1
|
Mu L, Wang G, Yang X, Liang J, Tong H, Li L, Geng K, Bo Y, Hu X, Yang R, Xu X, Zhang Y, Zhang H. Physiological premature aging of ovarian blood vessels leads to decline in fertility in middle-aged mice. Nat Commun 2025; 16:72. [PMID: 39747922 PMCID: PMC11695630 DOI: 10.1038/s41467-024-55509-y] [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: 05/25/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Ovarian function declines significantly as females enter middle-age, but the mechanisms underlying this decline remain unclear. Here, we utilize whole-organ imaging to observe a notable decrease in ovarian blood vessel (oBV) density and angiogenesis intensity of middle-aged mice. This leads to a diminished blood supply to the ovaries, resulting in inadequate development and maturation of ovarian follicles. Utilizing genetic-modified mouse models, we demonstrate that granulosa cell secreted VEGFA governs ovarian angiogenesis, but the physiological decline in oBV is not attributed to VEGFA insufficiency. Instead, through single-cell sequencing, we identify the aging of the ovarian vascular endothelium as the primary factor contributing to oBV decline. Consequently, the administration of salidroside, a natural compound that is functional to reverse oBV aging and promote ovarian angiogenesis, significantly enhances ovarian blood supply and improve fertility in older females. Our findings highlight that enhancing oBV function is a promising strategy to boost fertility in females.
Collapse
Affiliation(s)
- Lu Mu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Ge Wang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xuebing Yang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jing Liang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Huan Tong
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Lingyu Li
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Kaiying Geng
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yingnan Bo
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xindi Hu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Ruobing Yang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xueqiang Xu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yan Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Hua Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
| |
Collapse
|
2
|
Yang X, Zhang Y, Zhang H. Cellular and molecular regulations of oocyte selection and activation in mammals. Curr Top Dev Biol 2024; 162:283-315. [PMID: 40180512 DOI: 10.1016/bs.ctdb.2024.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Oocytes, a uniquely pivotal cell population, play a central role in species continuity. In mammals, oogenesis involves distinct processes characterized by sequential rounds of selection, arrest, and activation to produce a limited number of mature eggs, fitting their high-survival yet high-cost fertility. During the embryonic phase, oocytes undergo intensive selection via cytoplasmic and organelle enrichment, accompanied by the onset and arrest of meiosis, thereby establishing primordial follicles (PFs) as a finite reproductive reserve. Subsequently, the majority of primary oocytes enter a dormant state and are gradually recruited through a process termed follicle activation, essential for maintaining orderly fertility. Following activation, oocytes undergo rapid growth, experiencing cycles of arrest and activation regulated by endocrine and paracrine signals, ultimately forming fertilizable eggs. Over the past two decades, advancements in genetically modified animal models, high-resolution imaging, and omics technologies have significantly enhanced our understanding of the cellular and molecular mechanisms that govern mammalian oogenesis. These advances offer profound insights into the regulatory mechanisms of mammalian reproduction and associated female infertility disorders. In this chapter, we provide an overview of current knowledge in mammalian oogenesis, with a particular emphasis on oocyte selection and activation in vivo.
Collapse
Affiliation(s)
- Xuebing Yang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Yan Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, P.R. China
| | - Hua Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, P.R. China.
| |
Collapse
|
3
|
Yan Y, Zhang H, Xu R, Luo L, Yin L, Wu H, Zhang Y, Li C, Lu S, Tang Y, Zhao X, Pan M, Wei Q, Peng S, Ma B. Single-cell sequencing reveals the transcriptional alternations of 17β-estradiol suppressing primordial follicle formation in neonatal mouse ovaries. Cell Prolif 2024; 57:e13713. [PMID: 38988058 PMCID: PMC11503257 DOI: 10.1111/cpr.13713] [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: 02/22/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
Estrogen has been implicated in multiple biological processes, but the variation underlying estrogen-mediated primordial follicle (PF) formation remains unclear. Here, we show that 17β-estradiol (E2) treatment of neonatal mice led to the inhibition of PF formation and cell proliferation. Single-cell RNA sequencing (scRNA-seq) revealed that E2 treatment caused significant changes in the transcriptome of oocytes and somatic cells. E2 treatment disrupted the synchronised development of oocytes, pre-granulosa (PG) cells and stromal cells. Mechanistically, E2 treatment disrupted several signalling pathways critical to PF formation, especially down-regulating the Kitl and Smad1/3/4/5/7 expression, reducing the frequency and number of cell communication. In addition, E2 treatment influenced key gene expression, mitochondrial function of oocytes, the recruitment and maintenance of PG cells, the cell proliferation of somatic cells, as well as disordered the ovarian microenvironment. This study not only revealed insights into the regulatory role of estrogen during PF formation, but also filled in knowledge of dramatic changes in perinatal hormones, which are critical for the physiological significance of understanding hormone changes and reproductive protection.
Collapse
Affiliation(s)
- Yutong Yan
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Hui Zhang
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Rui Xu
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Linglin Luo
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Lu Yin
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Hao Wu
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Yiqian Zhang
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Chan Li
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Sihai Lu
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Yaju Tang
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Xiaoe Zhao
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Menghao Pan
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Qiang Wei
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Sha Peng
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| | - Baohua Ma
- College of Veterinary MedicineNorthwest A&F UniversityYanglingShaanxiChina
- Key Laboratory of Animal Biotechnology of the Ministry of AgricultureNorthwest A&F UniversityYanglingShaanxiChina
| |
Collapse
|
4
|
Nicol B, Estermann MA, Yao HHC, Mellouk N. Becoming female: Ovarian differentiation from an evolutionary perspective. Front Cell Dev Biol 2022; 10:944776. [PMID: 36158204 PMCID: PMC9490121 DOI: 10.3389/fcell.2022.944776] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 08/16/2022] [Indexed: 01/09/2023] Open
Abstract
Differentiation of the bipotential gonadal primordium into ovaries and testes is a common process among vertebrate species. While vertebrate ovaries eventually share the same functions of producing oocytes and estrogens, ovarian differentiation relies on different morphogenetic, cellular, and molecular cues depending on species. The aim of this review is to highlight the conserved and divergent features of ovarian differentiation through an evolutionary perspective. From teleosts to mammals, each clade or species has a different story to tell. For this purpose, this review focuses on three specific aspects of ovarian differentiation: ovarian morphogenesis, the evolution of the role of estrogens on ovarian differentiation and the molecular pathways involved in granulosa cell determination and maintenance.
Collapse
Affiliation(s)
- Barbara Nicol
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States,*Correspondence: Barbara Nicol,
| | - Martin A. Estermann
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Humphrey H-C Yao
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Namya Mellouk
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy en Josas, France
| |
Collapse
|
5
|
Abstract
In vitro systems capable of reconstituting the process of mouse oogenesis are now being established to help develop further understanding of the mechanisms underlying oocyte/follicle development and differentiation. These systems could also help increase the production of useful livestock or genetically modified animals, and aid in identifying the causes of infertility in humans. Recently, we revealed, using an in vitro system for recapitulating oogenesis, that the activation of the estrogen signaling pathway induces abnormal follicle formation, that blocking estrogen-induced expression of anti-Müllerian hormone is crucial for normal follicle formation, and that the production of α-fetoprotein in fetal liver tissue is involved in normal in vivo follicle formation. In mouse fetuses, follicle formation is not carried out by factors within the ovaries but is instead orchestrated by distal endocrine factors. This review outlines findings from genetics, endocrinology, and in vitro studies regarding the factors that can affect the formation of primordial follicles in mammals.
Collapse
|
6
|
Monget P, McNatty K, Monniaux D. The Crazy Ovary. Genes (Basel) 2021; 12:928. [PMID: 34207147 PMCID: PMC8234655 DOI: 10.3390/genes12060928] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/08/2021] [Accepted: 06/15/2021] [Indexed: 11/16/2022] Open
Abstract
From fetal life until senescence, the ovary is an extremely active tissue undergoing continuous structural and functional changes. These ever-changing events are best summarized by a quotation attributed to Plato when describing motion in space and time-'nothing ever is but is always becoming…'. With respect to the ovary, these changes include, at the beginning, the processes of follicular formation and thereafter those of follicular growth and atresia, steroidogenesis, oocyte maturation, and decisions relating to the number of mature oocytes that are ovulated for fertilization and the role of the corpus luteum. The aims of this review are to offer some examples of these complex and hitherto unknown processes. The ones herein have been elucidated from studies undertaken in vitro or from normal in vivo events, natural genetic mutations or after experimental inactivation of gene function. Specifically, this review offers insights concerning the initiation of follicular growth, pathologies relating to poly-ovular follicles, the consequences of premature loss of germ cells or oocytes loss, the roles of AMH (anti-Müllerian hormone) and BMP (bone morphogenetic protein) genes in regulating follicular growth and ovulation rate together with species differences in maintaining luteal function during pregnancy. Collectively, the evidence suggests that the oocyte is a key organizer of normal ovarian function. It has been shown to influence the phenotype of the adjacent somatic cells, the growth and maturation of the follicle, and to determine the ovulation rate. When germ cells or oocytes are lost prematurely, the ovary becomes disorganized and a wide range of pathologies may arise.
Collapse
Affiliation(s)
- Philippe Monget
- UMR INRAE-CNRS-IFCE-Université de Tours, 37380 Nouzilly, France;
| | - Ken McNatty
- School of Biological Sciences, Victoria University of Wellington, Wellington 6140, New Zealand;
| | | |
Collapse
|
7
|
Watanabe R, Sasaki S, Kimura N. Activation of autophagy in early neonatal mice increases primordial follicle number and improves lifelong fertility†. Biol Reprod 2021; 102:399-411. [PMID: 31566206 DOI: 10.1093/biolre/ioz179] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 08/21/2019] [Accepted: 09/14/2019] [Indexed: 12/17/2022] Open
Abstract
The number of stockpiled primordial follicles is thought to be responsible for the fate of female fertility and reproductive lifetime. We previously reported that starvation in nonsuckling early neonatal mice increases the number of primordial follicles with concomitant autophagy activation, suggesting that autophagy may accelerate the formation of primordial follicles. In this study, we attempted to upregulate the numbers of primordial follicles by administering an autophagy inducer and evaluated the progress of primordial follicle formation and their fertility during the life of the mice. To induce autophagy, mice were intraperitoneally injected with the Tat-beclin1 D-11 peptide (0.02 mg/g body weight) at 6-54 h or 60-84 h after birth. In animals that received Tat-beclin 1 D-11 by 54 h after birth, the primordial follicle numbers were significantly increased compared with the control group at 60 h. The ratio of expressed LC3-II/LC3-I proteins was also significantly greater. The numbers of littermates from pregnant females that had been treated with Tat-beclin 1 D-11 were maintained at remarkably greater levels until 10 months old. These results were supported by an abundance of primordial follicles at even 13-15 months old.
Collapse
Affiliation(s)
- Ren Watanabe
- Laboratory of Animal Reproduction, Graduate School of Agricultural Science, Yamagata University, Tsuruoka, Japan.,Japan Society for the Promotion of Science (JSPS) Research Fellowships for Young Scientists, Tokyo, Japan
| | - Sho Sasaki
- Laboratory of Animal Reproduction, Graduate School of Agricultural Science, Yamagata University, Tsuruoka, Japan
| | - Naoko Kimura
- Laboratory of Animal Reproduction, Graduate School of Agricultural Science, Yamagata University, Tsuruoka, Japan
| |
Collapse
|
8
|
Guo S, Quan S, Zou S. Roles of the Notch Signaling Pathway in Ovarian Functioning. Reprod Sci 2021; 28:2770-2778. [PMID: 34008156 DOI: 10.1007/s43032-021-00610-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/05/2021] [Indexed: 11/30/2022]
Abstract
The Notch signaling pathway regulates cell invasion, adhesion, proliferation, apoptosis, and differentiation via cell-to-cell interactions and plays important physiological roles in the ovary. This review summarizes current knowledge about the Notch signaling pathway in relation to ovarian functions and reveals the potential underlying mechanisms. We conducted an in-depth review of relevant literature to determine the current status of research into the Notch signaling pathway in relation to ovarian functioning and reveal potential underlying mechanisms. The activation of different Notch receptors promotes the formation of primordial follicles and proliferation of granulosa cells and inhibits steroid secretion. Abnormal regulation of the Notch signaling pathway or direct mutations might lead to over-activation or under-activation of the receptors, resulting in Notch upregulation or downregulation. It can also disrupt the normal physiological functions of the ovary. The lncRNA HOTAIR and growth hormones improved premature ovarian failure (POF) and promoted follicle maturation in a mouse model of POF by upregulating Notch1 expression. They also stimulated the Notch1 signaling pathway, increased the level of plasma estradiol, and decreased the level of plasma follicle-stimulating hormone. Thus, Notch1 could serve as a novel therapeutic target for POF. Several studies have reported multiple roles of Notch in regulating female primordial follicle formation and follicle maturation. Direct mutations in Notch-related molecules or abnormal gene regulation in the signaling pathway can lead to ovarian dysfunction. However, the underlying mechanisms are not fully understood.
Collapse
Affiliation(s)
- Shuhan Guo
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Song Quan
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - Siyi Zou
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Nanfang Hospital/The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Cai H, Liu B, Wang H, Sun G, Feng L, Chen Z, Zhou J, Zhang J, Zhang T, He M, Yang T, Guo Q, Teng Z, Xin Q, Zhou B, Zhang H, Xia G, Wang C. SP1 governs primordial folliculogenesis by regulating pregranulosa cell development in mice. J Mol Cell Biol 2021; 12:230-244. [PMID: 31282930 PMCID: PMC7181717 DOI: 10.1093/jmcb/mjz059] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/20/2019] [Accepted: 06/12/2019] [Indexed: 01/05/2023] Open
Abstract
Establishment of the primordial follicle (PF) pool is pivotal for the female reproductive lifespan; however, the mechanism of primordial folliculogenesis is poorly understood. Here, the transcription factor SP1 was shown to be essential for PF formation in mice. Our results showed that SP1 is present in both oocytes and somatic cells during PF formation in the ovary. Knockdown of Sp1 expression, especially in pregranulosa cells, significantly suppressed nest breakdown, oocyte apoptosis, and PF formation, suggesting that SP1 expressed by somatic cells functions in the process of primordial folliculogenesis. We further demonstrated that SP1 governs the recruitment and maintenance of Forkhead box L2-positive (FOXL2+) pregranulosa cells using an Lgr5-EGFP-IRES-CreERT2 (Lgr5-KI) reporter mouse model and a FOXL2+ cell-specific knockdown model. At the molecular level, SP1 functioned mainly through manipulation of NOTCH2 expression by binding directly to the promoter of the Notch2 gene. Finally, consistent with the critical role of granulosa cells in follicle survival in vitro, massive loss of oocytes in Sp1 knockdown ovaries was evidenced before puberty after the ovaries were transplanted under the renal capsules. Conclusively, our results reveal that SP1 controls the establishment of the ovarian reserve by regulating pregranulosa cell development in the mammalian ovary.
Collapse
Affiliation(s)
- Han Cai
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Bingying Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Huarong Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen 361102, China
| | - Guanghong Sun
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Lizhao Feng
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Ziqi Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jiaqi Zhou
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jiawei Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Tuo Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Meina He
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Tingting Yang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qirui Guo
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Zhen Teng
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qiliang Xin
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Bo Zhou
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hua Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.,Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Sciences, Ningxia University, Yinchuan 750021, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
10
|
Orzechowska M, Anusewicz D, Bednarek AK. Functional Gene Expression Differentiation of the Notch Signaling Pathway in Female Reproductive Tract Tissues-A Comprehensive Review With Analysis. Front Cell Dev Biol 2021; 8:592616. [PMID: 33384996 PMCID: PMC7770115 DOI: 10.3389/fcell.2020.592616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
The Notch pathway involves evolutionarily conserved signaling regulating the development of the female tract organs such as breast, ovary, cervix, and uterine endometrium. A great number of studies revealed Notch aberrancies in association with their carcinogenesis and disease progression, the management of which is still challenging. The present study is a comprehensive review of the available literature on Notch signaling during the normal development and carcinogenesis of the female tract organs. The review has been enriched with our analyses of the TCGA data including breast, cervical, ovarian, and endometrial carcinomas concerning the effects of Notch signaling at two levels: the core components and downstream effectors, hence filling the lack of global overview of Notch-driven carcinogenesis and disease progression. Phenotype heterogeneity regarding Notch signaling was projected in two uniform manifold approximation and projection algorithm dimensions, preceded by the principal component analysis step reducing the data burden. Additionally, overall and disease-free survival analyses were performed with the optimal cutpoint determination by Evaluate Cutpoints software to establish the character of particular Notch components in tumorigenesis. In addition to the review, we demonstrated separate models of the examined cancers of the Notch pathway and its targets, although expression profiles of all normal tissues were much more similar to each other than to its cancerous compartments. Such Notch-driven cancerous differentiation resulted in a case of opposite association with DFS and OS. As a consequence, target genes also show very distinct profiles including genes associated with cell proliferation and differentiation, energy metabolism, or the EMT. In conclusion, the observed Notch associations with the female tract malignancies resulted from differential expression of target genes. This may influence a future analysis to search for new therapeutic targets based on specific Notch pathway profiles.
Collapse
Affiliation(s)
| | - Dorota Anusewicz
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
11
|
Heffner K, Hizal DB, Majewska NI, Kumar S, Dhara VG, Zhu J, Bowen M, Hatton D, Yerganian G, Yerganian A, O'Meally R, Cole R, Betenbaugh M. Expanded Chinese hamster organ and cell line proteomics profiling reveals tissue-specific functionalities. Sci Rep 2020; 10:15841. [PMID: 32985598 PMCID: PMC7522264 DOI: 10.1038/s41598-020-72959-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/31/2020] [Indexed: 02/07/2023] Open
Abstract
Chinese hamster ovary (CHO) cells are the predominant production vehicle for biotherapeutics. Quantitative proteomics data were obtained from two CHO cell lines (CHO-S and CHO DG44) and compared with seven Chinese hamster (Cricetulus griseus) tissues (brain, heart, kidney, liver, lung, ovary and spleen) by tandem mass tag (TMT) labeling followed by mass spectrometry, providing a comprehensive hamster tissue and cell line proteomics atlas. Of the 8470 unique proteins identified, high similarity was observed between CHO-S and CHO DG44 and included increases in proteins involved in DNA replication, cell cycle, RNA processing, and chromosome processing. Alternatively, gene ontology and pathway analysis in tissues indicated increased protein intensities related to important tissue functionalities. Proteins enriched in the brain included those involved in acidic amino acid metabolism, Golgi apparatus, and ion and phospholipid transport. The lung showed enrichment in proteins involved in BCAA catabolism, ROS metabolism, vesicle trafficking, and lipid synthesis while the ovary exhibited enrichments in extracellular matrix and adhesion proteins. The heart proteome included vasoconstriction, complement activation, and lipoprotein metabolism enrichments. These detailed comparisons of CHO cell lines and hamster tissues will enhance understanding of the relationship between proteins and tissue function and pinpoint potential pathways of biotechnological relevance for future cell engineering.
Collapse
Affiliation(s)
- Kelley Heffner
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,AstraZeneca, Cell Culture and Fermentation Sciences, Gaithersburg, MD, USA
| | - Deniz Baycin Hizal
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Natalia I Majewska
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,AstraZeneca, Cell Culture and Fermentation Sciences, Gaithersburg, MD, USA
| | - Swetha Kumar
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Venkata Gayatri Dhara
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jie Zhu
- AstraZeneca, Cell Culture and Fermentation Sciences, Gaithersburg, MD, USA
| | - Michael Bowen
- Allogene Therapeutics, Product and Process Development, South San Francisco, CA, USA
| | - Diane Hatton
- AstraZeneca, Cell Culture and Fermentation Sciences, Gaithersburg, MD, USA
| | | | | | - Robert O'Meally
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Robert Cole
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Michael Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
12
|
Grigoletto L, Santana MHA, Bressan FF, Eler JP, Nogueira MFG, Kadarmideen HN, Baruselli PS, Ferraz JBS, Brito LF. Genetic Parameters and Genome-Wide Association Studies for Anti-Müllerian Hormone Levels and Antral Follicle Populations Measured After Estrus Synchronization in Nellore Cattle. Animals (Basel) 2020; 10:E1185. [PMID: 32668804 PMCID: PMC7401547 DOI: 10.3390/ani10071185] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022] Open
Abstract
Reproductive efficiency plays a major role in the long-term sustainability of livestock industries and can be improved through genetic and genomic selection. This study aimed to estimate genetic parameters (heritability and genetic correlation) and identify genomic regions and candidate genes associated with anti-Müllerian hormone levels (AMH) and antral follicle populations measured after estrous synchronization (AFP) in Nellore cattle. The datasets included phenotypic records for 1099 and 289 Nellore females for AFP and AMH, respectively, high-density single nucleotide polymorphism (SNP) genotypes for 944 animals, and 4129 individuals in the pedigree. The heritability estimates for AMH and AFP were 0.28 ± 0.07 and 0.30 ± 0.09, and the traits were highly and positively genetically correlated (rG = 0.81 ± 0.02). These findings indicated that these traits can be improved through selective breeding, and substantial indirect genetic gains are expected by selecting for only one of the two traits. A total of 31 genomic regions were shown to be associated with AMH or AFP, and two genomic regions located on BTA1 (64.9-65.0 Mb and 109.1-109.2 Mb) overlapped between the traits. Various candidate genes were identified to be potentially linked to important biological processes such as ovulation, tissue remodeling, and the immune system. Our findings support the use of AMH and AFP as indicator traits to genetically improve fertility rates in Nellore cattle and identify better oocyte donors.
Collapse
Affiliation(s)
- Laís Grigoletto
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, 13635-900 São Paulo, Brazil; (M.H.A.S.); (F.F.B.); (J.P.E.); (J.B.S.F.)
| | - Miguel Henrique Almeida Santana
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, 13635-900 São Paulo, Brazil; (M.H.A.S.); (F.F.B.); (J.P.E.); (J.B.S.F.)
| | - Fabiana Fernandes Bressan
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, 13635-900 São Paulo, Brazil; (M.H.A.S.); (F.F.B.); (J.P.E.); (J.B.S.F.)
| | - Joanir Pereira Eler
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, 13635-900 São Paulo, Brazil; (M.H.A.S.); (F.F.B.); (J.P.E.); (J.B.S.F.)
| | - Marcelo Fábio Gouveia Nogueira
- Department of Biological Sciences, School of Sciences and Languages, São Paulo State University, Assis, 19806-900 São Paulo, Brazil;
| | - Haja N. Kadarmideen
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, 102500 Lyngby, Denmark;
| | - Pietro Sampaio Baruselli
- College of Veterinary Medicine and Animal Science, University of Sao Paulo, 05508-270 São Paulo, Brazil;
| | - José Bento Sterman Ferraz
- Department of Veterinary Medicine, College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, 13635-900 São Paulo, Brazil; (M.H.A.S.); (F.F.B.); (J.P.E.); (J.B.S.F.)
| | - Luiz F. Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
13
|
Yoshino T, Saito D. Epithelial-to-mesenchymal transition–based morphogenesis of dorsal mesentery and gonad. Semin Cell Dev Biol 2019; 92:105-112. [DOI: 10.1016/j.semcdb.2018.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/30/2018] [Accepted: 09/02/2018] [Indexed: 12/26/2022]
|
14
|
Huang K, Dang Y, Zhang P, Shen C, Sui X, Xia G, Qin Y, Jiao X, Wang C, Huo R, Chen ZJ. CAV1 regulates primordial follicle formation via the Notch2 signalling pathway and is associated with premature ovarian insufficiency in humans. Hum Reprod 2019; 33:2087-2095. [PMID: 30304446 DOI: 10.1093/humrep/dey299] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/19/2018] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION What is the function of CAV1 in folliculogenesis and female reproduction? SUMMARY ANSWER CAV1 regulates germline cyst breakdown and primordial follicle (PF) formation in mice, and CAV1 mutation may be related to premature ovarian insufficiency (POI). WHAT IS KNOWN ALREADY Pre-granulosa cells are essential for the establishment of the PF pool, which determines female fertility and reproductive lifespan. Cav1 participates in vascularization in fetal mouse ovaries. However, the role of CAV1 in early folliculogenesis and POI pathogenesis remains unclear. STUDY DESIGN, SIZE, DURATION Cav1 function was investigated in mice and Human Embryonic Kidney 293 cells. Ovaries (six per group) were randomly assigned to Cav1-vivo-morpholino, control and control-morpholino groups, and all experiments were repeated at least three times. To investigate CAV1 mutations in women, 200 Chinese women with POI and 200 control individuals with regular menstrual cycles and normal endocrine profiles were recruited from the Center for Reproductive Medicine of Shandong University between September 2012 and December 2013. PARTICIPANTS/MATERIALS, SETTING, METHODS Wild-type CD1 mice, Lgr5-EGFP-ires-CreERT2 (Lgr5-KI) reporter mice and Human Embryonic Kidney 293 cells were used for these experiments. Protein expression was detected by Western blot, and quantitative RT-PCR was used to detect gene expression. The expression pattern of CAV1 in mouse ovaries and the phenotype of Cav1 deficiency in mice were detected by immunofluorescence. Pre-granulosa cell proliferation in ovaries was detected by bromodeoxyuridine (BrdU) assay and immunofluorescence. The coding region of the CAV1 gene was sequenced in 200 women with POI and 200 controls. The functional effect of the novel mutation c.142 G > C (p.Glu48Gln) was investigated by Cell Counting Kit-8 (CCK8) assays and Western blot. MAIN RESULTS AND THE ROLE OF CHANCE We confirmed that Cav1 deficiency in mouse ovary induced by CAV1-vivo-morpholino resulted in more multi-oocyte follicles than in the control and control-morpholino groups (P < 0.01). Suppression of Cav1 decreased Leucine rich repeat containing G protein coupled receptor 5 (Lgr5)-positive cell proliferation (P < 0.01) and reduced the number of Lgr5 and Forkhead box L2 (Foxl2) double-positive cells (P < 0.01). Furthermore, suppression of Cav1 inhibited ovarian epithelial Lgr5-positive cell proliferation and differentiation through the Notch2 signalling pathway. Two of the POI women carried novel CAV1 mutations (c.45 C > G synonymous and c.142 G > C [Glu48Gln]). The deleterious effect of p.Glu48Gln was corroborated by showing that it adversely affected the function of CAV1 in cell proliferation and NOTCH2 expression in HEK293FT cells. LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The novel Glu48Gln mutation was only detected in one of 200 POI patients and we were unable to investigate its effects in the ovary. WIDER IMPLICATIONS OF THE FINDINGS The identification of CAV1 as a potentially causative gene for POI provides a theoretical basis to devise treatments for POI in women. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Basic Research Program of China (973 Programs: 2012CB944700; 2013CB945501; 2013CB911400; 2014CB943202), the National Key Research and Development Program of China (2016YFC1000604, 2017YFC1001301), the State Key Program of National Natural Science Foundation of China (81430029), and the National Natural Science Foundation of China (31571540, 81522018, 81471509, 81601245, 81701406, 81571406). The authors declare no competing financial interests.
Collapse
Affiliation(s)
- Kun Huang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yujie Dang
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics; The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education; Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China.,Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Pan Zhang
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Cong Shen
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Xuesong Sui
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yingying Qin
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics; The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education; Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Xue Jiao
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics; The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education; Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Ran Huo
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Zi-Jiang Chen
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics; The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education; Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan, China.,Center for Reproductive Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
15
|
da Silva AVA, Figueiredo FB, Menezes RC, Mendes-Junior AA, de Miranda LHM, Cupolillo E, Porrozzi R, Morgado FN. Morphophysiological changes in the splenic extracellular matrix of Leishmania infantum-naturally infected dogs is associated with alterations in lymphoid niches and the CD4+ T cell frequency in spleens. PLoS Negl Trop Dis 2018; 12:e0006445. [PMID: 29677186 PMCID: PMC5931683 DOI: 10.1371/journal.pntd.0006445] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 05/02/2018] [Accepted: 04/11/2018] [Indexed: 12/05/2022] Open
Abstract
The spleen is one of the main affected organs in canine visceral leishmaniasis (CVL). Disorganization of the splenic white pulp (SWP) has been associated with immunosuppression and disease progression. This study aims to assess structural and cellular changes in the splenic extracellular matrix of dogs with CVL, correlating these changes with the parasite load and clinical signs. Splenic fragments were collected from 41 naturally infected animals for parasite load quantification by quantitative PCR, histopathological analysis and immunohistochemistry for CD3+, CD4+, and CD8+ T cells; CD21+ B cells; Ki-67+, IFN-γ+, and IL-10+ cells; and the MMP-9 and ADAM-10 enzymes. Laminin, collagen and fibronectin deposition were also evaluated. The animals were grouped according to the level of SWP organization. SWP disorganization was accompanied by a reduction in the quantity of lymphoid follicles/mm2 (p > 0.0001). Animals with moderate to intense SWP disorganization showed more clinical signs (p = 0.021), higher laminin (p = 0.045) and collagen deposition (p = 0.036), higher MMP-9 expression (p = 0.035) and lower numbers of CD4+ T cells (p = 0.027) in the spleen than the animals with organized SWP. These data suggest that splenic structure and function are drastically altered and compromised during CVL. Infected dogs play important roles in the transmission of visceral leishmaniasis. These dogs are considered reservoirs of parasites in urban areas and fail to mount an efficient anti-Leishmania immune response. However, the specific immunosuppression profile is not completely understood. In our report, we evaluate and discuss the morphophysiological alterations in the spleens of dogs with visceral leishmaniasis. We found an association between extracellular matrix alterations and a failure to control the parasite load. We suggest a role for these alterations in hindering an immune response that is otherwise able to control the parasite load, thereby leading to disease progression. Our research contributes to the current knowledge of the immunopathology of canine visceral leishmaniasis.
Collapse
Affiliation(s)
- Aurea Virginia Andrade da Silva
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
- Graduate student of the Masters Program in Cellular and Molecular Biology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiano Borges Figueiredo
- Laboratório de Pesquisa Clínica em Dermatozoonoses em Animais Domésticos, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo Caldas Menezes
- Laboratório de Pesquisa Clínica em Dermatozoonoses em Animais Domésticos, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Arthur Augusto Mendes-Junior
- Laboratório de Pesquisa Clínica em Dermatozoonoses em Animais Domésticos, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luisa Helena Monteiro de Miranda
- Laboratório de Pesquisa Clínica em Dermatozoonoses em Animais Domésticos, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elisa Cupolillo
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato Porrozzi
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Nazaré Morgado
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
16
|
Huang K, Wang Y, Zhang T, He M, Sun G, Wen J, Yan H, Cai H, Yong C, Xia G, Wang C. JAK signaling regulates germline cyst breakdown and primordial follicle formation in mice. Biol Open 2018; 7:bio029470. [PMID: 29242197 PMCID: PMC5827266 DOI: 10.1242/bio.029470] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/11/2017] [Indexed: 01/15/2023] Open
Abstract
In female mammals, primordial follicles consist of two types of cells, namely, oocytes and pregranulosa cells that surround the oocytes. The size of the primordial follicle pool determines the reproductive ability of female mammals. However, the underlying mechanisms controlling primordial follicle assembly remain unclear. In this study, we show that oocyte-derived Janus kinase (JAK) signaling is vital for germline cyst breakdown and primordial follicle formation in vitro JAK2 and JAK3 activity is increased while germline cysts are breaking down. Inhibition of either JAK2 or JAK3 prevents germline cyst breakdown and primordial follicle formation. We further show that specific suppression of JAK2 delays germ cell loss through the downregulation of p53, but has no influence on pregranulosa cell proliferation. Alternatively, specific inhibition of JAK3 decreases pregranulosa cell proliferation by downregulating Notch2 signaling, implying that JAK3 acts on pregranulosa cells by controlling the extracellular secretion of oocyte-derived factors. In summary, our results indicate that JAK signaling contributes to germline cyst breakdown and primordial follicle formation by regulating oocyte loss and pregranulosa cell proliferation in the fetal mouse ovary. Our findings contribute to a better understanding of the molecular mechanism of mammalian folliculogenesis.
Collapse
Affiliation(s)
- Kun Huang
- Department of Physiology, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Ye Wang
- Department of Physiology, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Tuo Zhang
- Department of Physiology, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Meina He
- Department of Physiology, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Guanghong Sun
- Department of Physiology, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jia Wen
- Department of Physiology, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hao Yan
- Department of Physiology, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Han Cai
- Department of Physiology, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Changfu Yong
- Animal Disease Control and Prevention Center of Shapotou District, Zhongwei, Ningxia 755000, China
| | - Guoliang Xia
- Department of Physiology, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Chao Wang
- Department of Physiology, State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
17
|
Júnior GAO, Perez BC, Cole JB, Santana MHA, Silveira J, Mazzoni G, Ventura RV, Júnior MLS, Kadarmideen HN, Garrick DJ, Ferraz JBS. Genomic study and Medical Subject Headings enrichment analysis of early pregnancy rate and antral follicle numbers in Nelore heifers. J Anim Sci 2017; 95:4796-4812. [PMID: 29293733 PMCID: PMC6292327 DOI: 10.2527/jas2017.1752] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/24/2017] [Indexed: 12/18/2022] Open
Abstract
Zebu animals () are known to take longer to reach puberty compared with taurine animals (), limiting the supply of animals for harvest or breeding and impacting profitability. Genomic information can be a helpful tool to better understand complex traits and improve genetic gains. In this study, we performed a genomewide association study (GWAS) to identify genetic variants associated with reproductive traits in Nelore beef cattle. Heifer pregnancy (HP) was recorded for 1,267 genotyped animals distributed in 12 contemporary groups (CG) with an average pregnancy rate of 0.35 (±0.01). Disregarding one of these CG, the number of antral follicles (NF) was also collected for 937 of these animals, with an average of 11.53 (±4.43). The animals were organized in CG: 12 and 11 for HP and NF, respectively. Genes in linkage disequilibrium (LD) with the associated variants can be considered in a functional enrichment analysis to identify biological mechanisms involved in fertility. Medical Subject Headings (MeSH) were detected using the MESHR package, allowing the extraction of broad meanings from the gene lists provided by the GWAS. The estimated heritability for HP was 0.28 ± 0.07 and for NF was 0.49 ± 0.09, with the genomic correlation being -0.21 ± 0.29. The average LD between adjacent markers was 0.23 ± 0.01, and GWAS identified genomic windows that accounted for >1% of total genetic variance on chromosomes 5, 14, and 18 for HP and on chromosomes 2, 8, 11, 14, 15, 16, and 22 for NF. The MeSH enrichment analyses revealed significant ( < 0.05) terms associated with HP-"Munc18 Proteins," "Fucose," and "Hemoglobins"-and with NF-"Cathepsin B," "Receptors, Neuropeptide," and "Palmitic Acid." This is the first study in Nelore cattle introducing the concept of MeSH analysis. The genomic analyses contributed to a better understanding of the genetic control of the reproductive traits HP and NF and provide new selection strategies to improve beef production.
Collapse
Affiliation(s)
| | - B. C. Perez
- Universidade de São Paulo (USP), Pirassununga, SP, Brazil
| | - J. B. Cole
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705-2350
| | | | - J. Silveira
- Universidade de São Paulo (USP), Pirassununga, SP, Brazil
| | - G. Mazzoni
- Department of Veterinary and Animal Sciences, University of Copenhagen, Denmark
- Section of Systems Genomics, Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet, 2800 Kgs. Lyngby, Denmark
| | - R. V. Ventura
- Beef Improvement Opportunities, Guelph, ON N1K1E5, Canada
- Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, ON N1G2W1, Canada
| | | | - H. N. Kadarmideen
- Section of Systems Genomics, Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet, 2800 Kgs. Lyngby, Denmark
| | | | | |
Collapse
|
18
|
Wang C, Zhou B, Xia G. Mechanisms controlling germline cyst breakdown and primordial follicle formation. Cell Mol Life Sci 2017; 74:2547-2566. [PMID: 28197668 PMCID: PMC11107689 DOI: 10.1007/s00018-017-2480-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 12/11/2022]
Abstract
In fetal females, oogonia proliferate immediately after sex determination. The progress of mitosis in oogonia proceeds so rapidly that the incompletely divided cytoplasm of the sister cells forms cysts. The oogonia will then initiate meiosis and arrest at the diplotene stage of meiosis I, becoming oocytes. Within each germline cyst, oocytes with Balbiani bodies will survive after cyst breakdown (CBD). After CBD, each oocyte is enclosed by pre-granulosa cells to form a primordial follicle (PF). Notably, the PF pool formed perinatally will be the sole lifelong oocyte source of a female. Thus, elucidating the mechanisms of CBD and PF formation is not only meaningful for solving mysteries related to ovarian development but also contributes to the preservation of reproduction. However, the mechanisms that regulate these phenomena are largely unknown. This review summarizes the progress of cellular and molecular research on these processes in mice and humans.
Collapse
Affiliation(s)
- Chao Wang
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Bo Zhou
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Guoliang Xia
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
19
|
Vanorny DA, Mayo KE. The role of Notch signaling in the mammalian ovary. Reproduction 2017; 153:R187-R204. [PMID: 28283672 DOI: 10.1530/rep-16-0689] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/03/2017] [Accepted: 03/09/2017] [Indexed: 12/21/2022]
Abstract
The Notch pathway is a contact-dependent, or juxtacrine, signaling system that is conserved in metazoan organisms and is important in many developmental processes. Recent investigations have demonstrated that the Notch pathway is active in both the embryonic and postnatal ovary and plays important roles in events including follicle assembly and growth, meiotic maturation, ovarian vasculogenesis and steroid hormone production. In mice, disruption of the Notch pathway results in ovarian pathologies affecting meiotic spindle assembly, follicle histogenesis, granulosa cell proliferation and survival, corpora luteal function and ovarian neovascularization. These aberrations result in abnormal folliculogenesis and reduced fertility. The knowledge of the cellular interactions facilitated by the Notch pathway is an important area for continuing research, and future studies are expected to enhance our understanding of ovarian function and provide critical insights for improving reproductive health. This review focuses on the expression of Notch pathway components in the ovary, and on the multiple functions of Notch signaling in follicle assembly, maturation and development. We focus on the mouse, where genetic investigations are possible, and relate this information to the human ovary.
Collapse
Affiliation(s)
- Dallas A Vanorny
- Department of Molecular Biosciences and Center for Reproductive ScienceNorthwestern University, Evanston, Illinois, USA
| | - Kelly E Mayo
- Department of Molecular Biosciences and Center for Reproductive ScienceNorthwestern University, Evanston, Illinois, USA
| |
Collapse
|
20
|
York JP, Ren YA, Zeng J, Bin Zhang, Wang F, Chen R, Liu J, Xia X, Zhang P. Growth Arrest Specific 2 (GAS2) is a Critical Mediator of Germ Cell Cyst Breakdown and Folliculogenesis in Mice. Sci Rep 2016; 6:34956. [PMID: 27734842 PMCID: PMC5062118 DOI: 10.1038/srep34956] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/21/2016] [Indexed: 01/25/2023] Open
Abstract
In the mouse ovary, the primordial follicle pool is established through a diverse array of signaling pathways and tissue remodeling events. Growth arrest specific gene two (GAS2) is a highly conserved cytoskeleton-associated protein whose in vivo function remains unclear. In Drosophila, loss of the GAS2 homolog, Pigs, results in infertility. We demonstrate herein that, in the mouse ovary, GAS2 is expressed in the stromal cells surrounding the oocyte cysts on 16.5 dpc, and in stromal cells surrounding growing follicles during juvenile and adult life. We have generated genetically engineered mice with inactivated Gas2. Gas2 homozygous mutant mice are viable but have severely impaired fertility in females, in which oocyte cyst breakdown is disrupted and follicle growth is impaired, with significantly reduced numbers of large antral follicles and corpora lutea. In these mutant mice, the organization of the basal lamina surrounding developing follicles is severely defective at multiple stages of folliculogenesis. We also found that Notch signaling activity was altered in ovaries from Gas2 null mice around the time of birth and during follicular development later in life. These results indicate that GAS2 is a critical and novel regulator of tissue remodeling in the ovary during oocyte cyst breakdown and folliculogenesis.
Collapse
Affiliation(s)
- J Philippe York
- Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yi Athena Ren
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jie Zeng
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Bin Zhang
- Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA
| | - Fang Wang
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Rui Chen
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Jianqiao Liu
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Xuefeng Xia
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China.,Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA.,Biochemistry and Molecular Biology, Baylor College of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Pumin Zhang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Biochemistry and Molecular Biology, Baylor College of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|