1
|
Kanatsu-Shinohara M, Morimoto H, Liu T, Tamura M, Shinohara T. Sendai virus-mediated RNA delivery restores fertility to congenital and chemotherapy-induced infertile female mice. PNAS NEXUS 2024; 3:pgae375. [PMID: 39262851 PMCID: PMC11388103 DOI: 10.1093/pnasnexus/pgae375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/13/2024] [Indexed: 09/13/2024]
Abstract
Current infertility treatment strategies focus on mature gametes, leaving a significant proportion of cases with gamete progenitors that stopped complete differentiation. On the other hand, recent advancements in next-generation sequencing have identified many candidate genes that may promote maturation of germ cells. Although gene therapy has shown success in mice, concerns about the integration of DNA vectors into oocytes hinder clinical applications. Here, we present the restoration of fertility in female mice through Sendai virus (SeV)-mediated RNA delivery. Ovaries lacking Kitl expression exhibit only primordial follicles due to impaired signaling to oocytes expressing the KIT tyrosine kinase. Despite SeVs being immunogenic and larger than the blood-follicle barrier, the administration of Kitl-expressing SeVs reinitiated oogenesis in genetically infertile mice that have only primordial follicles, resulting in the birth of normal offspring through natural mating. This virus also effectively addressed iatrogenic infertility induced by busulfan, a widely used cancer chemotherapy agent. Offspring born through SeV administration and natural mating displayed normal genomic imprinting patterns and fertility. Since SeVs pose no genotoxicity risk, the successful restoration of fertility by SeVs represents a promising approach for treating congenital infertility with somatic cell defects and protecting fertility of cancer patients who may become infertile due to loss of oocytes during cancer therapy.
Collapse
Affiliation(s)
- Mito Kanatsu-Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Yoshida Konoe, Sakyo-ku, Kyoto 606-8501, Japan
- AMED-CREST, AMED, 1-7-1 Otemachi, Chiyodaku, Tokyo 100-0004, Japan
| | - Hiroko Morimoto
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Yoshida Konoe, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tianjiao Liu
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Yoshida Konoe, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masaru Tamura
- Technology and Development Team for Mouse Phenotype Analysis Division, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Takashi Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Yoshida Konoe, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
2
|
Dai F, Wang R, Deng Z, Yang D, Wang L, Wu M, Hu W, Cheng Y. Comparison of the different animal modeling and therapy methods of premature ovarian failure in animal model. Stem Cell Res Ther 2023; 14:135. [PMID: 37202808 DOI: 10.1186/s13287-023-03333-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/06/2023] [Indexed: 05/20/2023] Open
Abstract
Incidence of premature ovarian failure (POF) is higher with the increase of the pace of life. The etiology of POF is very complex, which is closely related to genes, immune diseases, drugs, surgery, and psychological factors. Ideal animal models and evaluation indexes are essential for drug development and mechanism research. In our review, we firstly summarize the modeling methods of different POF animal models and compare their advantages and disadvantages. Recently, stem cells are widely studied for tumor treatment and tissue repair with low immunogenicity, high homing ability, high ability to divide and self-renew. Hence, we secondly reviewed recently published data on transplantation of stem cells in the POF animal model and analyzed the possible mechanism of their function. With the further insights of immunological and gene therapy, the combination of stem cells with other therapies should be actively explored to promote the treatment of POF in the future. Our article may provide guidance and insight for POF animal model selection and new drug development.
Collapse
Affiliation(s)
- Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Ruiqi Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhimin Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Linlin Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Mali Wu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wei Hu
- Department of Obstetrics and Gynecology Ultrasound, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
3
|
Du R, Cheng X, Ji J, Lu Y, Xie Y, Wang W, Xu Y, Zhang Y. Mechanism of ferroptosis in a rat model of premature ovarian insufficiency induced by cisplatin. Sci Rep 2023; 13:4463. [PMID: 36932163 PMCID: PMC10023701 DOI: 10.1038/s41598-023-31712-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023] Open
Abstract
Ferroptosis is widely present in fibrosis-related diseases. The basic pathology of premature ovarian insufficiency (POI) involves ovarian tissue fibrosis, and there are currently fewer relevant studies addressing the association between ferroptosis and POI. This study aimed to demonstrate that ferroptosis induced by cisplatin (CDDP) caused ovarian tissue fibrosis, leading to POI. Vitamin E (VE), a ferroptosis inhibitor, could repair damaged ovarian function. CDDP was used to establish a rat model of POI, and VE was administered to reverse the reproductive toxicity of CDDP. Ovarian function was assessed by histological section staining, follicle counts, sex hormone levels, as well as fertility assays. The extent of ferroptosis was assessed by transmission electron microscopy (TEM), malondialdehyde (MDA), Perls staining. CCK-8, Ethynyl-2-Deoxyuridine (EdU), and scratch assays were used to determine the effect of CDDP and VE on ovarian granulosa cell (GC) viability. Western blot, quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and immunohistochemistry were performed to evaluate ferroptosis-related molecular changes. Our results showed that CDDP caused follicle development disorders and ovarian tissue fibrosis, the levels of sex hormones suggested impaired ovarian function, and VE could reverse the reproductive toxicity of CDDP. The results of TEM, MDA and Perls staining suggested that the typical mitochondrial signature of ferroptosis was altered in ovarian GCs from the CDDP group, with significantly higher levels of lipid peroxidation and significant iron deposition in ovarian tissue, whereas VE mitigated the extent of ferroptosis. Molecular experiments then confirmed that the ferroptosis-related molecules acetyl CoA synthetase long chain family member 4 (ACSl4), 15-lipoxygenase-1 (ALOX15), solute carrier family 7 member 11 (SLC7A11), and glutathione peroxidase 4 (GPX4) were differentially expressed in each group. In summary, our study preliminarily demonstrated that CDDP may promote GCs to undergo ferroptosis, cause follicle development disorders, ovarian tissue fibrosis, and induce POI by regulating the expression of ACSl4, ALOX15, SLC7A11, and GPX4, while VE improved impaired ovarian function.
Collapse
Grants
- MS12021003, KYCX20_2799, KYCX21_3118 XiCheng
- MS12021003, KYCX20_2799, KYCX21_3118 XiCheng
- MS12021003, KYCX20_2799, KYCX21_3118 XiCheng
- MS12021003, KYCX20_2799, KYCX21_3118 XiCheng
- MS12021003, KYCX20_2799, KYCX21_3118 XiCheng
- MS12021003, KYCX20_2799, KYCX21_3118 XiCheng
- MS12021003, KYCX20_2799, KYCX21_3118 XiCheng
- BE2018672 Yuquan Zhang
- BE2018672 Yuquan Zhang
- BE2018672 Yuquan Zhang
- BE2018672 Yuquan Zhang
- BE2018672 Yuquan Zhang
- BE2018672 Yuquan Zhang
- BE2018672 Yuquan Zhang
Collapse
Affiliation(s)
- Rong Du
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Xi Cheng
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Jingjing Ji
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Yang Lu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Yuanyuan Xie
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Weina Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Yanhua Xu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Yuquan Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, No.20, Xisi Road, Nantong, Jiangsu Province, 226001, China.
| |
Collapse
|
4
|
Regulatory T Cells Overexpressing Peli1 Show Better Efficacy in Repairing Ovarian Endocrine Function in Autoimmune Premature Ovarian Insufficiency. J Transl Med 2023; 103:100005. [PMID: 37039145 DOI: 10.1016/j.labinv.2022.100005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/12/2022] [Accepted: 09/21/2022] [Indexed: 01/11/2023] Open
Abstract
Regulatory T (Treg) cell dysfunction is involved in the pathogenesis of autoimmune premature ovarian insufficiency (POI). Adoptive transfer of Treg cells has been shown to be effective in the treatment of autoimmune POI in mice. However, the therapeutic effect of Treg cell therapy is limited because the phenotype and function of Treg cells is not properly maintained when they are reinfused in an inflammatory environment. Therefore, enhancing the function of Treg cells using genetic engineering is of great significance for improving the efficacy of Treg cells in the treatment of immune diseases. In this study, we investigated the role of the E3 ubiquitinated ligase Pellino 1 (Peli1) in the proliferation and immunosuppressive function of Treg cells and the therapeutic effect of Treg cells overexpressing Peli1 on autoimmune POI. The results showed that the overexpression of Peli1 promoted cell proliferation and enhanced the immunosuppressive function of Treg cells in vitro. After the adoptive transfer of Treg cells overexpressing Peli1 in autoimmune POI mice, the apoptosis rate of ovarian granulosa cells declined. The levels of the inflammatory inhibitors interleukin 10 and transforming growth factor-β as well as the ovarian hormone estradiol were elevated. The number of primordial, primary, secondary, and mature follicles was restored to a certain extent compared with those in control subjects. These results revealed that the adoptive transfer of Treg cells overexpressing Peli1 promoted its efficacy against zona pellucida protein 3 peptide-induced POI, which provides new insights into the treatment of autoimmune POI.
Collapse
|
5
|
Shi L, Zhang Z, Deng M, Zheng F, Liu W, Ye S. Biological mechanisms and applied prospects of mesenchymal stem cells in premature ovarian failure. Medicine (Baltimore) 2022; 101:e30013. [PMID: 35960112 PMCID: PMC9371578 DOI: 10.1097/md.0000000000030013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/05/2022] [Accepted: 06/06/2022] [Indexed: 01/04/2023] Open
Abstract
Premature ovarian failure (POF), also known as primary ovarian insufficiency (POI), refers to the loss of ovarian function in women after puberty and before the age of 40 characterized by high serum gonadotropins and low estrogen, irregular menstruation, amenorrhea, and decreased fertility. However, the specific pathogenesis of POF is unexplained, and there is no effective therapy for its damaged ovarian tissue structure and reduced reserve function. Mesenchymal stem cells (MSCs), with multidirectional differentiation potential and self-renewal ability, as well as the cytokines and exosomes they secrete, have been studied and tested to play an active therapeutic role in a variety of degenerative pathologies, and MSCs are the most widely used stem cells in regenerative medicine. MSCs can reverse POI and enhance ovarian reserve function through differentiation into granulosa cells (GCs), immune regulation, secretion of cytokines and other nutritional factors, reduction of GCs apoptosis, and promotion of GCs regeneration. Many studies have proved that MSCs may have a restorative effect on the structure and fertility of injured ovarian tissues and turn to be a useful clinical approach to the treatment of patients with POF in recent years. We intend to use MSCs-based therapy to completely reverse POI in the future.
Collapse
Affiliation(s)
- Lan Shi
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Zhifen Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Miao Deng
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Fangyuan Zheng
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Wenhua Liu
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Shujin Ye
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
6
|
Kanatsu-Shinohara M, Lee J, Miyazaki T, Morimoto H, Shinohara T. Adeno-associated-virus-mediated gene delivery to ovaries restores fertility in congenital infertile mice. Cell Rep Med 2022; 3:100606. [PMID: 35584625 PMCID: PMC9133397 DOI: 10.1016/j.xcrm.2022.100606] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/24/2022] [Accepted: 03/24/2022] [Indexed: 11/20/2022]
Abstract
Oocytes and granulosa cells closely interact with each other during follicular development, and a lack of appropriate signaling between them results in infertility. Attempts to manipulate oocyte microenvironment have been impeded by the impermeability of the blood-follicle barrier (BFB). To establish a strategy for manipulating oogenesis, we use adeno-associated viruses (AAVs), which have a unique ability of transcytosis. Microinjecting of AAVs into the ovarian stroma penetrates the BFB and achieves long-term gene expression. Introduction of an AAV carrying the mouse Kitl gene restores oogenesis in congenitally infertile KitlSl-t/KitlSl-t mutant mouse ovaries, which lack Kitl expression but contain only primordial follicles. Healthy offspring without AAV integration are born by natural mating. Therefore, AAV-mediated gene delivery not only provides a means for studying oocyte-granulosa interactions through the manipulation of the oocyte microenvironment but could also be a powerful method to treat female infertility resulting from somatic cell defects.
Collapse
Affiliation(s)
- Mito Kanatsu-Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; AMED-CREST, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Jiyoung Lee
- Department of Epigenetics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Takehiro Miyazaki
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Hiroko Morimoto
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Takashi Shinohara
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; AMED-CREST, Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
7
|
Sun B, Xu L, Bi W, Ou WB. SALL4 Oncogenic Function in Cancers: Mechanisms and Therapeutic Relevance. Int J Mol Sci 2022; 23:ijms23042053. [PMID: 35216168 PMCID: PMC8876671 DOI: 10.3390/ijms23042053] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023] Open
Abstract
SALL4, a member of the SALL family, is an embryonic stem cell regulator involved in self-renewal and pluripotency. Recently, SALL4 overexpression was found in malignant cancers, including lung cancer, hepatocellular carcinoma, breast cancer, gastric cancer, colorectal cancer, osteosarcoma, acute myeloid leukemia, ovarian cancer, and glioma. This review updates recent advances of our knowledge of the biology of SALL4 with a focus on its mechanisms and regulatory functions in tumors and human hematopoiesis. SALL4 overexpression promotes proliferation, development, invasion, and migration in cancers through activation of the Wnt/β-catenin, PI3K/AKT, and Notch signaling pathways; expression of mitochondrial oxidative phosphorylation genes; and inhibition of the expression of the Bcl-2 family, caspase-related proteins, and death receptors. Additionally, SALL4 regulates tumor progression correlated with the immune microenvironment involved in the TNF family and gene expression through epigenetic mechanisms, consequently affecting hematopoiesis. Therefore, SALL4 plays a critical oncogenic role in gene transcription and tumor growth. However, there are still some scientific hypotheses to be tested regarding whether SALL4 is a therapeutic target, such as different tumor microenvironments and drug resistance. Thus, an in-depth understanding and study of the functions and mechanisms of SALL4 in cancer may help develop novel strategies for cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Wen-Bin Ou
- Correspondence: ; Tel./Fax: +86-571-8684-3303
| |
Collapse
|
8
|
Medenica S, Abazovic D, Ljubić A, Vukovic J, Begovic A, Cucinella G, Zaami S, Gullo G. The Role of Cell and Gene Therapies in the Treatment of Infertility in Patients with Thyroid Autoimmunity. Int J Endocrinol 2022; 2022:4842316. [PMID: 36081621 PMCID: PMC9448571 DOI: 10.1155/2022/4842316] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/03/2022] [Indexed: 12/02/2022] Open
Abstract
There is a rising incidence of infertility worldwide, and many couples experience difficulties conceiving nowadays. Thyroid autoimmunity (TAI) is recognized as one of the major female infertility causes related to a diminished ovarian reserve and potentially impaired oocyte maturation and embryo development, causing adverse pregnancy outcomes. Growing evidence has highlighted its impact on spontaneously achieved pregnancy and pregnancy achieved by in vitro fertilization. Despite the influence of thyroid hormones on the male reproductive system, there is insufficient data on the association between TAI and male infertility. In past years, significant progress has been achieved in cell and gene therapies as emerging treatment options for infertility. Cell therapies utilize living cells to restore healthy tissue microenvironment and homeostasis and usually involve platelet-rich plasma and various stem cells. Using stem cells as therapeutic agents has many advantages, including simple sampling, abundant sources, poor immunogenicity, and elimination of ethical concerns. Mesenchymal Stem Cells (MSCs) represent a heterogeneous fraction of self-renewal, multipotent non-hematopoietic stem cells that display profound immunomodulatory and immunosuppressive features and promising therapeutic effects. Infertility has a genetic component in about half of all cases, although most of its genetic causes are still unknown. Hence, it is essential to identify genes involved in meiosis, DNA repair, ovarian development, steroidogenesis, and folliculogenesis, as well as those involved in spermatogenesis in order to develop potential gene therapies for infertility. Despite advances in therapy approaches such as biological agents, autoimmune disorders remain impossible to cure. Recent research demonstrates the remarkable therapeutic effectiveness of MSCs in a wide array of autoimmune diseases. TAI is one of many autoimmune disorders that can benefit from the use of MSCs, which can be derived from bone marrow and adipose tissue. Cell and gene therapies hold great potential for treating autoimmune conditions, although further research is still needed.
Collapse
Affiliation(s)
- Sanja Medenica
- Department of Endocrinology, Internal Medicine Clinic, Clinical Center of Montenegro, School of Medicine, University of Montenegro, Podgorica, Montenegro
| | | | - Aleksandar Ljubić
- Biocell Hospital, Belgrade, Serbia
- Special Gynecology Hospital with Maternity Ward Jevremova, Belgrade, Serbia
- Libertas International University, Dubrovnik, Croatia
| | | | | | - Gaspare Cucinella
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF UNIT, University of Palermo, Palermo, Italy
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Giuseppe Gullo
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF UNIT, University of Palermo, Palermo, Italy
| |
Collapse
|
9
|
Genetic etiologic analysis in 74 Chinese Han women with idiopathic premature ovarian insufficiency by combined molecular genetic testing. J Assist Reprod Genet 2021; 38:965-978. [PMID: 33538981 DOI: 10.1007/s10815-021-02083-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/19/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To identify the disease-causing genes of Chinese Han women with idiopathic premature ovarian insufficiency (POI). METHODS Seventy-four Chinese Han women with idiopathic POI were collected to analyze the genetic etiology. Triplet repeat-primed polymerase chain reaction (TP-PCR) was performed to screen the FMR1 (CGG)n premutation, and then 60 POI-related genes were sequenced by targeted next-generation sequencing (NGS) in POI patients with normal FMR1. RESULTS A total of one patient (1/74) with FMR1 premutation was identified. Targeted NGS revealed that 15.07% (11/73) patients had pathogenic or likely pathogenic variants of Mendelian genes (FOXL2, EIF2B2, CYP17A1, CLPP, MCM9, GDF9, MSH5, ERCC6, POLG). Ten novel variants in six Mendelian genes were identified, such as CLPP c.355A>C (p.I119L) and c.688A>C (p.M230L), MCM9 c.1157C>T (p.T386M) and c.1291A>G (p.M431V), GDF9 c. 238C>T (p.Q80X), MSH5 c.604G>C (p.G202R) and c.2063T>C (p.I688T), ERCC6 c.C1769C>T (p.P590L), POLG c.2832G>C (p.E944D), and c.2821A>G (p.I941V). CONCLUSION This study suggested targeted NGS was an efficient etiologic test for idiopathic POI patients without FMR1 premutation and enriched the variant spectrum of POI-related genes.
Collapse
|
10
|
Treatment potential of bone marrow-derived stem cells in women with diminished ovarian reserves and premature ovarian failure. Curr Opin Obstet Gynecol 2020; 31:156-162. [PMID: 30855290 DOI: 10.1097/gco.0000000000000531] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW We review the techniques recently tested in both animal models and humans to provide a state-of-the-art on adult stem cell ovarian transplant to achieve ovarian rejuvenation in patients with diminished ovarian reserves. RECENT FINDINGS As the firsts reports of spontaneous pregnancies achieved after bone marrow transplantation in oncologic women with primary ovarian insufficiency, increasing evidence supports the regenerative effects of stem cell-based therapies in the ovarian niche. Adult stem cells from several origins promote follicular development, increase ovarian local vascularization, increase follicle and stromal cell proliferation and reduce cell apoptosis and follicular atresia, although they do not modify embryo quality. Therefore, residual quiescent follicles of aged or damaged ovaries might produce competent oocytes in an adequate ovarian environment. Nevertheless, further research is needed to properly evaluate underlying mechanisms, identify best cell sources and design less invasive infusion techniques. SUMMARY Stem cells may be a relevant therapeutic alternative for ovary regeneration and follicular development in patients with impaired ovaries, such as poor ovarian responders or women diagnosed with primary ovarian insufficiency.
Collapse
|
11
|
Rai S, Ashish, Kumari P, Singh A, Singh R. Correlation of follicle-stimulating hormone receptor gene Asn 680 Ser (rs6166) polymorphism with female infertility. J Family Med Prim Care 2019; 8:3356-3361. [PMID: 31742168 PMCID: PMC6857416 DOI: 10.4103/jfmpc.jfmpc_685_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 08/30/2019] [Accepted: 09/19/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND AIMS Female infertility is a complex multifactorial, and polygenic disease associated with genetic factors plays an essential role in its formation and follicle development, oocyte maturation, and steroidogenesis regulation in the ovary. The aim here is too study the genetic association between follicle-stimulating hormone receptor (FSHR) Asn680Ser; (rs6166) gene polymorphism with female Infertility in our population. METHODS In this prospective case-control study, we enrolled 106 infertile and 164 unrelated healthy control individuals. Genomic DNA was extracted from the 5 ml of venous blood using the modified salting-out method. A polymerase chain reaction-amplified exon 10 of FSHR and purified PCR products were sequenced on an ABI 3730XL DNA sequencer. The data were analyzed statistically. RESULTS We found that the presence of rare allele "G" and heterozygous and common homozygous genotypes significantly increased the risk of female infertility. No significant change in the FSHR 191756 G >A genotype frequency was observed, regardless of chromosomal integrity. The genotype frequency distribution of locus 680 was consistent with the Hardy-Weinberg Equilibrium (HWE) in both groups (P > 0.05). CONCLUSION No significant differences were found in allelic variants frequency and genotype distribution between each category of subjects when analyzing the FSHR SNPs in the exonic region (P value >0.05). FSHR Asn680Ser polymorphisms and female infertility (P > 0.05). Variations in FSHR gene have an essential influence on ovarian function and can account for several defects of female fertility. FSHR Asn680Ser (rs6166) gene polymorphism is associated with female infertility and can be used as a relevant molecular biomarker to identify the risk of infertility in our population. This finding can be important for disease pathogenesis.
Collapse
Affiliation(s)
- Sangeeta Rai
- Department of Obstetrics and Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashish
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Preeti Kumari
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Anup Singh
- Department of Geriatric Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Royana Singh
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|