1
|
Karami N, Taei A, Hassani SN, Alizadeh N, Eftekhari-Yazdi P, Hassani F. The effects of insulin-transferrin-selenium (ITS) and CHIR99021 on the development of pre-implantation human arrested embryos in vitro. Sci Rep 2025; 15:5006. [PMID: 39929940 PMCID: PMC11811121 DOI: 10.1038/s41598-025-89460-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
Pre-implantation development arrest poses a significant challenge in infertility treatment cycles. This study aims to evaluate the effect of Insulin, Transferrin, Selenium (ITS), and CHIR99021 on arrested human embryos. Arrested human embryos were obtained from the Embryology Department of the Royan Institute. After determining optimal concentrations, the embryos were assigned to control, CHIR99021, and ITS groups and cultured for 48-72 h. The arrest rate significantly decreased in the ITS and CHIR99021 groups compared to the control group (P < 0.05). The developmental rate up to the pre-morula stage significantly increased in the CHIR99021 group compared to the control group (P < 0.05). Additionally, there were significant increases in the expression of SOX2 in the CHIR99021 group and CCNA2 in the ITS group compared to the control group (P < 0.05). Immunofluorescent staining confirmed the expression of NANOG protein in the experimental groups. GSK3 inhibition by CHIR99021 and the application of ITS can alleviate arrest in human embryos, promote cell cycle induction, and enable progression to the blastocyst stage. Comprehensive characterization of these blastocysts in future studies is crucial to support ITS and CHIR99021 probable application in culture systems, particularly for women of advanced maternal age and those experiencing severe male factor infertility.
Collapse
Affiliation(s)
- Narges Karami
- MSC., Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Adeleh Taei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nazanin Alizadeh
- MSC., Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, P.O. Box 16635-148, Tehran, Iran.
| | - Fatemeh Hassani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, P.O. Box 16635-148, Tehran, Iran.
| |
Collapse
|
2
|
Wang Y, Tian F, Yue S, Li J, Li A, Liu Y, Liang J, Gao Y, Xue S. miR-17-5p-Mediated RNA Activation Upregulates KPNA2 Expression and Inhibits High-Glucose-Induced Apoptosis of Sheep Granulosa Cells. Int J Mol Sci 2025; 26:943. [PMID: 39940713 PMCID: PMC11817598 DOI: 10.3390/ijms26030943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/17/2025] [Accepted: 01/19/2025] [Indexed: 02/16/2025] Open
Abstract
The glucose metabolism homeostasis in the follicular fluid microenvironment plays an important role in follicular maturation and ovulation, and excessively high or low glucose concentrations have adverse effects on the differentiation of follicular granulosa cells (GCs). However, a limited number of microRNAs (miRNA) have been reported to be involved in glucose-stimulated GCs differentiation. In this study, we characterized the miRNA expression profiles of sheep ovarian GCs cultured in high-glucose and optimal glucose concentrations and focused on a differentially expressed miRNA: miR-17-5p, which may be involved in regulating high-glucose-induced GC apoptosis by targeting KPNA2. We found that overexpression of miR-17-5p significantly promoted GCs proliferation and inhibited cell apoptosis, while downregulated the mRNA and protein expression of apoptosis-related makers (Bax, Caspase-3, Caspase-9, and Bcl-2). In contrast to the classical mechanism of miRNA silencing target gene expression, miR-17-5p overexpression significantly upregulated the expression of target gene KPNA2. A dual luciferase reporter gene assay verified the targeted binding relationship between miR-17-5p and KPNA2 promoter. Meanwhile, overexpression of KPNA2 further promoted the downregulation of apoptosis-related genes driven by miR-17-5p mimics. Knockdown of KPNA2 blocked the inhibitory effect of miR-17-5p mimics on the expression of apoptosis-related genes. Our results demonstrated that miR-17-5p activated the KPNA2 promoter region and upregulated KPNA2 expression, thereby inhibiting GCs apoptosis under high glucose.
Collapse
Affiliation(s)
- Yong Wang
- Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot 010030, China; (Y.W.); (F.T.); (J.L.); (Y.L.); (J.L.); (Y.G.)
| | - Feng Tian
- Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot 010030, China; (Y.W.); (F.T.); (J.L.); (Y.L.); (J.L.); (Y.G.)
| | - Sicong Yue
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China; (S.Y.); (A.L.)
| | - Jiuyue Li
- Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot 010030, China; (Y.W.); (F.T.); (J.L.); (Y.L.); (J.L.); (Y.G.)
| | - Ao Li
- College of Animal Science and Technology, Hebei Agricultural University, Baoding 071000, China; (S.Y.); (A.L.)
| | - Yang Liu
- Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot 010030, China; (Y.W.); (F.T.); (J.L.); (Y.L.); (J.L.); (Y.G.)
| | - Jianyong Liang
- Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot 010030, China; (Y.W.); (F.T.); (J.L.); (Y.L.); (J.L.); (Y.G.)
| | - Yuan Gao
- Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot 010030, China; (Y.W.); (F.T.); (J.L.); (Y.L.); (J.L.); (Y.G.)
| | - Shuyuan Xue
- Inner Mongolia Academy of Agriculture and Animal Husbandry Sciences, Hohhot 010030, China; (Y.W.); (F.T.); (J.L.); (Y.L.); (J.L.); (Y.G.)
| |
Collapse
|
3
|
Zhang J, Lv J, Qin J, Zhang M, He X, Ma B, Wan Y, Gao Y, Wang M, Hong Z. Unraveling the mysteries of early embryonic arrest: genetic factors and molecular mechanisms. J Assist Reprod Genet 2024; 41:3301-3316. [PMID: 39325344 PMCID: PMC11706821 DOI: 10.1007/s10815-024-03259-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024] Open
Abstract
Early embryonic arrest (EEA) is a critical impediment in assisted reproductive technology (ART), affecting 40% of infertile patients by halting the development of early embryos from the zygote to blastocyst stage, resulting in a lack of viable embryos for successful pregnancy. Despite its prevalence, the molecular mechanism underlying EEA remains elusive. This review synthesizes the latest research on the genetic and molecular factors contributing to EEA, with a focus on maternal, paternal, and embryonic factors. Maternal factors such as irregularities in follicular development and endometrial environment, along with mutations in genes like NLRP5, PADI6, KPNA7, IGF2, and TUBB8, have been implicated in EEA. Specifically, PATL2 mutations are hypothesized to disrupt the maternal-zygotic transition, impairing embryo development. Paternal contributions to EEA are linked to chromosomal variations, epigenetic modifications, and mutations in genes such as CFAP69, ACTL7A, and M1AP, which interfere with sperm development and lead to infertility. Aneuploidy may disrupt spindle assembly checkpoints and pathways including Wnt, MAPK, and Hippo signaling, thereby contributing to EEA. Additionally, key genes involved in embryonic genome activation-such as ZSCAN4, DUXB, DUXA, NANOGNB, DPPA4, GATA6, ARGFX, RBP7, and KLF5-alongside functional disruptions in epigenetic modifications, mitochondrial DNA, and small non-coding RNAs, play critical roles in the onset of EEA. This review provides a comprehensive understanding of the genetic and molecular underpinnings of EEA, offering a theoretical foundation for the diagnosis and potential therapeutic strategies aimed at improving pregnancy outcomes.
Collapse
Affiliation(s)
- Jinyi Zhang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Jing Lv
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Juling Qin
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Ming Zhang
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Xuanyi He
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Binyu Ma
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Yingjing Wan
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Ying Gao
- Center for Reproductive Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Mei Wang
- 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.
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, Hubei, P.R. China.
| | - Zhidan Hong
- 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.
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan, Hubei, P.R. China.
| |
Collapse
|
4
|
Gu R, Wu T, Fu J, Sun YJ, Sun XX. Advances in the genetic etiology of female infertility. J Assist Reprod Genet 2024; 41:3261-3286. [PMID: 39320554 PMCID: PMC11707141 DOI: 10.1007/s10815-024-03248-w] [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: 04/19/2024] [Accepted: 08/07/2024] [Indexed: 09/26/2024] Open
Abstract
Human reproduction is a complex process involving gamete maturation, fertilization, embryo cleavage and development, blastocyst formation, implantation, and live birth. If any of these processes are abnormal or arrest, reproductive failure will occur. Infertility is a state of reproductive dysfunction caused by various factors. Advances in molecular genetics, including cell and molecular genetics, and high-throughput sequencing technologies, have found that genetic factors are important causes of infertility. Genetic variants have been identified in infertile women or men and can cause gamete maturation arrest, poor quality gametes, fertilization failure, and embryonic developmental arrest during assisted reproduction technology (ART), and thus reduce the clinical success rates of ART. This article reviews clinical studies on repeated in vitro fertilization (IVF)/intracytoplasmic sperm injection (ICSI) failures caused by ovarian dysfunction, oocyte maturation defects, oocyte abnormalities, fertilization disorders, and preimplantation embryonic development arrest due to female genetic etiology, the accumulation of pathogenic genes and gene pathogenic loci, and the functional mechanism and clinical significance of pathogenic genes in gametogenesis and early embryonic development.
Collapse
Affiliation(s)
- Ruihuan Gu
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China
| | - Tianyu Wu
- Institute of Pediatrics, State Key Laboratory of Genetic Engineering, Institutes of BiomedicalSciences, Shanghai Key Laboratory of Medical Epigenetics, Children's Hospital of Fudan University, Fudan University, Shanghai, 200032, China
| | - Jing Fu
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China
| | - Yi-Juan Sun
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China.
| | - Xiao-Xi Sun
- Department of Shanghai Ji'ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, 352 Dalin Road, Shanghai, 200011, China.
| |
Collapse
|
5
|
Rother F, Abu Hweidi D, Hartmann E, Bader M. Normal male fertility in a mouse model of KPNA2 deficiency. PLoS One 2024; 19:e0304189. [PMID: 39423201 PMCID: PMC11488728 DOI: 10.1371/journal.pone.0304189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
The nuclear transport of proteins is mediated by karyopherins and has been implicated to be crucial for germ cell and embryonic development. Deletion of distinct members of the karyopherin alpha family has been shown to cause male and female infertility in mice. Using a genetrap approach, we established mice deficient for KPNA2 (KPNA2 KO) and investigated the role of this protein in male germ cell development and fertility. Breeding of male KPNA2 KO mice leads to healthy offsprings in all cases albeit the absence of KPNA2 resulted in a reduction in sperm number by 60%. Analyses of the KPNA2 expression in wild-type mice revealed a strong KPNA2 presence in meiotic germ cells of all stages while a rapid decline is found in round spermatids. The high KPNA2 expression throughout all meiotic stages of sperm development suggests a possible function of KPNA2 during this phase, hence in its absence the spermatogenesis is not completely blocked. In KPNA2 KO mice, a higher portion of sperms presented with morphological abnormalities in the head and neck region, but a severe spermiogenesis defect was not found. Thus, we conclude that the function of KPNA2 in round spermatids is dispensable, as our mice do not show any signs of infertility. Our data provide evidence that KPNA2 is not crucial for male germ cell development and fertility.
Collapse
Affiliation(s)
- Franziska Rother
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | | | - Enno Hartmann
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
6
|
Pan Z, Wang W, Wu L, Yao Z, Wang W, Chen Y, Gu H, Dong J, Mu J, Zhang Z, Fu J, Li Q, Wang L, Sun X, Kuang Y, Sang Q, Chen B. Bi-allelic missense variants in MEI4 cause preimplantation embryonic arrest and female infertility. Hum Genet 2024; 143:1049-1060. [PMID: 38252283 DOI: 10.1007/s00439-023-02633-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024]
Abstract
Preimplantation embryonic arrest is an important pathogenesis of female infertility, but little is known about the genetic factors behind this phenotype. MEI4 is an essential protein for DNA double-strand break formation during meiosis, and Mei4 knock-out female mice are viable but sterile, indicating that MEI4 plays a crucial role in reproduction. To date, MEI4 has not been found to be associated with any human reproductive diseases. Here, we identified six compound heterozygous and homozygous MEI4 variants-namely, c.293C > T, p.(Ser98Leu), c.401C > G, p.(Pro134Arg), c.391C > G, p.(Pro131Ala), c.914A > T, p.(Tyr305Phe), c.908C > G, p.(Ala303Gly), and c.899A > T, p.(Gln300Leu)-in four independent families that were responsible for female infertility mainly characterized by preimplantation embryonic arrest. In vitro, we found that these variants reduced the interaction between MEI4 and DNA. In vivo, we generated a knock-in mouse model and demonstrated that female mice were infertile and were characterized by developmental defects during oogenesis. Our findings reveal the important roles of MEI4 in human reproduction and provide a new diagnostic marker for genetic counseling of clinical infertility patients.
Collapse
Affiliation(s)
- Zhiqi Pan
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Weijie Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Ling Wu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhongyuan Yao
- The Reproductive Medical Center of Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wenjing Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Yao Chen
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Hao Gu
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Jie Dong
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Jian Mu
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Zhihua Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Jing Fu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Qiaoli Li
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University, The Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China.
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, 200032, China.
| |
Collapse
|
7
|
Li X, Zou Y, Geng B, Liu P, Cao L, Zhang Z, Hu S, Wang C, Zhao Y, Wu Q, Tan J. Transcriptome analysis reveals that defects in cell cycle regulation contribute to preimplantation embryo arrest. Genomics 2024; 116:110946. [PMID: 39326642 DOI: 10.1016/j.ygeno.2024.110946] [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: 06/18/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Patients with preimplantation embryo arrest (PREMBA) often experience assisted reproductive failure primarily due to the lack of transferable embryos, and the molecular mechanisms underlying PREMBA remain unclear. In our study, the embryos from five women with recurrent preimplantation embryo arrest and three women with tubal factor infertility were used for single-embryo transcriptome sequencing. Meanwhile, the transcriptomes of normal human preimplantation embryos obtained from GSE36552 were utilized to perform a comparative analysis with the transcriptomes of PREMBA embryos. Our results showed dysregulation of the cell cycle phase transition might be a potential pathogenic factor contributing to PREMBA. Through integrated analysis of the differentially expressed genes (DEGs) and weighted gene co-expression network analysis (WGCNA), we identified a number of hub genes using the protein-protein interaction network. The top 5 hub genes were as follows: CCNB2, BUB1B, CDC25A, CCNB3, and PLK3. The expression of hub genes was validated in PREMBA embryos and donated embryos using RT-qPCR. The knockdown of Ccnb2 in mouse zygotes led to an increase in embryo fragmentation, a rise in apoptosis, and a reduction in blastocyst formation. Furthermore, silencing the expression of CDC25A in HEK293T cells resulted in a decrease in cell proliferation and an increase in apoptosis, providing further support for our findings. Our findings could predict the development outcomes of preimplantation embryos and be used as potential therapeutic targets to prevent recurrent failures of IVF/ICSI attempts.
Collapse
Affiliation(s)
- Xin Li
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Yang Zou
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Baobao Geng
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, SuZhou, Jiangsu Province, China
| | - Peipei Liu
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Liyun Cao
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Zhiqin Zhang
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Shaofeng Hu
- JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Changhua Wang
- JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Yan Zhao
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China.
| | - Qiongfang Wu
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China.
| | - Jun Tan
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China.
| |
Collapse
|
8
|
Wang W, Sang Q, Wang L. Genetic factors of oocyte maturation arrest: an important cause for recurrent IVF/ICSI failures. J Assist Reprod Genet 2024; 41:1951-1953. [PMID: 38980564 PMCID: PMC11339007 DOI: 10.1007/s10815-024-03195-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/10/2024] Open
Abstract
Oocyte maturation arrest (OMA) is a common phenotype observed in IVF/ICSI cycles, characterized by the production of immature oocytes which lead to infertility. Previous studies have demonstrated that genetic factors play an important role in OMA, but the genetic mechanisms underlying a group of patients remain to be elucidated. In the recent issue of Journal of Assisted Reproduction and Genetics, Hu et al. and Wan et al. identified novel PATL2 or ZFP36L2 variants in OMA patients, respectively. By conducting in vitro experiments, they demonstrated the destructive effect of the variants on protein function. These findings expand the mutational spectrum of PATL2 and ZFP36L2, and provide precise reference for genetic counseling of OMA patients.
Collapse
Affiliation(s)
- Weijie Wang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Lei Wang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, and the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
9
|
Huang J, Wang C, Kuo C, Chang T, Liu Y, Hsiao T, Wang C, Yu C. Oxidative stress mediates nucleocytoplasmic shuttling of KPNA2 via AKT1-CDK1 axis-regulated S62 phosphorylation. FASEB Bioadv 2024; 6:276-288. [PMID: 39114447 PMCID: PMC11301272 DOI: 10.1096/fba.2024-00078] [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: 05/21/2024] [Revised: 06/25/2024] [Accepted: 06/28/2024] [Indexed: 08/10/2024] Open
Abstract
Karyopherin α 2 (KPNA2, importin α1), a transport factor shuttling between the nuclear and cytoplasmic compartments, is involved in the nuclear import of proteins and participates in cellular processes such as cell cycle regulation, apoptosis, and transcriptional regulation. However, it is still unclear which signaling regulates the nucleocytoplasmic distribution of KPNA2 in response to cellular stress. In this study, we report that oxidative stress increases nuclear retention of KPNA2 through alpha serine/threonine-protein kinase (AKT1)-mediated reduction of serine 62 (S62) phosphorylation. We first found that AKT1 activation was required for H2O2-induced nuclear accumulation of KPNA2. Immunoprecipitation and quantitative proteomic analysis revealed that the phosphorylation of KPNA2 at S62 was decreased under H2O2-induced oxidative stress. We showed that cyclin-dependent kinase 1 (CDK1), a kinase responsible for KPNA2 S62 phosphorylation, contributes to the localization of KPNA2 in the cytoplasm. AKT1 knockdown increased KPNA2 S62 phosphorylation and inhibited CDK1 activation. Furthermore, H2O2-induced AKT1 activation promoted nuclear KPNA2 interaction with nucleophosmin 1 (NPM1), resulting in attenuation of NPM1-mediated cyclin D1 gene transcription. Thus, we infer that the AKT1-CDK1 axis regulates the nucleocytoplasmic shuttling and function of KPNA2 through spatiotemporal regulation of KPNA2 S62 phosphorylation under oxidative stress conditions.
Collapse
Affiliation(s)
- Jie‐Xin Huang
- Graduate Institute of Biomedical Sciences, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Chun‐I Wang
- Department of Biochemistry, School of MedicineChina Medical UniversityTaichungTaiwan
| | - Chia‐Yu Kuo
- Department of Cell and Molecular Biology, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Ting‐Wei Chang
- Institute of Molecular Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Yu‐Chin Liu
- Department of Cell and Molecular Biology, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Ting‐Feng Hsiao
- Graduate Institute of Biomedical Sciences, College of MedicineChang Gung UniversityTaoyuanTaiwan
- Molecular Medicine Research CenterChang Gung UniversityTaoyuanTaiwan
| | - Chih‐Liang Wang
- School of Medicine, College of MedicineChang Gung UniversityTaoyuanTaiwan
- Department of Thoracic MedicineChang Gung Memorial HospitalTaoyuanTaiwan
| | - Chia‐Jung Yu
- Graduate Institute of Biomedical Sciences, College of MedicineChang Gung UniversityTaoyuanTaiwan
- Department of Cell and Molecular Biology, College of MedicineChang Gung UniversityTaoyuanTaiwan
- Molecular Medicine Research CenterChang Gung UniversityTaoyuanTaiwan
- Department of Thoracic MedicineChang Gung Memorial HospitalTaoyuanTaiwan
| |
Collapse
|
10
|
Hu HY, Zhang GH, Deng WF, Wei TY, Feng ZK, Li CX, Li SJ, Liu JE, Tian YP. Novel PATL2 variants cause female infertility with oocyte maturation defect. J Assist Reprod Genet 2024; 41:1965-1976. [PMID: 38954294 PMCID: PMC11339215 DOI: 10.1007/s10815-024-03150-5] [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: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 07/04/2024] Open
Abstract
PURPOSE Oocyte maturation defect (OOMD) is a rare cause of in vitro fertilization failure characterized by the production of immature oocytes. Compound heterozygous or homozygous PATL2 mutations have been associated with oocyte arrest at the germinal vesicle (GV), metaphase I (MI), and metaphase II (MII) stages, as well as morphological changes. METHODS In this study, we recruited three OOMD cases and conducted a comprehensive multiplatform laboratory investigation. RESULTS Whole exome sequence (WES) revealed four diagnostic variants in PATL2, nonsense mutation c.709C > T (p.R237*) and frameshift mutation c.1486_1487delinsT (p.A496Sfs*4) were novel mutations that have not been reported previously. Furthermore, the pathogenicity of these variants was predicted using in silico analysis, which indicated detrimental effects. Molecular dynamic analysis suggested that the A496S variant disrupted the hydrophobic segment, leading to structural changes that affected the overall protein folding and stability. Additionally, biochemical and molecular experiments were conducted on cells transfected with wild-type (WT) or mutant PATL2 (p.R237* and p.A496Sfs*4) plasmid vectors. CONCLUSIONS The results demonstrated that PATL2A496Sfs*4 and PATL2R237* had impacts on protein size and expression level. Interestingly, expression levels of specific genes involved in oocyte maturation and early embryonic development were found to be simultaneously deregulated. The findings in our study expand the variation spectrum of the PATL2 gene, provide solid evidence for counseling on future pregnancies in affected families, strongly support the application of in the diagnosis of OOMD, and contribute to the understanding of PATL2 function.
Collapse
Affiliation(s)
- Hua-Ying Hu
- Birth Defects Prevention and Control Technology Research Center, Medical Innovation Research Division of Chinese, PLA General Hospital, Beijing, 100853, China
| | - Ge-Han Zhang
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese, PLA General Hospital, Beijing, China
| | - Wei-Fen Deng
- Shenzhen Hengsheng Hospital, Shenzhen, Guangdong, China
| | - Tian-Ying Wei
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, 100191, China
| | - Zhan-Ke Feng
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, 100191, China
| | - Cun-Xi Li
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, 100191, China
| | - Song Jun Li
- The Reproduction Medical Center, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518001, Guangdong, China.
| | - Jia-En Liu
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, 100191, China.
| | - Ya-Ping Tian
- Birth Defects Prevention and Control Technology Research Center, Medical Innovation Research Division of Chinese, PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
11
|
Rother F, Depping R, Popova E, Huegel S, Heiler A, Hartmann E, Bader M. Karyopherin α2 is a maternal effect gene required for early embryonic development and female fertility in mice. FASEB J 2024; 38:e23623. [PMID: 38656660 DOI: 10.1096/fj.202301572rr] [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: 08/03/2023] [Revised: 02/26/2024] [Accepted: 04/08/2024] [Indexed: 04/26/2024]
Abstract
The nuclear transport of proteins plays an important role in mediating the transition from egg to embryo and distinct karyopherins have been implicated in this process. Here, we studied the impact of KPNA2 deficiency on preimplantation embryo development in mice. Loss of KPNA2 results in complete arrest at the 2cell stage and embryos exhibit the inability to activate their embryonic genome as well as a severely disturbed nuclear translocation of Nucleoplasmin 2. Our findings define KPNA2 as a new maternal effect gene.
Collapse
Affiliation(s)
- Franziska Rother
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | | | - Elena Popova
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Stefanie Huegel
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Ariane Heiler
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Enno Hartmann
- Institute for Biology, University of Lübeck, Lübeck, Germany
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
12
|
Veraguas-Dávila D, Zapata-Rojas C, Aguilera C, Saéz-Ruiz D, Saravia F, Castro FO, Rodriguez-Alvarez L. Proteomic Analysis of Domestic Cat Blastocysts and Their Secretome Produced in an In Vitro Culture System without the Presence of the Zona Pellucida. Int J Mol Sci 2024; 25:4343. [PMID: 38673927 PMCID: PMC11050229 DOI: 10.3390/ijms25084343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Domestic cat blastocysts cultured without the zona pellucida exhibit reduced implantation capacity. However, the protein expression profile has not been evaluated in these embryos. The objective of this study was to evaluate the protein expression profile of domestic cat blastocysts cultured without the zona pellucida. Two experimental groups were generated: (1) domestic cat embryos generated by IVF and cultured in vitro (zona intact, (ZI)) and (2) domestic cat embryos cultured in vitro without the zona pellucida (zona-free (ZF group)). The cleavage, morula, and blastocyst rates were estimated at days 2, 5 and 7, respectively. Day 7 blastocysts and their culture media were subjected to liquid chromatography-tandem mass spectrometry (LC-MS/MS). The UniProt Felis catus database was used to identify the standard proteome. No significant differences were found in the cleavage, morula, or blastocyst rates between the ZI and ZF groups (p > 0.05). Proteomic analysis revealed 22 upregulated and 20 downregulated proteins in the ZF blastocysts. Furthermore, 14 proteins involved in embryo development and implantation were present exclusively in the culture medium of the ZI blastocysts. In conclusion, embryo culture without the zona pellucida did not affect in vitro development, but altered the protein expression profile and release of domestic cat blastocysts.
Collapse
Affiliation(s)
- Daniel Veraguas-Dávila
- Escuela de Medicina Veterinaria, Departamento de Ciencias Agrarias, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Km 6 Los Niches, Curicó 3340000, Chile
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Av. Vicente Méndez 595, Chillan 3780000, Chile; (C.Z.-R.); (D.S.-R.); (F.S.); (F.O.C.); (L.R.-A.)
| | - Camila Zapata-Rojas
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Av. Vicente Méndez 595, Chillan 3780000, Chile; (C.Z.-R.); (D.S.-R.); (F.S.); (F.O.C.); (L.R.-A.)
| | - Constanza Aguilera
- School of Veterinary Medicine, Faculty of Natural Sciences, San Sebastián University, Concepción 4081339, Chile;
| | - Darling Saéz-Ruiz
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Av. Vicente Méndez 595, Chillan 3780000, Chile; (C.Z.-R.); (D.S.-R.); (F.S.); (F.O.C.); (L.R.-A.)
| | - Fernando Saravia
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Av. Vicente Méndez 595, Chillan 3780000, Chile; (C.Z.-R.); (D.S.-R.); (F.S.); (F.O.C.); (L.R.-A.)
| | - Fidel Ovidio Castro
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Av. Vicente Méndez 595, Chillan 3780000, Chile; (C.Z.-R.); (D.S.-R.); (F.S.); (F.O.C.); (L.R.-A.)
| | - Lleretny Rodriguez-Alvarez
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepción, Av. Vicente Méndez 595, Chillan 3780000, Chile; (C.Z.-R.); (D.S.-R.); (F.S.); (F.O.C.); (L.R.-A.)
| |
Collapse
|
13
|
Zhang J, Su T, Fan Y, Cheng C, Xu L, LiTian. Spotlight on iron overload and ferroptosis: Research progress in female infertility. Life Sci 2024; 340:122370. [PMID: 38141854 DOI: 10.1016/j.lfs.2023.122370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Iron is an essential trace element for organisms. However, iron overload, which is common in haematological disorders (e.g. haemochromatosis, myelodysplastic syndromes, aplastic anaemia, and thalassaemia, blood transfusion-dependent or not), can promote reactive oxygen species generation and induce ferroptosis, a novel form of programmed cell death characterised by excess iron and lipid peroxidation, thus causing cell and tissue damage. Infertility is a global health concern. Recent evidence has indicated the emerging role of iron overload and ferroptosis in female infertility by inducing hypogonadism, causing ovary dysfunction, impairing preimplantation embryos, attenuating endometrial receptivity, and crosstalk between subfertility-related disorders, such as polycystic ovary syndrome and endometriosis. In addition, gut microbiota and their metabolites are involved in iron metabolism, ferroptosis, and female infertility. In this review, we systematically elaborate on the current research progress in female infertility with a novel focus on iron overload and ferroptosis and summarise promising therapies targeting iron overload and ferroptosis to recover fertility in women. In summary, our study provides new insights into female infertility and offers literature references for the clinical management of female infertility associated with iron overload and ferroptosis, which may be beneficial for females with haematopoietic disorders suffering from both iron overload and infertility.
Collapse
Affiliation(s)
- Jinghua Zhang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Tiantian Su
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Yuan Fan
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Cheng Cheng
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China
| | - Lanping Xu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People's Hospital & Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - LiTian
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China; Reproductive Medical Center, Peking University People's Hospital, Beijing 100044, China.
| |
Collapse
|
14
|
Umphred-Wilson K, Adoro S. Hypomorphic human SEL1L and HRD1 variants uncouple multilayered ER-associated degradation machinery. J Clin Invest 2024; 134:e175448. [PMID: 38226624 PMCID: PMC10786685 DOI: 10.1172/jci175448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024] Open
Abstract
The suppressor of lin-12-like-HMG-CoA reductase degradation 1 (SEL1L-HRD1) complex of the endoplasmic reticulum-associated degradation (ERAD) machinery is a key cellular proteostasis pathway. Although previous studies have shown ERAD as promoting the development and maintenance of many cell types in mice, its importance to human physiology remained undetermined. In two articles in this issue of the JCI, Qi and colleagues describe four biallelic hypomorphic SEL1L and HRD1 variants that were associated with neurodevelopment disorders, locomotor dysfunction, impaired immunity, and premature death in patients. These pathogenic SEL1L-HRD1 variants shine a light on the critical importance of ERAD in humans and pave the way for future studies dissecting ERAD mechanisms in specific cell types.
Collapse
|
15
|
Weis D, Lin LL, Wang HH, Li ZJ, Kusikova K, Ciznar P, Wolf HM, Leiss-Piller A, Wang Z, Wei X, Weis S, Skalicka K, Hrckova G, Danisovic L, Soltysova A, Yang TT, Feichtinger RG, Mayr JA, Qi L. Biallelic Cys141Tyr variant of SEL1L is associated with neurodevelopmental disorders, agammaglobulinemia, and premature death. J Clin Invest 2024; 134:e170882. [PMID: 37943617 PMCID: PMC10786703 DOI: 10.1172/jci170882] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023] Open
Abstract
Suppressor of lin-12-like-HMG-CoA reductase degradation 1 (SEL1L-HRD1) ER-associated degradation (ERAD) plays a critical role in many physiological processes in mice, including immunity, water homeostasis, and energy metabolism; however, its relevance and importance in humans remain unclear, as no disease variant has been identified. Here, we report a biallelic SEL1L variant (p. Cys141Tyr) in 5 patients from a consanguineous Slovakian family. These patients presented with not only ERAD-associated neurodevelopmental disorders with onset in infancy (ENDI) syndromes, but infantile-onset agammaglobulinemia with no mature B cells, resulting in frequent infections and early death. This variant disrupted the formation of a disulfide bond in the luminal fibronectin II domain of SEL1L, largely abolishing the function of the SEL1L-HRD1 ERAD complex in part via proteasomal-mediated self destruction by HRD1. This study reports a disease entity termed ENDI-agammaglobulinemia (ENDI-A) syndrome and establishes an inverse correlation between SEL1L-HRD1 ERAD functionality and disease severity in humans.
Collapse
Affiliation(s)
- Denisa Weis
- Department of Medical Genetics, Kepler University Hospital, School of Medicine, Johannes Kepler University, Linz, Austria
- Department of Pediatrics, Faculty of Medicine, Comenius University Bratislava and National Institute of Children’s Diseases, Bratislava, Slovakia
| | - Liangguang L. Lin
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular & Integrative Physiology and
| | - Huilun H. Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular & Integrative Physiology and
| | - Zexin Jason Li
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Katarina Kusikova
- Department of Pediatric Neurology, Faculty of Medicine, Comenius University Bratislava and National Institute of Children’s Diseases, Bratislava, Slovakia
| | - Peter Ciznar
- Department of Pediatrics, Faculty of Medicine, Comenius University Bratislava and National Institute of Children’s Diseases, Bratislava, Slovakia
| | - Hermann M. Wolf
- Immunology Outpatient Clinic, Vienna, Austria
- Sigmund Freud Private University–Medical School, Vienna, Austria
| | | | - Zhihong Wang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular & Integrative Physiology and
| | - Xiaoqiong Wei
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular & Integrative Physiology and
| | - Serge Weis
- Division of Neuropathology, Neuromed Campus, Department of Pathology and Molecular Pathology, Kepler University Hospital, Johannes Kepler University, Linz, Austria
| | - Katarina Skalicka
- Department of Pediatrics, Faculty of Medicine, Comenius University Bratislava and National Institute of Children’s Diseases, Bratislava, Slovakia
| | - Gabriela Hrckova
- Department of Pediatrics, Faculty of Medicine, Comenius University Bratislava and National Institute of Children’s Diseases, Bratislava, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, and
| | - Andrea Soltysova
- Faculty of Natural Sciences, Department of Molecular Biology, Comenius University, Bratislava, Slovakia
- Institute for Clinical and Translational Research, Biomedical Research Centre, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - René Günther Feichtinger
- University Children’s Hospital, Salzburger Landeskliniken Universitätsklinikum (SALK) and Paracelsus Medical University (PMU), Salzburg, Austria
| | - Johannes A. Mayr
- University Children’s Hospital, Salzburger Landeskliniken Universitätsklinikum (SALK) and Paracelsus Medical University (PMU), Salzburg, Austria
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular & Integrative Physiology and
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
16
|
Wei Y, Wang J, Qu R, Zhang W, Tan Y, Sha Y, Li L, Yin T. Genetic mechanisms of fertilization failure and early embryonic arrest: a comprehensive review. Hum Reprod Update 2024; 30:48-80. [PMID: 37758324 DOI: 10.1093/humupd/dmad026] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/07/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND Infertility and pregnancy loss are longstanding problems. Successful fertilization and high-quality embryos are prerequisites for an ongoing pregnancy. Studies have proven that every stage in the human reproductive process is regulated by multiple genes and any problem, at any step, may lead to fertilization failure (FF) or early embryonic arrest (EEA). Doctors can diagnose the pathogenic factors involved in FF and EEA by using genetic methods. With the progress in the development of new genetic technologies, such as single-cell RNA analysis and whole-exome sequencing, a new approach has opened up for us to directly study human germ cells and reproductive development. These findings will help us to identify the unique mechanism(s) that leads to FF and EEA in order to find potential treatments. OBJECTIVE AND RATIONALE The goal of this review is to compile current genetic knowledge related to FF and EEA, clarifying the mechanisms involved and providing clues for clinical diagnosis and treatment. SEARCH METHODS PubMed was used to search for relevant research articles and reviews, primarily focusing on English-language publications from January 1978 to June 2023. The search terms included fertilization failure, early embryonic arrest, genetic, epigenetic, whole-exome sequencing, DNA methylation, chromosome, non-coding RNA, and other related keywords. Additional studies were identified by searching reference lists. This review primarily focuses on research conducted in humans. However, it also incorporates relevant data from animal models when applicable. The results were presented descriptively, and individual study quality was not assessed. OUTCOMES A total of 233 relevant articles were included in the final review, from 3925 records identified initially. The review provides an overview of genetic factors and mechanisms involved in the human reproductive process. The genetic mutations and other genetic mechanisms of FF and EEA were systematically reviewed, for example, globozoospermia, oocyte activation failure, maternal effect gene mutations, zygotic genome activation abnormalities, chromosome abnormalities, and epigenetic abnormalities. Additionally, the review summarizes progress in treatments for different gene defects, offering new insights for clinical diagnosis and treatment. WIDER IMPLICATIONS The information provided in this review will facilitate the development of more accurate molecular screening tools for diagnosing infertility using genetic markers and networks in human reproductive development. The findings will also help guide clinical practice by identifying appropriate interventions based on specific gene mutations. For example, when an individual has obvious gene mutations related to FF, ICSI is recommended instead of IVF. However, in the case of genetic defects such as phospholipase C zeta1 (PLCZ1), actin-like7A (ACTL7A), actin-like 9 (ACTL9), and IQ motif-containing N (IQCN), ICSI may also fail to fertilize. We can consider artificial oocyte activation technology with ICSI to improve fertilization rate and reduce monetary and time costs. In the future, fertility is expected to be improved or restored by interfering with or supplementing the relevant genes.
Collapse
Affiliation(s)
- Yiqiu Wei
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingxuan Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Qu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiqian Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiling Tan
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanwei Sha
- Department of Andrology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Vogel OA, Forwood JK, Leung DW, Amarasinghe GK, Basler CF. Viral Targeting of Importin Alpha-Mediated Nuclear Import to Block Innate Immunity. Cells 2023; 13:71. [PMID: 38201275 PMCID: PMC10778312 DOI: 10.3390/cells13010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Cellular nucleocytoplasmic trafficking is mediated by the importin family of nuclear transport proteins. The well-characterized importin alpha (IMPA) and importin beta (IMPB) nuclear import pathway plays a crucial role in the innate immune response to viral infection by mediating the nuclear import of transcription factors such as IRF3, NFκB, and STAT1. The nuclear transport of these transcription factors ultimately leads to the upregulation of a wide range of antiviral genes, including IFN and IFN-stimulated genes (ISGs). To replicate efficiently in cells, viruses have developed mechanisms to block these signaling pathways. One strategy to evade host innate immune responses involves blocking the nuclear import of host antiviral transcription factors. By binding IMPA proteins, these viral proteins prevent the nuclear transport of key transcription factors and suppress the induction of antiviral gene expression. In this review, we describe examples of proteins encoded by viruses from several different families that utilize such a competitive inhibition strategy to suppress the induction of antiviral gene expression.
Collapse
Affiliation(s)
- Olivia A. Vogel
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Jade K. Forwood
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW 2678, Australia;
| | - Daisy W. Leung
- Department of Internal Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA;
| | - Gaya K. Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA;
| | - Christopher F. Basler
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| |
Collapse
|
18
|
Fan H, Zhou Z, Zheng W, Guan Y, Meng Q, Wang W, Dong J, Wan L, Zhu J, Zeng Y, Liu R, Gu H, Lin G, Chen B, Sang Q, Wang L. Homozygous variants in CDC23 cause female infertility characterized by oocyte maturation defects. Hum Genet 2023; 142:1621-1631. [PMID: 37768355 DOI: 10.1007/s00439-023-02606-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
Oocyte maturation defects are major phenotypes resulting in female infertility. Although many genetic factors have been found to be responsible for these phenotypes, the underlying pathogenic genes and variants remain to be identified. The anaphase promoting complex or cyclosome (APC/C) is known to be essential in the metaphase-to-anaphase transition. In this study, we identified two homozygous missense variants (c.986A > G, p.Y329C and c.988C > T, p.R330C) in CDC23 that are responsible for female infertility characterized by oocyte maturation defects in three infertile individuals. CDC23 (cell division cycle 23) is one of the core subunits of the APC/C. In vitro experiments showed that the variant c.986A > G (p.Y329C) led to a decrease in CDC23 protein level and the variant c.988C > T (p.R330C) changed the localization of CDC23 in HeLa cells and mouse oocytes. In vivo studies showed that Cdc23Y329C/Y329C mice successfully mimicked the patients' phenotype by causing low expression of CDC23 and APC4 and the accumulation of securin and cyclin B1 in oocytes. AZ3146 treatment was able to rescue the phenotype. Taken together, our findings reveal the important roles of CDC23 in human oocyte maturation and provide a new genetic marker for female infertility.
Collapse
Affiliation(s)
- Huizhen Fan
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Zhou Zhou
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, 200237, China
| | - Wei Zheng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Yichun Guan
- Department of Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qingxia Meng
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215000, China
| | - Wenjing Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Jie Dong
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Liuxia Wan
- Department of Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jiawei Zhu
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215000, China
| | - Yang Zeng
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Ruyi Liu
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Hao Gu
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Ge Lin
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, 410078, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, 200237, China.
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China.
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
19
|
Abstract
Reproduction involves a wide range of biological processes, including organ formation and development, neuroendocrine regulation, hormone production, and meiosis and mitosis. Infertility, the failure of reproduction, has become a major issue for human reproductive health and affects up to one in seven couples worldwide. Here, we review various aspects of human infertility, including etiology, mechanisms, and treatments, with a particular emphasis on genetics. We focus on gamete production and gamete quality, which is the core of successful reproduction. We also discuss future research opportunities and challenges to further expand our understanding of human infertility and improve patient care by providing precision diagnosis and personalized treatments.
Collapse
Affiliation(s)
- Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Pierre F Ray
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Team Genetics Epigenetics and Therapies of Infertility, Institute for Advanced Biosciences, 380000 Grenoble, France
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| |
Collapse
|
20
|
Wang W, Guo J, Shi J, Li Q, Chen B, Pan Z, Qu R, Fu J, Shi R, Xue X, Mu J, Zhang Z, Wu T, Wang W, Zhao L, Li Q, He L, Sun X, Sang Q, Lin G, Wang L. Bi-allelic pathogenic variants in PABPC1L cause oocyte maturation arrest and female infertility. EMBO Mol Med 2023:e17177. [PMID: 37052235 DOI: 10.15252/emmm.202217177] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Oocyte maturation arrest is one of the important causes of female infertility, but the genetic factors remain largely unknown. PABPC1L, a predominant poly(A)-binding protein in Xenopus, mouse, and human oocytes and early embryos prior to zygotic genome activation, plays a key role in translational activation of maternal mRNAs. Here, we identified compound heterozygous and homozygous variants in PABPC1L that are responsible for female infertility mainly characterized by oocyte maturation arrest in five individuals. In vitro studies demonstrated that these variants resulted in truncated proteins, reduced protein abundance, altered cytoplasmic localization, and reduced mRNA translational activation by affecting the binding of PABPC1L to mRNA. In vivo, three strains of Pabpc1l knock-in (KI) female mice were infertile. RNA-sequencing analysis showed abnormal activation of the Mos-MAPK pathway in the zygotes of KI mice. Finally, we activated this pathway in mouse zygotes by injecting human MOS mRNA, and this mimicked the phenotype of KI mice. Our findings reveal the important roles of PABPC1L in human oocyte maturation and add a genetic potential candidate gene to be screened for causes of infertility.
Collapse
Affiliation(s)
- Weijie Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Jing Guo
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Juanzi Shi
- Reproductive Medicine Center, Shaanxi Maternal and Child Care Service Center, Xi'an, China
| | - Qun Li
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Zhiqi Pan
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Ronggui Qu
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Jing Fu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Rong Shi
- Reproductive Medicine Center, Shaanxi Maternal and Child Care Service Center, Xi'an, China
| | - Xia Xue
- Reproductive Medicine Center, Shaanxi Maternal and Child Care Service Center, Xi'an, China
| | - Jian Mu
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Zhihua Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Tianyu Wu
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Wenjing Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Lin Zhao
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Qiaoli Li
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Lin He
- Bio-X Center, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Ge Lin
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, the Shanghai Key Laboratory of Medical Epigenetics, the Institutes of Biomedical Sciences, the State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Sharif M, Detti L, Van den Veyver IB. Take your mother's ferry: preimplantation embryo development requires maternal karyopherins for nuclear transport. J Clin Invest 2023; 133:e166279. [PMID: 36647833 PMCID: PMC9843045 DOI: 10.1172/jci166279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The genetic basis of preimplantation embryo arrest is slowly being unraveled. Recent discoveries point to maternally expressed proteins required for cellular functions before the embryonic genome is activated. In this issue of the JCI, Wang, Miyamoto, et al. suggest a critical role for karyopherin-mediated protein cargo transport between oocyte cytoplasm and nucleus. Defective maternal oocyte-expressed human karyopherin subunit α7 (KPNA7) and mouse KPNA2 fail to bind a critical substrate, ribosomal L1 domain-containing protein 1 (RSL1D1), affecting its transport to the nucleus. As shown in embryos of Kpna2-null females, the consequences are disrupted zygotic genome activation and arrest of development. These findings have important implications for diagnosis and treatment of female infertility.
Collapse
Affiliation(s)
| | - Laura Detti
- Department of Obstetrics and Gynecology
- Division of Reproductive Endocrinology and Infertility
| | - Ignatia B. Van den Veyver
- Department of Obstetrics and Gynecology
- Divisions of Maternal Fetal Medicine and Prenatal and Reproductive Genetics, and
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|