lncRNAs) of mature rabbit oocyte, 2-cell, 4-cell, 8-cell, and morula embryos using RNA-seq. A total of 2673 known rabbit lncRNAs were identified. The lncRNAs exhibited dynamic expression patterns during pre-implantation development. Moreover, 107 differentially expressed lncRNAs (DE lncRNAs) were detected between mature oocyte and 2-cell embryo, while 419 DE lncRNAs were detected between 8-cell embryo and morula, consistent with the occurrence of minor and major zygotic genome activation (ZGA) wave of rabbit pre-implanted embryo. This study then predicted the potential target genes of DE lncRNAs based on the trans-regulation mechanism of lncRNAs. The GO and KEGG analyses showed that lncRNAs with stage-specific expression patterns promoted embryo cleavage and synchronic development by regulating gene transcription and translation, intracellular metabolism and organelle organization, and intercellular signaling transduction. The correlation analysis between mRNAs and lncRNAs identified that lncRNAs ENSOCUG00000034943 and ENSOCUG00000036338 may play a vital role in the late-period pre-implantation development by regulating ILF2 gene. This study also found that the sequential degradation of maternal lncRNAs occurred through maternal and zygotic pathways. Furthermore, the function analysis of the late-degraded lncRNAs suggested that these lncRNAs may play a role in the mRNA degradation in embryos via mRNA surveillance pathway. Therefore, this work provides a global view of known lncRNAs in rabbit pre-implantation development and highlights the role of lncRNAs in embryogenesis regulation.
Collapse
|
334
|
Korkidakis A, Au J, Albert A, Havelock J. Higher blastocyst implantation in frozen versus fresh embryo transfers in good prognosis patients. Minerva Obstet Gynecol 2021; 73:776-781. [PMID: 34905881 DOI: 10.23736/s2724-606x.21.04722-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND There is emerging evidence that frozen embryo transfers provide a more favorable environment for implantation as compared to fresh embryo transfers. Our objective was to determine if there is a clinical benefit to frozen versus fresh blastocyst transfers in good prognosis patients. METHODS Subjects undergoing their first or second IVF/ICSI cycle <38 years of age in an OCP pretreated GnRH antagonist stimulation protocol with supernumerary embryos available for blastocyst cryopreservation were eligible for analysis. Primary transfer was exclusively blastocyst transfer. Exclusion criteria consisted of rescue ICSI, preimplantation genetic testing, donor oocytes, and surrogacy. The cohort was divided into two groups based on whether they underwent a fresh vs. frozen primary transfer. The implantation rates were compared using mixed-effects logistic regression. The clinical pregnancy and live birth rates were compared using logistic regression adjusted for number of oocytes retrieved and number of embryos transferred. All models included age, reason for treatment, and number of prior births as covariates. RESULTS A total of 615 subjects were included in the study. There were no differences in the two groups with respect to age, BMI, baseline ovarian reserve testing, total gonadotropin dosage, and duration of stimulation. The implantation rate was higher in the frozen-embryo group as compared to the fresh-embryo group (59% and 48% respectively; OR 1.58; 95% CI 1.02-2.44). There was a trend towards higher clinical pregnancy and live birth rates in the frozen-embryo group. These differences persisted in the adjusted analysis. CONCLUSIONS Among good prognosis patients undergoing IVF, frozen embryo transfer was associated with improved implantation rates. Consideration should be given to primary frozen blastocyst transfer in this population.
Collapse
Affiliation(s)
- Ann Korkidakis
- Division of Reproductive Endocrinology and Infertility, University of British Columbia, Vancouver, BC, Canada -
| | - Jason Au
- Pacific Center for Reproductive Medicine, Burnaby, BC, Canada
| | - Arianne Albert
- Women's Health Research Institute, BC Women's Hospital and Health Center, Vancouver, BC, Canada
| | - Jon Havelock
- Division of Reproductive Endocrinology and Infertility, University of British Columbia, Vancouver, BC, Canada.,Women's Health Research Institute, BC Women's Hospital and Health Center, Vancouver, BC, Canada
| |
Collapse
|
335
|
Yang H, Kolben T, Meister S, Paul C, van Dorp J, Eren S, Kuhn C, Rahmeh M, Mahner S, Jeschke U, von Schönfeldt V. Factors Influencing the In Vitro Maturation (IVM) of Human Oocyte. Biomedicines 2021; 9:1904. [PMID: 34944731 PMCID: PMC8698296 DOI: 10.3390/biomedicines9121904] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023] Open
Abstract
In vitro maturation (IVM) of oocytes is a promising assisted reproductive technology (ART) deemed as a simple and safe procedure. It is mainly used in patients with impaired oocyte maturation and in fertility preservation for women facing the risk of losing fertility. However, to date, it is still not widely used in clinical practice because of its underperformance. The influencing factors, such as biphasic IVM system, culture medium, and the supplementation, have a marked effect on the outcomes of oocyte IVM. However, the role of different culture media, supplements, and follicular priming regimens in oocyte IVM have yet to be fully clarified and deserve further investigation.
Collapse
Affiliation(s)
- Huixia Yang
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
| | - Thomas Kolben
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
| | - Sarah Meister
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
| | - Corinna Paul
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
| | - Julia van Dorp
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
| | - Sibel Eren
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
| | - Christina Kuhn
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
- Department of Obstetrics and Gynecology, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Martina Rahmeh
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
- Department of Obstetrics and Gynecology, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Viktoria von Schönfeldt
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians-University, 81377 Munich, Germany; (H.Y.); (T.K.); (S.M.); (C.P.); (J.v.D.); (S.E.); (C.K.); (M.R.); (S.M.); (V.v.S.)
| |
Collapse
|
336
|
Sinha N, Lydia Walker G, Sen A. Looking at the Future Through the Mother's Womb: Gestational Diabetes and Offspring Fertility. Endocrinology 2021; 162:6379047. [PMID: 34597389 PMCID: PMC8520322 DOI: 10.1210/endocr/bqab209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Indexed: 12/12/2022]
Abstract
Altered nutrition or intrauterine exposure to various adverse conditions during fetal development or earlier in a mother's life can lead to epigenetic changes in fetal tissues, predisposing those tissues to diseases that manifest when offspring become adults. An example is a maternal obesity associated with gestational diabetes (GDM), where fetal exposure to a hyperglycemic, hyperinsulinemic, and/or hyperlipidemic gestational environment can provoke epigenetic changes that predispose offspring to various diseased conditions later in life. While it is now well established that offspring exposed to GDM have an increased risk of developing obesity, metabolic disorders, and/or cardiovascular disease in adult life, there are limited studies assessing the reproductive health of these offspring. This mini-review discusses the long-term effect of in utero exposure to GDM-associated adverse prenatal environment on the reproductive health of the offspring. Moreover, using evidence from various animal models and human epidemiological studies, this review offers molecular insight and understanding of how epigenetic reprogramming of genes culminates in reproductive dysfunction and the development of subfertility or infertility later in adult life.
Collapse
Affiliation(s)
- Niharika Sinha
- Reproductive and Developmental Sciences Program, East Lansing, Michigan 48824, USA
- Department of Animal Sciences, Michigan State University, East Lansing, Michigan 48824, USA
| | - Gretchen Lydia Walker
- Reproductive and Developmental Sciences Program, East Lansing, Michigan 48824, USA
- Department of Animal Sciences, Michigan State University, East Lansing, Michigan 48824, USA
| | - Aritro Sen
- Reproductive and Developmental Sciences Program, East Lansing, Michigan 48824, USA
- Department of Animal Sciences, Michigan State University, East Lansing, Michigan 48824, USA
- Correspondence: Aritro Sen, PhD, Reproductive and Developmental Sciences Program, 3013 Interdisciplinary Science & Technology Building, Michigan State University, 766 Service Rd, East Lansing, MI 48824, USA.
| |
Collapse
|
337
|
Wang Y, Wang X, Long Q, Liu Y, Yin T, Sirota I, Ren F, Gu Z, Luo J. Reducing embryonic mtDNA copy number alters epigenetic profile of key hepatic lipolytic genes and causes abnormal lipid accumulation in adult mice. FEBS J 2021; 288:6828-6843. [PMID: 34258867 DOI: 10.1111/febs.16121] [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: 03/23/2021] [Revised: 06/30/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022]
Abstract
Adverse fetal environment, in particular a shortage or excess of nutrients, is associated with increased risks of metabolic diseases later in life. However, the molecular mechanisms underlying this developmental origin of adult diseases remain unclear. Here, we directly tested the role of mitochondrial stress in mediating fetal programming in mice by enzymatically depleting mtDNA in zygotes. mtDNA-targeted plasmid microinjection is used to reduce embryonic mtDNA copy number directly, followed by embryo transfer. Mice with reduced zygote mtDNA copy number were born morphologically normal and showed no accelerated body weight gain. However, at 5 months of age these mice showed markedly increased hepatic lipidosis and became glucose-intolerant. Hepatic mRNA and protein expressions of peroxisome proliferator-activated receptor α (Pparα), a key transcriptional regulator of lipid metabolism, were significantly decreased as a result of increased DNA methylation in its proximal regulatory region. These results indicate that perturbation of mitochondrial function around the periconceptional period causes hypermethylation and thus suppressed expression of PPARα in fetal liver, leading to impaired hepatic lipid metabolism. Our findings provide the first direct evidence that mitochondrial stress mediates epigenetic changes associated with fetal programming of adult diseases in a mammalian system.
Collapse
Affiliation(s)
- Yakun Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Xuan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Qiaoming Long
- Cam-Su Mouse Genomic Resource Center, Soochow University, China
| | - Yuanwu Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Tao Yin
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Inna Sirota
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Junjie Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
338
|
Chiaratti MR. Uncovering the important role of mitochondrial dynamics in oogenesis: impact on fertility and metabolic disorder transmission. Biophys Rev 2021; 13:967-981. [PMID: 35059021 PMCID: PMC8724343 DOI: 10.1007/s12551-021-00891-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Oocyte health is tightly tied to mitochondria given their role in energy production, metabolite supply, calcium (Ca2+) buffering, and cell death regulation, among others. In turn, mitochondrial function strongly relies on these organelle dynamics once cyclic events of fusion and fission (division) are required for mitochondrial turnover, positioning, content homogenization, metabolic flexibility, interaction with subcellular compartments, etc. Importantly, during oogenesis, mitochondria change their architecture from an "orthodox" elongated shape characterized by the presence of numerous transversely oriented cristae to a round-to-oval morphology containing arched and concentrically arranged cristae. This, along with evidence showing that mitochondrial function is kept quiescent during most part of oocyte development, suggests an important role of mitochondrial dynamics in oogenesis. To investigate this, recent works have downregulated/upregulated in oocytes the expression of key effectors of mitochondrial dynamics, including mitofusins 1 (MFN1) and 2 (MFN2) and the dynamin-related protein 1 (DRP1). As a result, both MFN1 and DRP1 were found to be essential to oogenesis and fertility, while MFN2 deletion led to offspring with increased weight gain and glucose intolerance. Curiously, neither MFN1/MFN2 deficiency nor DRP1 overexpression enhanced mitochondrial fragmentation, indicating that mitochondrial size is strictly regulated in oocytes. Therefore, the present work seeks to discuss the role of mitochondria in supporting oogenesis as well as recent findings connecting defective mitochondrial dynamics in oocytes with infertility and transmission of metabolic disorders.
Collapse
Affiliation(s)
- Marcos Roberto Chiaratti
- Departamento de Genética e Evolução, Centro de Ciências Biológicas e da Saúde, Universidade Federal de São Carlos, São Carlos, 13565-905 Brazil
| |
Collapse
|
339
|
Priya K, Setty M, Babu UV, Pai KSR. Implications of environmental toxicants on ovarian follicles: how it can adversely affect the female fertility? ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:67925-67939. [PMID: 34628616 PMCID: PMC8718383 DOI: 10.1007/s11356-021-16489-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/07/2021] [Indexed: 05/06/2023]
Abstract
The pool of primordial follicles formed in the ovaries during early development determines the span and quality of fertility in the reproductive life of a woman. As exposure to occupational and environmental toxicants (ETs) has become inevitable, consequences on female fertility need to be established. This review focuses on the ETs, especially well-studied prototypes of the classes endocrine disrupting chemicals (EDCs), heavy metals, agrochemicals, cigarette smoke, certain chemicals used in plastic, cosmetic and sanitary product industries etc that adversely affect the female fertility. Many in vitro, in vivo and epidemiological studies have indicated that these ETs have the potential to affect folliculogenesis and cause reduced fertility in women. Here, we emphasize on four main conditions: polycystic ovary syndrome, primary ovarian insufficiency, multioocytic follicles and meiotic defects including aneuploidies which can be precipitated by ETs. These are considered main causes for reduced female fertility by directly altering the follicular recruitment, development and oocytic meiosis. Although substantial experimental evidence is drawn with respect to the detrimental effects, it is clear that establishing the role of one ET as a risk factor in a single condition is difficult as multiple conditions have common risk factors. Therefore, it is important to consider this as a matter of public and wildlife health.
Collapse
Affiliation(s)
- Keerthi Priya
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Manjunath Setty
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Uddagiri Venkanna Babu
- Phytochemistry Department, R & D Centre, The Himalaya Drug Company, Makali, Tumkur Road, Bangalore, Karnataka, 562162, India
| | - Karkala Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
340
|
Dehghan Z, Mohammadi-Yeganeh S, Sameni M, Mirmotalebisohi SA, Zali H, Salehi M. Repurposing new drug candidates and identifying crucial molecules underlying PCOS Pathogenesis Based On Bioinformatics Analysis. Daru 2021; 29:353-366. [PMID: 34480296 PMCID: PMC8416576 DOI: 10.1007/s40199-021-00413-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 08/16/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUNDS Polycystic ovary syndrome affects 7% of women of reproductive ages. Poor-quality oocytes, along with lower cleavage and implantation rates, reduce fertilization. OBJECTIVE This study aimed to determine crucial molecular mechanisms behind PCOS pathogenesis and repurpose new drug candidates interacting with them. To predict a more in-depth insight, we applied a novel bioinformatics approach to analyze interactions between the drug-related and PCOS proteins in PCOS patients. METHODS The newest proteomics data was retrieved from 16 proteomics datasets and was used to construct the PCOS PPI network using Cytoscape. The topological network analysis determined hubs and bottlenecks. The MCODE Plugin was used to identify highly connected regions, and the associations between PCOS clusters and drug-related proteins were evaluated using the Chi-squared/Fisher's exact test. The crucial PPI hub-bottlenecks and the shared molecules (between the PCOS clusters and drug-related proteins) were then investigated for their drug-protein interactions with previously US FDA-approved drugs to predict new drug candidates. RESULTS The PI3K/AKT pathway was significantly related to one PCOS subnetwork and most drugs (metformin, letrozole, pioglitazone, and spironolactone); moreover, VEGF, EGF, TGFB1, AGT, AMBP, and RBP4 were identified as the shared proteins between the PCOS subnetwork and the drugs. The shared top biochemical pathways between another PCOS subnetwork and rosiglitazone included metabolic pathways, carbon metabolism, and citrate cycle, while the shared proteins included HSPB1, HSPD1, ACO2, TALDO1, VDAC1, and MDH2. We proposed some new candidate medicines for further PCOS treatment investigations, such as copper and zinc compounds, reteplase, alteplase, gliclazide, Etc. CONCLUSION Some of the crucial molecules suggested by our model have already been experimentally reported as critical molecules in PCOS pathogenesis. Moreover, some repurposed medications have already shown beneficial effects on infertility treatment. These previous experimental reports confirm our suggestion for investigating our other repurposed drugs (in vitro and in vivo).
Collapse
Affiliation(s)
- Zeinab Dehghan
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular & Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Cellular & Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Sameni
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular & Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Mirmotalebisohi
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular & Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hakimeh Zali
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Salehi
- Cellular & Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
341
|
Ye Q, Zeng X, Cai S, Qiao S, Zeng X. Mechanisms of lipid metabolism in uterine receptivity and embryo development. Trends Endocrinol Metab 2021; 32:1015-1030. [PMID: 34625374 DOI: 10.1016/j.tem.2021.09.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/19/2021] [Accepted: 09/06/2021] [Indexed: 02/05/2023]
Abstract
Metabolic regulation plays important roles in embryo development and uterine receptivity during early pregnancy, ultimately influencing pregnancy efficiency in mammals. The important roles of lipid metabolism during early pregnancy have not been fully understood. Here, we described the regulatory roles of phospholipid, sphingolipid, and cholesterol metabolism on early embryo development, implantation, and uterine receptivity through production of cannabinoids, prostaglandins, lysophosphatidic acid, sphingosine-1-phosphate, and steroid hormones. Moreover, the impacts of lipids and fatty acids on embryo development potential and the related epigenetic modifications are also discussed. This review aims to elucidate the modulations of lipid metabolism on uterine receptivity and embryo development, contributing to novel strategies to establish dietary balanced lipids and fatty acids for reducing early embryo loss.
Collapse
Affiliation(s)
- Qianhong Ye
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, P. R. China; Beijing Key Laboratory of Biofeed Additives, Beijing 100193, P. R. China
| | - Xiangzhou Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, P. R. China; Beijing Key Laboratory of Biofeed Additives, Beijing 100193, P. R. China
| | - Shuang Cai
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, P. R. China; Beijing Key Laboratory of Biofeed Additives, Beijing 100193, P. R. China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, P. R. China; Beijing Key Laboratory of Biofeed Additives, Beijing 100193, P. R. China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, P. R. China; Beijing Key Laboratory of Biofeed Additives, Beijing 100193, P. R. China.
| |
Collapse
|
342
|
Bülow NS, Holt MD, Skouby SO, Petersen KB, Englund ALM, Pinborg A, Macklon NS. Co-treatment with letrozole during ovarian stimulation for IVF/ICSI: a systematic review and meta-analysis. Reprod Biomed Online 2021; 44:717-736. [DOI: 10.1016/j.rbmo.2021.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022]
|
343
|
Zhang L, Wang W, Liu L, Zhang Y, Zhang X. Peroxisome proliferator-activated receptor gamma coactivator 1-alpha protects a fibrotic liver from partial hepatectomy-induced advanced liver injury through regulating cell cycle arrest. Basic Clin Pharmacol Toxicol 2021; 130:254-267. [PMID: 34845850 PMCID: PMC9300180 DOI: 10.1111/bcpt.13697] [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: 08/04/2021] [Revised: 11/04/2021] [Accepted: 11/22/2021] [Indexed: 11/30/2022]
Abstract
Background A fibrotic liver may have an impaired regenerative capacity. Because liver transplantation is donor limited, understanding the regenerative ability of a fibrotic liver is important. Methods A two‐thirds partial hepatectomy (PH) was performed in C57Bl/6 mice with or without carbon tetrachloride (CCl4) treatment. Liver regeneration in the fibrotic liver after PH was assessed by the intrahepatic expression of the cell cycle regulators p53, p21, cyclin D1, c‐Fos and CDK2 using Western blot analysis. In addition, the expression of PGC‐1α and the cell proliferation‐related proteins PCNA and phosphate histone H3 was determined by Western blot and immunohistochemical staining analyses. Histone epigenetic modification of the PGC‐1α promoter was investigated through chromatin immunoprecipitation (ChIP) and reverse transcription–quantitative polymerase chain reaction (RT‐qPCR) assays. The impact of PGC‐1α on liver regeneration after PH was further evaluated in PGC‐1α‐knockout mice. Results A decreased expression of PGC‐1α and liver regeneration‐related genes in the fibrotic liver was detected after a PH. Histone acetylation at the PGC‐1α promoter led to increases in PGC‐1α expression and the survival rate in the fibrotic group after a PH. PGC‐1α‐mediated liver regeneration was further demonstrated in PGC‐1αf/falbcre+/0 mice. Conclusion Targeting PGC‐1α may represent a strategy to improve the treatment of PH in patients with liver fibrosis.
Collapse
Affiliation(s)
- Linzhong Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Lipeng Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yanghao Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiuying Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
344
|
Imprinted lncRNA Dio3os preprograms intergenerational brown fat development and obesity resistance. Nat Commun 2021; 12:6845. [PMID: 34824246 PMCID: PMC8617289 DOI: 10.1038/s41467-021-27171-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
Maternal obesity (MO) predisposes offspring to obesity and metabolic disorders but little is known about the contribution of offspring brown adipose tissue (BAT). We find that MO impairs fetal BAT development, which persistently suppresses BAT thermogenesis and primes female offspring to metabolic dysfunction. In fetal BAT, MO enhances expression of Dio3, which encodes deiodinase 3 (D3) to catabolize triiodothyronine (T3), while a maternally imprinted long noncoding RNA, Dio3 antisense RNA (Dio3os), is inhibited, leading to intracellular T3 deficiency and suppression of BAT development. Gain and loss of function shows Dio3os reduces D3 content and enhances BAT thermogenesis, rendering female offspring resistant to high fat diet-induced obesity. Attributing to Dio3os inactivation, its promoter has higher DNA methylation in obese dam oocytes which persists in fetal and adult BAT, uncovering an oocyte origin of intergenerational obesity. Overall, our data uncover key features of Dio3os activation in BAT to prevent intergenerational obesity and metabolic dysfunctions. Maternal obesity predisposes offspring to obesity and metabolic disorders through incompletely understood mechanisms. Here the authors report that Dio3os is an imprinted long-coding RNA that modulates brown adipose tissue development and obesity resistance in the offspring.
Collapse
|
345
|
Lin T, Sun L, Lee JE, Kim SY, Jin DI. DNA damage repair is suppressed in porcine aged oocytes. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2021; 63:984-997. [PMID: 34796342 PMCID: PMC8564305 DOI: 10.5187/jast.2021.e90] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022]
Abstract
This study sought to evaluate DNA damage and repair in porcine postovulatory aged
oocytes. The DNA damage response, which was assessed by H2A.X expression,
increased in porcine aged oocytes over time. However, the aged oocytes exhibited
a significant decrease in the expression of RAD51, which reflects the DNA damage
repair capacity. Further experiments suggested that the DNA repair ability was
suppressed by the downregulation of genes involved in the homologous
recombination (HR) and nonhomologous end-joining (NHEJ) pathways. The expression
levels of the cell cycle checkpoint genes, CHEK1 and
CHEK2, were upregulated in porcine aged oocytes in response
to induced DNA damage. Immunofluorescence results revealed that the expression
level of H3K79me2 was significantly lower in porcine aged oocytes than in
control oocytes. In addition, embryo quality was significantly reduced in aged
oocytes, as assessed by measuring the cell proliferation capacity. Our results
provide evidence that DNA damage is increased and the DNA repair ability is
suppressed in porcine aged oocytes. These findings increase our understanding of
the events that occur during postovulatory oocyte aging.
Collapse
Affiliation(s)
- Tao Lin
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China.,Division of Animal & Dairy Science, Chungnam National University, Daejeon 34134, Korea
| | - Ling Sun
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China.,Division of Animal & Dairy Science, Chungnam National University, Daejeon 34134, Korea
| | - Jae Eun Lee
- Division of Animal & Dairy Science, Chungnam National University, Daejeon 34134, Korea
| | - So Yeon Kim
- Division of Animal & Dairy Science, Chungnam National University, Daejeon 34134, Korea
| | - Dong Il Jin
- Division of Animal & Dairy Science, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
346
|
Orsolini MF, Meyers SA, Dini P. An Update on Semen Physiology, Technologies, and Selection Techniques for the Advancement of In Vitro Equine Embryo Production: Section II. Animals (Basel) 2021; 11:ani11113319. [PMID: 34828049 PMCID: PMC8614388 DOI: 10.3390/ani11113319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/09/2021] [Accepted: 11/18/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary In order to improve fertilization and pregnancy rates within artificial insemination or in vitro fertilization techniques in horses, producers may choose to select the best sperm within an ejaculate. In this paper, we review conventional and novel methods of sperm selection. Abstract As the use of assisted reproductive technologies (ART) and in vitro embryo production (IVP) expand in the equine industry, it has become necessary to further our understanding of available semen selection techniques. This segment of our two-section review will focus on the selection of spermatozoa based on quality and sex for equine intracytoplasmic sperm injection (ICSI), as well as current and future developments in sperm sorting technologies. Ultimately, novel methods of semen selection will be assessed based on their efficacy in other species and their relevance and future application towards ARTs in the horse.
Collapse
Affiliation(s)
- Morgan F. Orsolini
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA;
| | - Stuart A. Meyers
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA;
| | - Pouya Dini
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA;
- Correspondence:
| |
Collapse
|
347
|
Bülow NS, Skouby SO, Warzecha AK, Udengaard H, Andersen CY, Holt MD, Grøndahl ML, Nyboe Andersen A, Sopa N, Mikkelsen ALE, Pinborg A, Macklon NS. Impact of letrozole co-treatment during ovarian stimulation with gonadotrophins for IVF: a multicentre, randomized, double-blinded placebo-controlled trial. Hum Reprod 2021; 37:309-321. [PMID: 34792133 DOI: 10.1093/humrep/deab249] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Does letrozole co-treatment during ovarian stimulation with gonadotrophins for IVF reduce the proportion of women with premature progesterone levels above 1.5 ng/ml at the time of triggering final oocyte maturation? SUMMARY ANSWER The proportion of women with premature progesterone above 1.5 ng/ml was not significantly affected by letrozole co-treatment. WHAT IS KNOWN ALREADY IVF creates multiple follicles with supraphysiological levels of sex steroids interrupting the endocrine milieu and affects the window of implantation. Letrozole is an effective aromatase inhibitor, normalizing serum oestradiol, thereby ameliorating some of the detrimental effects of IVF treatment. STUDY DESIGN, SIZE, DURATION A randomized, double-blinded placebo-controlled trial investigated letrozole intervention during stimulation for IVF with FSH. The trial was conducted at four fertility clinics at University Hospitals in Denmark from August 2016 to November 2018. PARTICIPANTS/MATERIALS, SETTING, METHODS A cohort of 129 women with expected normal ovarian reserve (anti-Müllerian hormone 8-32 nmol/l) completed an IVF cycle with fresh embryo transfer and received co-treatment with either 5 mg/day letrozole (n = 67) or placebo (n = 62), along with the FSH. Progesterone, oestradiol, FSH, LH and androgens were analysed in repeated serum samples collected from the start of the stimulation to the mid-luteal phase. In addition, the effect of letrozole on reproductive outcomes, total FSH consumption and adverse events were assessed. MAIN RESULTS AND THE ROLE OF CHANCE The proportion of women with premature progesterone >1.5 ng/ml was similar (6% vs 0% (OR 0.0, 95% CI [0.0; 1.6], P = 0.12) in the letrozole versus placebo groups, respectively), whereas the proportion of women with mid-luteal progesterone >30 ng/ml was significantly increased in the letrozole group: (59% vs 31% (OR 3.3, 95% CI [1.4; 7.1], P = 0.005)). Letrozole versus placebo decreased oestradiol levels on the ovulation trigger day by 68% (95% CI [60%; 75%], P < 0.0001). Other hormonal profiles, measured as AUC, showed the following results. The increase in LH in the letrozole group versus placebo group was 38% (95% CI [21%; 58%], P < 0.0001) and 34% (95% CI [11%; 61%], P = 0.006) in the follicular and luteal phases, respectively. In the letrozole group versus placebo group, testosterone increased by 79% (95% CI [55%; 105%], P < 0.0001) and 49% (95% CI [30%; 72%], P < 0.0001) in the follicular and luteal phases, respectively. In the letrozole group versus placebo group, the increase in androstenedione was by 85% (95% CI [59%; 114%], P < 0.0001) and 69% (95% CI [48%; 94%], P < 0.0001) in the follicular and luteal phases, respectively. The ongoing pregnancy rate was similar between the letrozole and placebo groups (31% vs 39% (risk-difference of 8%, 95% CI [-25%; 11%], P = 0.55)). No serious adverse reactions were recorded in either group. The total duration of exogenous FSH stimulation was 1 day shorter in the intervention group, significantly reducing total FSH consumption (mean difference -100 IU, 95% CI [-192; -21], P = 0.03). LIMITATIONS, REASONS FOR CAUTION Late follicular progesterone samples were collected on the day before and day of ovulation triggering for patient logistic considerations, and the recently emerged knowledge about diurnal variation of progesterone was not taken into account. The study was powered to detect hormonal variations but not differences in pregnancy outcomes. WIDER IMPLICATIONS OF THE FINDINGS Although the use of letrozole has no effect on the primary outcome, the number of women with a premature increase in progesterone on the day of ovulation triggering, the increased progesterone in the mid-luteal phase due to letrozole may contribute to optimizing the luteal phase endocrinology. The effect of letrozole on increasing androgens and reducing FSH consumption may be used in poor responders. However, the effect of letrozole on implantation and ongoing pregnancy rates should be evaluated in a meta-analysis or larger randomized controlled trial (RCT). STUDY FUNDING/COMPETING INTEREST(S) Funding was received from EU Interreg for ReproUnion and Ferring Pharmaceuticals, and Roche Diagnostics contributed with assays. N.S.M. and A.P. have received grants from Ferring, Merck Serono, Anecova and Gedeon Richter, and/or personal fees from IBSA, Vivoplex, ArtPred and SPD, outside the submitted work. The remaining authors have no competing interests. TRIAL REGISTRATION NUMBERS NCT02939898 and NCT02946684. TRIAL REGISTRATION DATE 15 August 2016. DATE OF FIRST PATIENT’S ENROLMENT 22 August 2016.
Collapse
Affiliation(s)
- Nathalie Søderhamn Bülow
- Department of Obstetrics and Gynaecology, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Copenhagen, Denmark.,The Fertility Department, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sven Olaf Skouby
- Department of Obstetrics and Gynaecology, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Copenhagen, Denmark
| | - Agnieszka Katarzyna Warzecha
- Department of Obstetrics and Gynaecology, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Copenhagen, Denmark
| | - Hanne Udengaard
- Department of Obstetrics and Gynaecology, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Copenhagen, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marianne Dreyer Holt
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Zealand University Hospital, Køge, Denmark
| | - Marie Louise Grøndahl
- Department of Obstetrics and Gynaecology, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Copenhagen, Denmark
| | | | - Negjyp Sopa
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital, Hvidovre, Denmark
| | - Anne Lis Englund Mikkelsen
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Zealand University Hospital, Køge, Denmark
| | - Anja Pinborg
- The Fertility Department, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital, Hvidovre, Denmark
| | - Nicholas Stephen Macklon
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Zealand University Hospital, Køge, Denmark.,London Women's Clinic, London, UK
| |
Collapse
|
348
|
Mohamed WI, Schenk AD, Kempf G, Cavadini S, Basters A, Potenza A, Abdul Rahman W, Rabl J, Reichermeier K, Thomä NH. The CRL4 DCAF1 cullin-RING ubiquitin ligase is activated following a switch in oligomerization state. EMBO J 2021; 40:e108008. [PMID: 34595758 PMCID: PMC8591539 DOI: 10.15252/embj.2021108008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 11/09/2022] Open
Abstract
The cullin‐4‐based RING‐type (CRL4) family of E3 ubiquitin ligases functions together with dedicated substrate receptors. Out of the ˜29 CRL4 substrate receptors reported, the DDB1‐ and CUL4‐associated factor 1 (DCAF1) is essential for cellular survival and growth, and its deregulation has been implicated in tumorigenesis. We carried out biochemical and structural studies to examine the structure and mechanism of the CRL4DCAF1 ligase. In the 8.4 Å cryo‐EM map of CRL4DCAF1, four CUL4‐RBX1‐DDB1‐DCAF1 protomers are organized into two dimeric sub‐assemblies. In this arrangement, the WD40 domain of DCAF1 mediates binding with the cullin C‐terminal domain (CTD) and the RBX1 subunit of a neighboring CRL4DCAF1 protomer. This renders RBX1, the catalytic subunit of the ligase, inaccessible to the E2 ubiquitin‐conjugating enzymes. Upon CRL4DCAF1 activation by neddylation, the interaction between the cullin CTD and the neighboring DCAF1 protomer is broken, and the complex assumes an active dimeric conformation. Accordingly, a tetramerization‐deficient CRL4DCAF1 mutant has higher ubiquitin ligase activity compared to the wild‐type. This study identifies a novel mechanism by which unneddylated and substrate‐free CUL4 ligases can be maintained in an inactive state.
Collapse
Affiliation(s)
- Weaam I Mohamed
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Andreas D Schenk
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Georg Kempf
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Simone Cavadini
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Anja Basters
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Alessandro Potenza
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | | | - Julius Rabl
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Kurt Reichermeier
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.,Genentech, South San Francisco, CA, USA
| | - Nicolas H Thomä
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| |
Collapse
|
349
|
Ntostis P, Iles D, Kokkali G, Vaxevanoglou T, Kanavakis E, Pantou A, Huntriss J, Pantos K, Picton HM. The impact of maternal age on gene expression during the GV to MII transition in euploid human oocytes. Hum Reprod 2021; 37:80-92. [PMID: 34755188 PMCID: PMC8730309 DOI: 10.1093/humrep/deab226] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/18/2021] [Indexed: 12/27/2022] Open
Abstract
STUDY QUESTION Are there age-related differences in gene expression during the germinal vesicle (GV) to metaphase II (MII) stage transition in euploid human oocytes? SUMMARY ANSWER A decrease in mitochondrial-related transcripts from GV to MII oocytes was observed, with a much greater reduction in MII oocytes with advanced age. WHAT IS KNOWN ALREADY Early embryonic development is dependent on maternal transcripts accumulated and stored within the oocyte during oogenesis. Transcriptional activity of the oocyte, which dictates its ultimate developmental potential, may be influenced by age and explain the reduced competence of advanced maternal age (AMA) oocytes compared with the young maternal age (YMA). Gene expression has been studied in human and animal oocytes; however, RNA sequencing could provide further insights into the transcriptome profiling of GV and in vivo matured MII euploid oocytes of YMA and AMA patients. STUDY DESIGN, SIZE, DURATION Fifteen women treated for infertility in a single IVF unit agreed to participate in this study. Five GV and 5 MII oocytes from 6, 21-26 years old women (YMA cohort) and 5 GV and 6 MII oocytes from 6, 41-44 years old women (AMA cohort) undergoing IVF treatment were donated. The samples were collected within a time frame of 4 months. RNA was isolated and deep sequenced at the single-cell level. All donors provided either GV or MII oocytes. PARTICIPANTS/MATERIALS, SETTING, METHODS Cumulus dissection from donated oocytes was performed 38 h after hCG injection, denuded oocytes were inserted into lysis buffer supplemented with RNase inhibitor. The samples were stored at -80°C until further use. Isolated RNA from GV and MII oocytes underwent library preparation using an oligo deoxy-thymidine (dT) priming approach (SMART-Seq v4 Ultra Low Input RNA assay; Takara Bio, Japan) and Nextera XT DNA library preparation assay (Illumina, USA) followed by deep sequencing. Data processing, quality assessment and bioinformatics analysis were performed using source-software, mainly including FastQC, HISAT2, StringTie and edgeR, along with functional annotation analysis, while scploid R package was employed to determine the ploidy status. MAIN RESULTS AND THE ROLE OF CHANCE Following deep sequencing of single GV and MII oocytes in both YMA and AMA cohorts, several hundred transcripts were found to be expressed at significantly different levels. When YMA and AMA MII oocyte transcriptomes were compared, the most significant of these were related to mitochondrial structure and function, including biological processes, mitochondrial respiratory chain complex I assembly and mitochondrial translational termination (false discovery rate (FDR) 6.0E-10 to 1.2E-7). These results indicate a higher energy potential of the YMA MII cohort that is reduced with ageing. Other biological processes that were significantly higher in the YMA MII cohort included transcripts involved in the translation process (FDR 1.9E-2). Lack of these transcripts could lead to inappropriate protein synthesis prior to or upon fertilisation of the AMA MII oocytes. LARGE SCALE DATA The RNA sequencing data were deposited in the Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo), under the accession number: GSE164371. LIMITATIONS, REASONS FOR CAUTION The relatively small sample size could be a reason for caution. However, the RNA sequencing results showed homogeneous clustering with low intra-group variation and five to six biological replicates derived from at least three different women per group minimised the potential impact of the sample size. WIDER IMPLICATIONS OF THE FINDINGS Understanding the effects of ageing on the oocyte transcriptome could highlight the mechanisms involved in GV to MII transition and identify biomarkers that characterise good MII oocyte quality. This knowledge has the potential to guide IVF regimes for AMA patients. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the Medical Research Council (MRC Grant number MR/K020501/1).
Collapse
Affiliation(s)
- P Ntostis
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - D Iles
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - G Kokkali
- Genesis Athens Clinic, Reproductive Medicine Unit, Athens, Greece
| | - T Vaxevanoglou
- Genesis Athens Clinic, Reproductive Medicine Unit, Athens, Greece
| | - E Kanavakis
- Genesis Athens Clinic, Reproductive Medicine Unit, Athens, Greece
| | - A Pantou
- Genesis Athens Clinic, Reproductive Medicine Unit, Athens, Greece
| | - J Huntriss
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - K Pantos
- Genesis Athens Clinic, Reproductive Medicine Unit, Athens, Greece
| | - H M Picton
- Genesis Genoma Laboratory, Athens, Greece
| |
Collapse
|
350
|
Zhao X, Huang JX, Zhang H, Gong X, Dong J, Ren HL, Liu Z. A comparison study of superovulation strategies for C57BL/6J and B6D2F1 mice in CRISPR-Cas9 mediated genome editing. Reprod Fertil Dev 2021; 33:772-781. [PMID: 34748725 DOI: 10.1071/rd21199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 08/12/2021] [Indexed: 11/23/2022] Open
Abstract
Reproductive techniques such as superovulation and in vitro fertilisation (IVF) have been widely used in generating genetically modified animals. The current gold standard for superovulation in mice is using coherent treatments of equine chorionic gonadotropin (eCG) and human chorionic gonadotropin (hCG). An alternative method using inhibin antiserum (IAS) instead of eCG has been recently reported. Here, we evaluate different superovulation strategies in C57BL/6J and B6D2F1 mice. Firstly, we found that using 5-week-old C57BL/6J and 4-week-old B6D2F1 donors could achieve better superovulation outcomes. Then, we compared eCG-hCG, IAS-hCG and eCG-IAS-hCG with different dosages in both mouse strains. Significantly increased numbers of oocytes were obtained by using IAS-hCG and eCG-IAS-hCG methods. However, low fertilisation rates (36.3-38.8%) were observed when natural mating was applied. We then confirmed that IVF could dramatically ameliorate the fertilisation rates up to 89.1%. Finally, we performed CRISPR-Cas9 mediated genome editing targeting Scn11a and Kcnh1 loci, and successfully obtained mutant pups using eCG-hCG and IAS-hCG induced zygotes, which were fertilised by either natural mating or IVF. Our results showed that IAS is a promising superovulation reagent, and the efficiency of genome editing is unlikely to be affected by using IAS-induced zygotes.
Collapse
Affiliation(s)
- Xue Zhao
- School of Life Science and Technology, Weifang Medical University, Weifang, Shandong 261053, China
| | - Johnny X Huang
- School of Life Science and Technology, Weifang Medical University, Weifang, Shandong 261053, China; and Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Hailong Zhang
- School of Life Science and Technology, Weifang Medical University, Weifang, Shandong 261053, China
| | - Xueyang Gong
- School of Life Science and Technology, Weifang Medical University, Weifang, Shandong 261053, China
| | - Jinhua Dong
- School of Life Science and Technology, Weifang Medical University, Weifang, Shandong 261053, China
| | - Hong-Lin Ren
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Xi An Da Lu 5333, Changchun 130062, China
| | - Zengshan Liu
- School of Life Science and Technology, Weifang Medical University, Weifang, Shandong 261053, China; and Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Xi An Da Lu 5333, Changchun 130062, China
| |
Collapse
|