1
|
Overland MR, Li Y, Derpinghaus A, Aksel S, Cao M, Ladwig N, Cunha GR, Himelreich-Perić M, Baskin LS. Development of the human ovary: Fetal through pubertal ovarian morphology, folliculogenesis and expression of cellular differentiation markers. Differentiation 2023; 129:37-59. [PMID: 36347737 DOI: 10.1016/j.diff.2022.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 01/28/2023]
Abstract
A definition of normal human fetal and early postnatal ovarian development is critical to the ability to accurately diagnose the presence or absence of functional ovarian tissue in clinical specimens. Through assembling an extensive histologic and immunohistochemical developmental ontogeny of human ovarian specimens from 8 weeks of gestation through 16 years of postnatal, we present a comprehensive immunohistochemical mapping of normal protein expression patterns in the early fetal through post-pubertal human ovary and detail a specific expression-based definition of the early stages of follicular development. Normal fetal and postnatal ovarian tissue is defined by the presence of follicular structures and characteristic immunohistochemical staining patterns, including granulosa cells expressing Forkhead Box Protein L2 (FOXL2). However, the current standard array of immunohistochemical markers poorly defines ovarian stromal tissue, and additional work is needed to identify new markers to advance our ability to accurately identify ovarian stromal components in gonadal specimens from patients with disorders of sexual differentiation.
Collapse
Affiliation(s)
- Maya R Overland
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Yi Li
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Sena Aksel
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Nicholas Ladwig
- Department of Pathology, University of California, 505 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| | - Marta Himelreich-Perić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
2
|
Cheng H, Shang D, Zhou R. Germline stem cells in human. Signal Transduct Target Ther 2022; 7:345. [PMID: 36184610 PMCID: PMC9527259 DOI: 10.1038/s41392-022-01197-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/06/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
The germline cells are essential for the propagation of human beings, thus essential for the survival of mankind. The germline stem cells, as a unique cell type, generate various states of germ stem cells and then differentiate into specialized cells, spermatozoa and ova, for producing offspring, while self-renew to generate more stem cells. Abnormal development of germline stem cells often causes severe diseases in humans, including infertility and cancer. Primordial germ cells (PGCs) first emerge during early embryonic development, migrate into the gentile ridge, and then join in the formation of gonads. In males, they differentiate into spermatogonial stem cells, which give rise to spermatozoa via meiosis from the onset of puberty, while in females, the female germline stem cells (FGSCs) retain stemness in the ovary and initiate meiosis to generate oocytes. Primordial germ cell-like cells (PGCLCs) can be induced in vitro from embryonic stem cells or induced pluripotent stem cells. In this review, we focus on current advances in these embryonic and adult germline stem cells, and the induced PGCLCs in humans, provide an overview of molecular mechanisms underlying the development and differentiation of the germline stem cells and outline their physiological functions, pathological implications, and clinical applications.
Collapse
Affiliation(s)
- Hanhua Cheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, 430072, Wuhan, China.
| | - Dantong Shang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, 430072, Wuhan, China
| | - Rongjia Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Renmin Hospital of Wuhan University, Wuhan University, 430072, Wuhan, China.
| |
Collapse
|
3
|
LIN28 Family in Testis: Control of Cell Renewal, Maturation, Fertility and Aging. Int J Mol Sci 2022; 23:ijms23137245. [PMID: 35806250 PMCID: PMC9266904 DOI: 10.3390/ijms23137245] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 12/12/2022] Open
Abstract
Male reproductive development starts early in the embryogenesis with somatic and germ cell differentiation in the testis. The LIN28 family of RNA-binding proteins promoting pluripotency has two members—LIN28A and LIN28B. Their function in the testis has been investigated but many questions about their exact role based on the expression patterns remain unclear. LIN28 expression is detected in the gonocytes and the migrating, mitotically active germ cells of the fetal testis. Postnatal expression of LIN28 A and B showed differential expression, with LIN28A expressed in the undifferentiated spermatogonia and LIN28B in the elongating spermatids and Leydig cells. LIN28 interferes with many signaling pathways, leading to cell proliferation, and it is involved in important testicular physiological processes, such as cell renewal, maturation, fertility, and aging. In addition, aberrant LIN28 expression is associated with testicular cancer and testicular disorders, such as hypogonadotropic hypogonadism and Klinefelter’s syndrome. This comprehensive review encompasses current knowledge of the function of LIN28 paralogs in testis and other tissues and cells because many studies suggest LIN28AB as a promising target for developing novel therapeutic agents.
Collapse
|
4
|
Lundgaard Riis M, Jørgensen A. Deciphering Sex-Specific Differentiation of Human Fetal Gonads: Insight From Experimental Models. Front Cell Dev Biol 2022; 10:902082. [PMID: 35721511 PMCID: PMC9201387 DOI: 10.3389/fcell.2022.902082] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Sex-specific gonadal differentiation is initiated by the expression of SRY in male foetuses. This promotes a signalling pathway directing testicular development, while in female foetuses the absence of SRY and expression of pro-ovarian factors promote ovarian development. Importantly, in addition to the initiation of a sex-specific signalling cascade the opposite pathway is simultaneously inhibited. The somatic cell populations within the gonads dictates this differentiation as well as the development of secondary sex characteristics via secretion of endocrine factors and steroid hormones. Opposing pathways SOX9/FGF9 (testis) and WNT4/RSPO1 (ovary) controls the development and differentiation of the bipotential mouse gonad and even though sex-specific gonadal differentiation is largely considered to be conserved between mice and humans, recent studies have identified several differences. Hence, the signalling pathways promoting early mouse gonad differentiation cannot be directly transferred to human development thus highlighting the importance of also examining this signalling in human fetal gonads. This review focus on the current understanding of regulatory mechanisms governing human gonadal sex differentiation by combining knowledge of these processes from studies in mice, information from patients with differences of sex development and insight from manipulation of selected signalling pathways in ex vivo culture models of human fetal gonads.
Collapse
Affiliation(s)
- Malene Lundgaard Riis
- Department of Growth and Reproduction, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| | - Anne Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
- International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital—Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
5
|
Chen J, Liu W, Lee KF, Liu K, Wong BPC, Shu-Biu Yeung W. Overexpression of Lin28a induces a primary ovarian insufficiency phenotype via facilitation of primordial follicle activation in mice. Mol Cell Endocrinol 2022; 539:111460. [PMID: 34543700 DOI: 10.1016/j.mce.2021.111460] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 01/28/2023]
Abstract
Lin28a is an RNA binding protein and increasing evidence has indicated its role in regulating female fertility. Lin28a has been reported to be involved in ovarian follicle activation. However, its role and mechanisms in regulating primordial follicle activation have not yet been explored. To test whether overexpression of Lin28a activates ovarian primordial follicles, studies were conducted in wild type (WT) and Lin28a Tg mice. Female Lin28a Tg mice at 4-month old exhibited significantly smaller litter size and fewer ovulated oocytes when compared with the WT mice. By 6-month of age, these parameters in Lin28a Tg mice were less than 20% of the WT mice. At postnatal day (PD) 14, the number of primordial follicles was significantly decreased but the number of primary follicles was significantly increased in the transgenic mice. The number of primordial follicles, secondary and antral follicles in these mice were drastically reduced at PD21. In the ovary of Lin28a Tg mice, there were activation of Wnt/β-catenin signaling and its downstream mTOR pathway. Interestingly, overexpression of Lin28a, which can also act as transcriptional activator, activated Wnt signaling through enhancing the transcription of Wnt co-receptor LRP5. In conclusion, overexpression of Lin28a induced a primary ovarian insufficiency phenotype in long term via facilitating Wnt/β-catenin signaling leading to activation of primordial follicles.
Collapse
Affiliation(s)
- Jing Chen
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China.
| | - Weimin Liu
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China.
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China.
| | - Kui Liu
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China.
| | - Benancy P C Wong
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China.
| | - William Shu-Biu Yeung
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China.
| |
Collapse
|
6
|
Lundgaard Riis M, Nielsen JE, Hagen CP, Rajpert-De Meyts E, Græm N, Jørgensen A, Juul A. Accelerated loss of oogonia and impaired folliculogenesis in females with Turner syndrome start during early fetal development. Hum Reprod 2021; 36:2992-3002. [PMID: 34568940 DOI: 10.1093/humrep/deab210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
STUDY QUESTION How are germ cell numbers and initiation of folliculogenesis affected in fetal Turner syndrome (TS) ovaries? SUMMARY ANSWER Germ cell development was severely affected already in early second trimester pregnancies, including accelerated oogonia loss and impaired initiation of primordial follicle formation in TS ovaries, while the phenotype in TS mosaic ovaries was less severe. WHAT IS KNOWN ALREADY Females with TS are characterized by premature ovarian insufficiency (POI). This phenotype is proposed to be a consequence of germ cell loss during development, but the timing and mechanisms behind this are not characterized in detail. Only few studies have evaluated germ cell development in fetal TS and TS mosaic ovaries, and with a sparse number of specimens included per study. STUDY DESIGN, SIZE, DURATION This study included a total of 102 formalin-fixed and paraffin-embedded fetal ovarian tissue specimens. Specimens included were from fetuses with 45,X (N = 42 aged gestational week (GW) 12-20, except one GW 40 sample), 45,X/46,XX (N = 7, aged GW 12-20), and from controls (N = 53, aged GW 12-42) from a biobank (ethics approval # H-2-2014-103). PARTICIPANTS/MATERIALS, SETTING, METHODS The number of OCT4 positive germ cells/mm2, follicles (primordial and primary)/mm2 and cPARP positive cells/mm2 were quantified in fetal ovarian tissue from TS, TS mosaic and controls following morphological and immunohistochemical analysis. MAIN RESULTS AND THE ROLE OF CHANCE After adjusting for gestational age, the number of OCT4+ oogonia was significantly higher in control ovaries (N = 53) versus 45,X ovaries (N = 40, P < 0.001), as well as in control ovaries versus 45,X/46,XX mosaic ovaries (N = 7, P < 0.043). Accordingly, the numbers of follicles were significantly higher in control ovaries versus 45,X and 45,X/46,XX ovaries from GW 16-20 with a median range of 154 (N = 11) versus 0 (N = 24) versus 3 (N = 5) (P < 0.001 and P < 0.015, respectively). The number of follicles was also significantly higher in 45,X/46,XX mosaic ovaries from GW 16-20 compared with 45,X ovaries (P < 0.005). Additionally, the numbers of apoptotic cells determined as cPARP+ cells/mm2 were significantly higher in ovaries 45,X (n = 39) versus controls (n = 15, P = 0.001) from GW 12-20 after adjusting for GW. LIMITATIONS, REASONS FOR CAUTION The analysis of OCT4+ cells/mm2, cPARP+ cells/mm2 and follicles (primordial and primary)/mm2 should be considered semi-quantitative as it was not possible to use quantification by stereology. The heterogeneous distribution of follicles in the ovarian cortex warrants a cautious interpretation of the exact quantitative numbers reported. Moreover, only one 45,X specimen and no 45,X/46,XX specimens aged above GW 20 were available for this study, which unfortunately made it impossible to assess whether the ovarian folliculogenesis was delayed or absent in the TS and TS mosaic specimens. WIDER IMPLICATIONS OF THE FINDINGS This human study provides insights about the timing of accelerated fetal germ cell loss in TS. Knowledge about the biological mechanism of POI in girls with TS is clinically useful when counseling patients about expected ovarian function and fertility preservation strategies. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC). TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Malene Lundgaard Riis
- Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - John E Nielsen
- Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Casper P Hagen
- Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Niels Græm
- Department of Pathology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Anne Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.,International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
7
|
Protein expression reveals a molecular sexual identity of avian primordial germ cells at pre-gonadal stages. Sci Rep 2021; 11:19236. [PMID: 34584135 PMCID: PMC8478952 DOI: 10.1038/s41598-021-98454-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 09/06/2021] [Indexed: 11/11/2022] Open
Abstract
In poultry, in vitro propagated primordial germ cells (PGCs) represent an important tool for the cryopreservation of avian genetic resources. However, several studies have highlighted sexual differences exhibited by PGCs during in vitro propagation, which may compromise their reproductive capacities. To understand this phenomenon, we compared the proteome of pregonadal migratory male (ZZ) and female (ZW) chicken PGCs propagated in vitro by quantitative proteomic analysis using a GeLC-MS/MS strategy. Many proteins were found to be differentially abundant in chicken male and female PGCs indicating their early sexual identity. Many of the proteins more highly expressed in male PGCs were encoded by genes localised to the Z sex chromosome. This suggests that the known lack of dosage compensation of the transcription of Z-linked genes between sexes persists at the protein level in PGCs, and that this may be a key factor of their autonomous sex differentiation. We also found that globally, protein differences do not closely correlate with transcript differences indicating a selective translational mechanism in PGCs. Male and female PGC expressed protein sets were associated with differential biological processes and contained proteins known to be biologically relevant for male and female germ cell development, respectively. We also discovered that female PGCs have a higher capacity to uptake proteins from the cell culture medium than male PGCs. This study presents the first evidence of an early predetermined sex specific cell fate of chicken PGCs and their sexual molecular specificities which will enable the development of more precise sex-specific in vitro culture conditions for the preservation of avian genetic resources.
Collapse
|
8
|
Tocci A. The safety of VASA pos presumptive adult ovarian stem cells. Reprod Biomed Online 2021; 43:587-597. [PMID: 34474974 DOI: 10.1016/j.rbmo.2021.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 01/16/2023]
Abstract
Isolation and characterization of presumptive human adult ovarian stem cells (OSC) has broken the long standing dogma of the absence of postnatal neo-oogenesis. Human adult OSC have been immunosorted by antibodies reacting against the RNA helicase VASA and have been reported to engraft into appropriate stem cell niches to promote neo-oogenesis. Analysis of published research, however, questions some of the findings on isolation, characterization, in-vitro self-renewal and clinical safety of the presumptive human adult OSC. In the present study, human VASApos embryo-fetal primordial germ cells and presumptive adult OSC are shown to share several pluripotency and early germ cell markers not ascertained in the initial characterization of adult OSC. A new hypothesis is made that the restoration of fertility claimed to result from presumptive human adult OSC may be attributed instead to VASApos embryo-fetal primordial germ cell remnants in the adult ovary, or alternatively to earlier VASAneg germ cells generated by in-vitro de-differentiation of the presumptive OSC. The suggested hypotheses have extensive implications for the practice and safety of adult OSC in the development of new treatments aimed at rescuing the ovarian reserve.
Collapse
Affiliation(s)
- Angelo Tocci
- Gruppo Donnamed, Reproductive Medicine Unit Via Cassia 1110 00189, Rome, Italy.
| |
Collapse
|
9
|
Scarlet D, Handschuh S, Reichart U, Podico G, Ellerbrock RE, Demyda-Peyrás S, Canisso IF, Walter I, Aurich C. Sexual Differentiation and Primordial Germ Cell Distribution in the Early Horse Fetus. Animals (Basel) 2021; 11:2422. [PMID: 34438878 PMCID: PMC8388682 DOI: 10.3390/ani11082422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/29/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022] Open
Abstract
It was the aim of this study to characterize the development of the gonads and genital ducts in the equine fetus around the time of sexual differentiation. This included the identification and localization of the primordial germ cell population. Equine fetuses between 45 and 60 days of gestation were evaluated using a combination of micro-computed tomography scanning, immunohistochemistry, and multiplex immunofluorescence. Fetal gonads increased in size 23-fold from 45 to 60 days of gestation, and an even greater increase was observed in the metanephros volume. Signs of mesonephros atrophy were detected during this time. Tubular structures of the fetal testes were present from day 50 onwards, whereas cell clusters dominated in the fetal ovary. The genital ducts were well-differentiated and presented a lumen in all samples. No sign of mesonephric or paramesonephric duct degeneration was detected. Expression of AMH was strong in the fetal testes but absent in ovaries. Irrespective of sex, primordial germ cells selectively expressed LIN28. Migration of primordial germ cells from the mesonephros to the gonad was detected at 45 days, but not at 60 days of development. Their number and distribution within the gonad were influenced (p < 0.05) by fetal sex. Most primordial germ cells (86.8 ± 3.2% in females and 84.6 ± 4.7% in males) were characterized as pluripotent according to co-localization with CD117. However, only a very small percentage of primordial germ cells were proliferating (7.5 ± 1.7% in females and 3.2 ± 1.2% in males) based on co-localization with Ki67. It can be concluded that gonadal sexual differentiation in the horse occurs asynchronously with regard to sex but already before 45 days of gestation.
Collapse
Affiliation(s)
- Dragos Scarlet
- Obstetrics, Gynecology and Andrology, Department for Small Animals and Horses, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
- Institute of Veterinary Anatomy and Clinic of Reproductive Medicine, Vetsuisse Faculty Zürich, Winterthurerstrasse 260, 8057 Zürich, Switzerland
| | - Stephan Handschuh
- Vetcore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (S.H.); (U.R.); (I.W.)
| | - Ursula Reichart
- Vetcore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (S.H.); (U.R.); (I.W.)
| | - Giorgia Podico
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA; (G.P.); (R.E.E.); (I.F.C.)
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Robyn E. Ellerbrock
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA; (G.P.); (R.E.E.); (I.F.C.)
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Sebastián Demyda-Peyrás
- Department of Animal Production, School of Veterinary Sciences, National University of La Plata and CONICET CCT-La Plata, Calle 60 and 118 S/N, 1900 La Plata, Argentina;
| | - Igor F. Canisso
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA; (G.P.); (R.E.E.); (I.F.C.)
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Ingrid Walter
- Vetcore Facility for Research, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (S.H.); (U.R.); (I.W.)
- Institute of Pathology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Christine Aurich
- Center for Artificial Insemination and Embryo Transfer, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria;
| |
Collapse
|
10
|
Dong LH, Hildorf S, Clasen-Linde E, Kvist K, Cortes D, Thorup J, Andersen CY. Postnatal germ cell development in cryptorchid boys. Asian J Androl 2021; 22:258-264. [PMID: 31274480 PMCID: PMC7275797 DOI: 10.4103/aja.aja_48_19] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cryptorchidism is associated with infertility in adulthood. Early orchiopexy is suggested to reduce the risk. Information is lacking on the potential link between infant germ cell maturation and the risk of future infertility. The objective of the study was to evaluate age-related germ cell development in cryptorchidism. Immunostaining for markers of germ cell development (octamer-binding transcription factor 3/4 [OCT3/4], placental alkaline phosphatase [PLAP], KIT proto-oncogene [C-KIT], podoplanin [D2-40], Lin-28 homolog A [LIN28], and G antigen 7 [GAGE-7]) was performed in testicular biopsies from 40 cryptorchid boys aged 4-35 months. Germ cell numbers and distributions were evaluated in cross sections of seminiferous tubules, with and without immunostaining. OCT3/4, D2-40, and LIN28 were generally expressed in the early stages of germ cell development, as shown by positive expression in germ cells in the central region of seminiferous tubules. In contrast, PLAP and GAGE-7 were expressed in both central and peripheral parts of the tubules in the early stages of development and expressed mainly in a peripheral position with advancing age. Germ cell maturation was delayed in this study population as compared with that observed in our previous study on germ cell markers in a healthy population. The number of GAGE-7-positive germ cells per tubular cross section obtained by immunostaining was significantly higher than that obtained by standard hematoxylin and eosin staining. Double immunostaining revealed heterogeneity in germ cell development in cryptorchid testes. These results shed light on the pathophysiology of germ cell development in boys with cryptorchidism.
Collapse
Affiliation(s)
- Li-Hua Dong
- Laboratory of Reproductive Biology, Copenhagen University Hospital Rigshospitalet, Copenhagen 2100, Denmark
| | - Simone Hildorf
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Erik Clasen-Linde
- Department of Pathology, Copenhagen University Hospital Rigshospitalet, Copenhagen 2100, Denmark
| | - Kolja Kvist
- Department of Pediatric Surgery, Copenhagen University Hospital Rigshospitalet, Copenhagen 2100, Denmark
| | - Dina Cortes
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.,Department of Pediatrics, Copenhagen University Hospital Hvidovre, Copenhagen 2650, Denmark
| | - Jørgen Thorup
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.,Department of Pediatric Surgery, Copenhagen University Hospital Rigshospitalet, Copenhagen 2100, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Copenhagen University Hospital Rigshospitalet, Copenhagen 2100, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
11
|
Zuo Q, Jin K, Wang M, Zhang Y, Chen G, Li B. BMP4 activates the Wnt- Lin28A- Blimp1-Wnt pathway to promote primordial germ cell formation via altering H3K4me2. J Cell Sci 2021; 134:jcs249375. [PMID: 33443086 PMCID: PMC7875490 DOI: 10.1242/jcs.249375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022] Open
Abstract
The unique developmental characteristics of chicken primordial germ cells (PGCs) enable them to be used in recovery of endangered bird species, gene editing and the generation of transgenic birds, but the limited number of PGCs greatly limits their application. Studies have shown that the formation of mammalian PGCs is induced by BMP4 signal, but the mechanism underlying chicken PGC formation has not been determined. Here, we confirmed that Wnt signaling activated via BMP4 activates transcription of Lin28A by inducing β-catenin to compete with LSD1 for binding to TCF7L2, causing LSD1 to dissociate from the Lin28A promoter and enhancing H3K4me2 methylation in this region. Lin28A promotes PGC formation by inhibiting gga-let7a-3p maturation to initiate Blimp1 expression. Interestingly, expression of Blimp1 helped sustain Wnt5A expression by preventing LSD1 binding to the Wnt5A promoter. We thus elucidated a positive feedback pathway involving Wnt-Lin28A-Blimp1-Wnt that ensures PGC formation. In summary, our data provide new insight into the development of PGCs in chickens.
Collapse
Affiliation(s)
- Qisheng Zuo
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Kai Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Man Wang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yani Zhang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Guohong Chen
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Bichun Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu 225009, China
| |
Collapse
|
12
|
Meinsohn MC, Hughes CHK, Estienne A, Saatcioglu HD, Pépin D, Duggavathi R, Murphy BD. A role for orphan nuclear receptor liver receptor homolog-1 (LRH-1, NR5A2) in primordial follicle activation. Sci Rep 2021; 11:1079. [PMID: 33441767 PMCID: PMC7807074 DOI: 10.1038/s41598-020-80178-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023] Open
Abstract
Liver receptor homolog-1 (NR5A2) is expressed specifically in granulosa cells of developing ovarian follicles where it regulates the late stages of follicle development and ovulation. To establish its effects earlier in the trajectory of follicular development, NR5A2 was depleted from granulosa cells of murine primordial and primary follicles. Follicle populations were enumerated in neonates at postnatal day 4 (PND4) coinciding with the end of the formation of the primordial follicle pool. The frequency of primordial follicles in PND4 conditional knockout (cKO) ovaries was greater and primary follicles were substantially fewer relative to control (CON) counterparts. Ten-day in vitro culture of PND4 ovaries recapitulated in vivo findings and indicated that CON mice developed primary follicles in the ovarian medulla to a greater extent than did cKO animals. Two subsets of primordial follicles were observed in wildtype ovaries: one that expressed NR5A2 and the second in which the transcript was absent. Neither expressed the mitotic marker. KI-67, indicating their developmental quiescence. RNA sequencing on PND4 demonstrated that loss of NR5A2 induced changes in 432 transcripts, including quiescence markers, inhibitors of follicle activation, and regulators of cellular migration and epithelial-to-mesenchymal transition. These experiments suggest that NR5A2 expression poises primordial follicles for entry into the developing pool.
Collapse
Affiliation(s)
- Marie-Charlotte Meinsohn
- Centre de recherche en reproduction et fertilité, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 7C6, Canada
| | - Camilla H K Hughes
- Centre de recherche en reproduction et fertilité, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 7C6, Canada
| | - Anthony Estienne
- Centre de recherche en reproduction et fertilité, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 7C6, Canada
| | - Hatice D Saatcioglu
- Pediatric Surgical Research Laboratories, Simches Research Center, Massachusetts General Hospital, 185 Cambridge St., Boston, MA, 02114, USA
| | - David Pépin
- Pediatric Surgical Research Laboratories, Simches Research Center, Massachusetts General Hospital, 185 Cambridge St., Boston, MA, 02114, USA
| | - Raj Duggavathi
- Department of Animal Science, McGill University, 21111 Lakeshore Rd., MS1085, Ste-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Bruce D Murphy
- Centre de recherche en reproduction et fertilité, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 7C6, Canada.
| |
Collapse
|
13
|
Study on the Function and Mechanism of Lin28B in the Formation of Chicken Primordial Germ Cells. Animals (Basel) 2020; 11:ani11010043. [PMID: 33379329 PMCID: PMC7823903 DOI: 10.3390/ani11010043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/20/2020] [Accepted: 12/24/2020] [Indexed: 12/24/2022] Open
Abstract
Lin28A and Lin28B are two homologues of the same family of RNA binding proteins (RBPs). The function and molecular mechanism of Lin28A in the formation of primordial germ cells (PGCs) are very clear, but the related research on Lin28B is rarely reported. Here, we found that the overexpression of Lin28B can promote the formation of PGC in vivo. Furthermore, the overexpression of Lin28B also resulted in the inhibition of totipotency gene expression and upregulated the PGCs marker genes, and a significant increase in the number of PGCs in genital ridge, as detected by Periodic Acid-Schiff(PAS) staining. However, the inhibited Lin28B expression showed completely opposite results, which were confirmed on the PGC induction model in vitro. Mechanistically, we found that the overexpression of Lin28B can inhibit the maturation of let-7a-3p, and the results of high-throughput sequencing indicated that let-7a-3p was a negative regulator of the formation process of PGCs. Therefore, we conclude that our results determine that Lin28B participates in the formation of PGCs through let-7a-3p, which set a theoretical foundation for improving the function and mechanism of Lin28 family in the formation of PGCs.
Collapse
|
14
|
Jin K, Li D, Jin J, Song J, Zhang Y, Chang G, Chen G, Li B. C1EIP Functions as an Activator of ENO1 to Promote Chicken PGCs Formation via Inhibition of the Notch Signaling Pathway. Front Genet 2020; 11:751. [PMID: 32849782 PMCID: PMC7396672 DOI: 10.3389/fgene.2020.00751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
The production of germ cells, especially primordial germ cells (PGCs), is important for avian stem cells and reproduction biology. However, key factors involved in the regulation of PGCs remain unknown. Here, we report a PGC-related marker gene: C1EIP (Chromosome 1 Expression in PGCs), whose activation and expression are regulated by the transcription factor STAT3 (signal transducer and activator of transcription 3), histone acetylation, and promoter methylation. C1EIP regulates PGCs formation by mediating the expression of PGC-associated genes, such as CVH (Chicken Vasa Homologous) and CKIT (Chicken KIT proto-oncogene). C1EIP knockdown during embryonic development reduces PGC generation efficiency both in vitro and in ovo. Conversely, C1EIP overexpression increases the formation efficiency of PGCs. C1EIP encodes a cytoplasmic protein that interacts with ENO1 (Enolase 1) in the cytoplasm, inhibits the Notch signaling pathway, and positively regulates PGC generation. Collectively, our findings demonstrate C1EIP as a novel gene involved in PGC formation, which regulates genes involved in embryonic stem cell differentiation through interaction with ENO1 and subsequent inhibition of the Notch signaling pathway by the impression of Myc (MYC proto-oncogene).
Collapse
Affiliation(s)
- Kai Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Dong Li
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, China
| | - Jing Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jiuzhou Song
- Animal & Avian Sciences, University of Maryland, College Park, College Park, MD, United States
| | - Yani Zhang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Guobing Chang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Guohong Chen
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Bichun Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
15
|
Regulation of human trophoblast surrogate Jeg-3 spheroids implantation potential by Wnt/β-catenin pathway and lin28a/let-7a axis. Exp Cell Res 2020; 388:111718. [DOI: 10.1016/j.yexcr.2019.111718] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/04/2019] [Accepted: 11/08/2019] [Indexed: 12/16/2022]
|
16
|
Zuo Q, Jin K, Song J, Zhang Y, Chen G, Li B. Interaction of the primordial germ cell-specific protein C2EIP with PTCH2 directs differentiation of embryonic stem cells via HH signaling activation. Cell Death Dis 2018; 9:497. [PMID: 29703892 PMCID: PMC5923244 DOI: 10.1038/s41419-018-0557-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/26/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022]
Abstract
Although many marker genes for germ cell differentiation have been identified, genes that specifically regulate primordial germ cell (PGC) generation are more difficult to determine. In the current study, we confirmed that C2EIP is a PGC marker gene that regulates differentiation by influencing the expression of pluripotency-associated genes such as Oct4 and Sox2. Knockout of C2EIP during embryonic development reduced PGC generation efficiency 1.5-fold, whereas C2EIP overexpression nearly doubled the generation efficiency both in vitro and in vivo. C2EIP encodes a cytoplasmic protein that interacted with PTCH2 at the intracellular membrane, promoted PTCH2 ubiquitination, activated the Hedgehog (HH) signaling pathway via competitive inhibition of the GPCR-like protein SMO, and positively regulated PGC generation. Activation and expression of C2EIP are regulated by the transcription factor STAT1, histone acetylation, and promoter methylation. Our data suggest that C2EIP is a novel, specific indicator of PGC generation whose gene product regulates embryonic stem cell differentiation by activating the HH signaling pathway via PTCH2 modification.
Collapse
Affiliation(s)
- Qisheng Zuo
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, 225009, P.R. China
| | - Kai Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, 225009, P.R. China
| | - Jiuzhou Song
- Animal & Avian Sciences, University of Maryland, Baltimore, MD, 20741, USA
| | - Yani Zhang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, 225009, P.R. China
| | - Guohong Chen
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, 225009, P.R. China
| | - Bichun Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, 225009, P.R. China.
| |
Collapse
|
17
|
Frydman N, Poulain M, Arkoun B, Duquenne C, Tourpin S, Messiaen S, Habert R, Rouiller-Fabre V, Benachi A, Livera G. Human foetal ovary shares meiotic preventing factors with the developing testis. Hum Reprod 2018; 32:631-642. [PMID: 28073973 DOI: 10.1093/humrep/dew343] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 12/14/2016] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION How can pre-meiotic germ cells persist in the human foetal ovary? SUMMARY ANSWER Numerous oogonia escaping meiotic entry were retrieved throughout human ovarian development simultaneously with the expression of signalling pathways preventing meiosis, typically described in the rodent embryonic testis. WHAT IS KNOWN ALREADY The transition from mitosis to meiosis is a key event in female germ cells that remains poorly documented in research on the human ovary. Previous reports described a strikingly asynchronous differentiation in the human female germ line during development, with the persistence of oogonia among oocytes and follicles during the second and third trimesters. The possible mechanisms allowing some cells to escape meiosis remain elusive. STUDY DESIGN SIZE, DURATION In order to document the extent of this phenomenon, we detailed the expression profile of germ cell differentiation markers using 73 ovaries ranging from 6.4 to 35 weeks post-fertilization. PARTICIPANTS/MATERIALS SETTING, METHODS Pre-meiotic markers were detected by immunohistochemistry or qRT-PCR. The expression of the main meiosis-preventing factors identified in mice was analysed, and their functionality assessed using organ cultures. MAIN RESULTS AND THE ROLE OF CHANCE Oogonia stained for AP2γ could be traced from the first trimester until the end of the third trimester. Female germ cell differentiation is organized both in time and space in a centripetal manner in the foetal human ovary. Unexpectedly, some features usually ascribed to rodent pre-spermatogonia could be observed in human foetal ovaries, such as NANOS2 expression and quiescence in some germ cells. The two main somatic signals known to inhibit meiosis in the mouse embryonic testis, CYP26B1 and FGF9, were detected in the human ovary and act simultaneously to repress STRA8 and meiosis in human foetal female germ cells. LARGE SCALE DATA N/A. LIMITATIONS REASON FOR CAUTION Our conclusions relied partly on in vitro experiments. Germ cells were not systematically identified with immunostaining and some may have thus escaped analysis. WIDER IMPLICATIONS OF THE FINDINGS We found evidence that a robust repression of meiotic entry is taking place in the human foetal ovary, possibly explaining the exceptional long-lasting presence of pre-meiotic germ cells until late gestational age. This result calls for a redefinition of the markers known as classical male markers, which may in fact characterize mammalian developing gonads irrespectively of their sex. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by the Université Paris Diderot-Paris 7 and Université Paris-Sud, CEA, INSERM, and Agence de la Biomédecine. The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Nelly Frydman
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. Paris-Sud, Université Paris-Saclay, Fontenay aux Roses F-92265, France.,AP-HP, Reproductive Biology Unit, Univ. Paris-Sud, Université Paris-Saclay, Hôpital Antoine Béclère, Clamart F-92140, France
| | - Marine Poulain
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. Paris-Sud, Université Paris-Saclay, Fontenay aux RosesF-92265, France
| | - Brahim Arkoun
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. Paris-Sud, Université Paris-Saclay, Fontenay aux RosesF-92265, France
| | - Clotilde Duquenne
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. Paris-Sud, Université Paris-Saclay, Fontenay aux RosesF-92265, France
| | - Sophie Tourpin
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. Paris-Sud, Université Paris-Saclay, Fontenay aux RosesF-92265, France
| | - Sébastien Messiaen
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. Paris-Sud, Université Paris-Saclay, Fontenay aux RosesF-92265, France
| | - René Habert
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. Paris-Sud, Université Paris-Saclay, Fontenay aux RosesF-92265, France
| | - Virginie Rouiller-Fabre
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. Paris-Sud, Université Paris-Saclay, Fontenay aux RosesF-92265, France
| | - Alexandra Benachi
- AP-HP, Department of Obstetrics and Gynaecology, Univ. Paris-Sud, Université Paris-Saclay, Hôpital Antoine Béclère, ClamartF-92140, France
| | - Gabriel Livera
- Laboratory of Development of the Gonads, Unit of Genetic Stability, Stem Cells and Radiation, UMR 967, INSERM, CEA/DSV/iRCM/SCSR, Univ. Paris Diderot, Sorbonne Paris Cité, Univ. Paris-Sud, Université Paris-Saclay, Fontenay aux RosesF-92265, France
| |
Collapse
|
18
|
Ibuprofen is deleterious for the development of first trimester human fetal ovary ex vivo. Hum Reprod 2018; 33:482-493. [DOI: 10.1093/humrep/dex383] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/01/2018] [Indexed: 12/17/2022] Open
|
19
|
Hayashi M, Kawaguchi T, Durcova-Hills G, Imai H. Generation of germ cells from pluripotent stem cells in mammals. Reprod Med Biol 2017; 17:107-114. [PMID: 29692667 PMCID: PMC5902460 DOI: 10.1002/rmb2.12077] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/07/2017] [Indexed: 01/01/2023] Open
Abstract
Background The germ cell lineage transmits genetic and epigenetic information to the next generation. Primordial germ cells (PGCs), the early embryonic precursors of sperm or eggs, have been studied extensively. Recently, in vitro models of PGC induction have been established in the mouse. Many attempts are reported to enhance our understanding of PGC development in other mammals, including human. Methods Here, original and review articles that have been published on PubMed are reviewed in order to give an overview of the literature that is focused on PGC development, including the specification of in vivo and in vitro in mice, human, porcine, and bovine. Results Mammalian PGC development, in vivo and in vitro, have been studied primarily by using the mouse model as a template to study PGC specification in other mammals, including human, porcine, and bovine. Conclusion The growing body of published works reveals similarities, as well as differences, in PGC establishment in and between mouse and human.
Collapse
Affiliation(s)
- Masafumi Hayashi
- Laboratory of Reproductive Biology Graduate School of Agriculture Kyoto University Kyoto Japan
| | - Takamasa Kawaguchi
- Laboratory of Reproductive Biology Graduate School of Agriculture Kyoto University Kyoto Japan.,The Fukui Research Institute Ono Pharmaceutical Companyy, Ltd. Fukui Japan
| | - Gabriela Durcova-Hills
- Laboratory of Reproductive Biology Graduate School of Agriculture Kyoto University Kyoto Japan
| | - Hiroshi Imai
- Laboratory of Reproductive Biology Graduate School of Agriculture Kyoto University Kyoto Japan
| |
Collapse
|
20
|
Wang Y, Zuo Q, Bi Y, Zhang W, Jin J, Zhang L, Zhang Y, Li B. miR‐31 Regulates Spermatogonial Stem Cells Meiosis via Targeting Stra8. J Cell Biochem 2017; 118:4844-4853. [DOI: 10.1002/jcb.26159] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
ABSTRACTStra8 (stimulated by retinoic acid gene 8) is a specific gene that is expressed in mammalian germ cells during transition from mitosis to meiosis and plays a key role in the initiation of meiosis in mammals and birds. So, the evaluation of the Stra8 pathway in cSSCs may provide a deeper insight into mammalian spermatogenesis. miRNA was also an important regulating factor for meiosis of SSCs. However, there is currently no data indicating that miRNA regulate the meiosis of SSCs via Stra8. Here, we predicted the prospective miRNA targeting to Stra8 using the online Bioinformatics database‐Targetscan, and performed an analysis of the dual‐luciferase recombinant vector, pGL3‐CMV‐LUC‐MCS‐Stra8‐3′UTR. miR‐31 mimics (miR‐31m), miR‐31 inhibitors (miR‐31i), Control (NC, scrambled oligonucleotides transfection) were transfected into cSSCs; Stra8 and miRNA were analyzed by RT‐qPCR, immunofluorescence, and Western blot. The detection of haploid was conducted by flow cytometry. The results showed that miR‐31 regulates meiosis of cSSCs via targeting Stra8 in vitro and in vivo. Our study identifies a new regulatory pathway that miR‐31 targets Stra8 and inhibits spermatogenesis. J. Cell. Biochem. 118: 4844–4853, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yingjie Wang
- College of Animal Science and Technology Yangzhou University Yangzhou 225009, Jiangsu Province P.R. China
- Key Laboratory for Animal Genetics, Breeding, Reproduction, and Molecular Design of Jiangsu Province Yangzhou, 225009, Jiangsu Province P.R. China
| | - Qisheng Zuo
- College of Animal Science and Technology Yangzhou University Yangzhou 225009, Jiangsu Province P.R. China
- Key Laboratory for Animal Genetics, Breeding, Reproduction, and Molecular Design of Jiangsu Province Yangzhou, 225009, Jiangsu Province P.R. China
| | - Yulin Bi
- College of Animal Science and Technology Yangzhou University Yangzhou 225009, Jiangsu Province P.R. China
- Key Laboratory for Animal Genetics, Breeding, Reproduction, and Molecular Design of Jiangsu Province Yangzhou, 225009, Jiangsu Province P.R. China
| | - Wenhui Zhang
- College of Animal Science and Technology Yangzhou University Yangzhou 225009, Jiangsu Province P.R. China
- Key Laboratory for Animal Genetics, Breeding, Reproduction, and Molecular Design of Jiangsu Province Yangzhou, 225009, Jiangsu Province P.R. China
| | - Jing Jin
- College of Animal Science and Technology Yangzhou University Yangzhou 225009, Jiangsu Province P.R. China
- Key Laboratory for Animal Genetics, Breeding, Reproduction, and Molecular Design of Jiangsu Province Yangzhou, 225009, Jiangsu Province P.R. China
| | - Liangliang Zhang
- College of Animal Science and Technology Yangzhou University Yangzhou 225009, Jiangsu Province P.R. China
- Key Laboratory for Animal Genetics, Breeding, Reproduction, and Molecular Design of Jiangsu Province Yangzhou, 225009, Jiangsu Province P.R. China
| | - Ya‐ni Zhang
- College of Animal Science and Technology Yangzhou University Yangzhou 225009, Jiangsu Province P.R. China
- Key Laboratory for Animal Genetics, Breeding, Reproduction, and Molecular Design of Jiangsu Province Yangzhou, 225009, Jiangsu Province P.R. China
| | - Bichun Li
- College of Animal Science and Technology Yangzhou University Yangzhou 225009, Jiangsu Province P.R. China
- Key Laboratory for Animal Genetics, Breeding, Reproduction, and Molecular Design of Jiangsu Province Yangzhou, 225009, Jiangsu Province P.R. China
| |
Collapse
|
21
|
Wang Y, Li J, Guo S, Ouyang Y, Yin L, Liu S, Zhao Z, Yang J, Huang W, Qin H, Zhao X, Ni B, Wang H. Lin28B facilitates the progression and metastasis of pancreatic ductal adenocarcinoma. Oncotarget 2017; 8:60414-60428. [PMID: 28947981 PMCID: PMC5601149 DOI: 10.18632/oncotarget.19578] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 06/25/2017] [Indexed: 12/19/2022] Open
Abstract
Lin28B, a Lin28 homologue, represses the biogenesis of let-7 microRNAs (miRNAs), has a role in tumorigenesis, and is considered a potential therapeutic target for various human malignancies. However, the associations between Lin28B and the clinical features and outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) remain unclear. In this study, we explored the clinical significance of Lin28B in PDAC and its association with metastasis by examining tissues from patients with PDAC and elucidated the molecular mechanisms using PDAC cell lines. In patients, high Lin28B expression was significantly correlated with high levels of lymphatic metastasis, distant metastasis and a poor prognosis. Furthermore, the multivariate analysis identified Lin28B expression as an independent prognostic factor in patients. In cell lines, stable silencing of Lin28B inhibited cell proliferation, cell cycle transition, migration and the epithelial-mesenchymal transition (EMT). It also increased the expression of the c-MYC, HMGA2 and KRAS genes, which are targeted by the cancer-suppressor miRNA let-7. Lin28B overexpression in the PDAC cell lines had the opposite effect. In human PDAC samples, high Lin28B expression was associated with decreased let-7 expression and increased c-MYC, HMGA2 and KRAS expression. Thus, Lin28B is a novel marker for predicting the prognosis of patients with PDAC and might be a potential therapeutic target for PDAC.
Collapse
Affiliation(s)
- Yunchao Wang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China.,Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing 400038, PR China
| | - Jian Li
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Shixiang Guo
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Yongsheng Ouyang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Liangyu Yin
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Songsong Liu
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Zhiping Zhao
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Jiali Yang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Wenjie Huang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, PR China
| | - Huan Qin
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Xin Zhao
- Department of General Surgery, The First Affiliated Hospital, Soochow University, Jiangsu 215006, PR China
| | - Bing Ni
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing 400038, PR China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| |
Collapse
|
22
|
Rosario R, Childs AJ, Anderson RA. RNA-binding proteins in human oogenesis: Balancing differentiation and self-renewal in the female fetal germline. Stem Cell Res 2017; 21:193-201. [PMID: 28434825 PMCID: PMC5446320 DOI: 10.1016/j.scr.2017.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/29/2017] [Accepted: 04/13/2017] [Indexed: 12/11/2022] Open
Abstract
Primordial germ cells undergo three significant processes on their path to becoming primary oocytes: the initiation of meiosis, the formation and breakdown of germ cell nests, and the assembly of single oocytes into primordial follicles. However at the onset of meiosis, the germ cell becomes transcriptionally silenced. Consequently translational control of pre-stored mRNAs plays a central role in coordinating gene expression throughout the remainder of oogenesis; RNA binding proteins are key to this regulation. In this review we examine the role of exemplars of such proteins, namely LIN28, DAZL, BOLL and FMRP, and highlight how their roles during germ cell development are critical to oogenesis and the establishment of the primordial follicle pool. RNA-binding proteins (RBPs) are key regulators of gene expression during oogenesis. RBPs LIN28, DAZL, BOLL and FMRP display stage-specific expression in fetal oocytes. LIN28 and DAZL may regulate self-renewal and progression into meiosis respectively. BOLL and FMRP may be involved in the later stages of prophase I and oocyte growth. RBPs may have critical roles in establishing the ovarian reserve during fetal life.
Collapse
Affiliation(s)
- Roseanne Rosario
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Andrew J Childs
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London NW1 0TU, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
23
|
Abstract
The somatic component of follicular structure is a mixture of different cell types, represented by Granulosa cells (GCs) that are the paracrine regulators of the oocyte growth. GCs finely support this process by a continuous bidirectional talk with oocyte, which ensure oocyte quality and competence. Specific pathways are involved in the cross-talk and in both GCs and oocyte development. This review summarizes data from GCs gene expression analysis concerning both their physiological role and their interaction with oocyte. We also explore the CGs transcriptome modifications induced by controlled ovarian stimulation (COS) or pathological conditions and their impact in reproduction. The transcriptome analysis of GCs could be a powerful tool to improve our knowledge about the pathways involved in oocyte development. This approach, associated with new technologies as RNA-seq could allow the identifications of new noninvasive biological markers of oocyte quality to increase the efficiency of clinical IVF. Moreover, GCs expression analysis could be useful to shed light on new therapeutic targets by providing new options for the treatment of infertility.
Collapse
Affiliation(s)
- Marco D'Aurora
- a Department of Psychological , Health and Territorial Sciences, School of Medicine, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
- b Center of excellence on Aging, CeSI-met, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
| | - Samantha Sperduti
- b Center of excellence on Aging, CeSI-met, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
- c Department of Medical , Oral and Biotechnological Sciences, School of Medicine, "G. d'Annunzio" University Chieti-Pescara , Italy
| | - Giovanna Di Emidio
- d Department of Health , Life and Environmental Sciences, University of L'Aquila , L'Aquila , Italy Coppito , and
| | - Liborio Stuppia
- a Department of Psychological , Health and Territorial Sciences, School of Medicine, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
- b Center of excellence on Aging, CeSI-met, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
| | - Paolo Giovanni Artini
- e Department of Experimental and Clinical Medicine , Division of Gynecology and Obstetrics, University of Pisa , Pisa , Italy
| | - Valentina Gatta
- a Department of Psychological , Health and Territorial Sciences, School of Medicine, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
- b Center of excellence on Aging, CeSI-met, "G. d'Annunzio" University Chieti-Pescara , Chieti , Italy
| |
Collapse
|
24
|
Rosario R, Filis P, Tessyman V, Kinnell H, Childs AJ, Gray NK, Anderson RA. FMRP Associates with Cytoplasmic Granules at the Onset of Meiosis in the Human Oocyte. PLoS One 2016; 11:e0163987. [PMID: 27695106 PMCID: PMC5047637 DOI: 10.1371/journal.pone.0163987] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/16/2016] [Indexed: 01/09/2023] Open
Abstract
Germ cell development and primordial follicle formation during fetal life is critical in establishing the pool of oocytes that subsequently determines the reproductive lifespan of women. Fragile X-associated primary ovarian insufficiency (FXPOI) is caused by inheritance of the FMR1 premutation allele and approximately 20% of women with the premutation allele develop ovarian dysfunction and premature ovarian insufficiency. However, the underlying disease mechanism remains obscure, and a potential role of FMRP in human ovarian development has not been explored. We have characterised the expression of FMR1 and FMRP in the human fetal ovary at the time of germ cell entry into meiosis through to primordial follicle formation. FMRP expression is exclusively in germ cells in the human fetal ovary. Increased FMRP expression in germ cells coincides with the loss of pluripotency-associated protein expression, and entry into meiosis is associated with FMRP granulation. In addition, we have uncovered FMRP association with components of P-bodies and stress granules, suggesting it may have a role in mRNA metabolism at the time of onset of meiosis. Therefore, this data support the hypothesis that FMRP plays a role regulating mRNAs during pivotal maturational processes in fetal germ cells, and ovarian dysfunction resulting from FMR1 premutation may have its origins during these stages of oocyte development.
Collapse
Affiliation(s)
- Roseanne Rosario
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Panagiotis Filis
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Victoria Tessyman
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Hazel Kinnell
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Andrew J. Childs
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Nicola K. Gray
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, Queen’s Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom
- * E-mail:
| |
Collapse
|
25
|
Jung D, Kee K. Insights into female germ cell biology: from in vivo development to in vitro derivations. Asian J Androl 2016; 17:415-20. [PMID: 25652637 PMCID: PMC4430939 DOI: 10.4103/1008-682x.148077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Understanding the mechanisms of human germ cell biology is important for developing infertility treatments. However, little is known about the mechanisms that regulate human gametogenesis due to the difficulties in collecting samples, especially germ cells during fetal development. In contrast to the mitotic arrest of spermatogonia stem cells in the fetal testis, female germ cells proceed into meiosis and began folliculogenesis in fetal ovaries. Regulations of these developmental events, including the initiation of meiosis and the endowment of primordial follicles, remain an enigma. Studying the molecular mechanisms of female germ cell biology in the human ovary has been mostly limited to spatiotemporal characterizations of genes or proteins. Recent efforts in utilizing in vitro differentiation system of stem cells to derive germ cells have allowed researchers to begin studying molecular mechanisms during human germ cell development. Meanwhile, the possibility of isolating female germline stem cells in adult ovaries also excites researchers and generates many debates. This review will mainly focus on presenting and discussing recent in vivo and in vitro studies on female germ cell biology in human. The topics will highlight the progress made in understanding the three main stages of germ cell developments: namely, primordial germ cell formation, meiotic initiation, and folliculogenesis.
Collapse
Affiliation(s)
| | - Kehkooi Kee
- Department of Basic Medical Sciences, Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
26
|
Dean A, van den Driesche S, Wang Y, McKinnell C, Macpherson S, Eddie SL, Kinnell H, Hurtado-Gonzalez P, Chambers TJ, Stevenson K, Wolfinger E, Hrabalkova L, Calarrao A, Bayne RA, Hagen CP, Mitchell RT, Anderson RA, Sharpe RM. Analgesic exposure in pregnant rats affects fetal germ cell development with inter-generational reproductive consequences. Sci Rep 2016; 6:19789. [PMID: 26813099 PMCID: PMC4728385 DOI: 10.1038/srep19789] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023] Open
Abstract
Analgesics which affect prostaglandin (PG) pathways are used by most pregnant women. As germ cells (GC) undergo developmental and epigenetic changes in fetal life and are PG targets, we investigated if exposure of pregnant rats to analgesics (indomethacin or acetaminophen) affected GC development and reproductive function in resulting offspring (F1) or in the F2 generation. Exposure to either analgesic reduced F1 fetal GC number in both sexes and altered the tempo of fetal GC development sex-dependently, with delayed meiotic entry in oogonia but accelerated GC differentiation in males. These effects persisted in adult F1 females as reduced ovarian and litter size, whereas F1 males recovered normal GC numbers and fertility by adulthood. F2 offspring deriving from an analgesic-exposed F1 parent also exhibited sex-specific changes. F2 males exhibited normal reproductive development whereas F2 females had smaller ovaries and reduced follicle numbers during puberty/adulthood; as similar changes were found for F2 offspring of analgesic-exposed F1 fathers or mothers, we interpret this as potentially indicating an analgesic-induced change to GC in F1. Assuming our results are translatable to humans, they raise concerns that analgesic use in pregnancy could potentially affect fertility of resulting daughters and grand-daughters.
Collapse
Affiliation(s)
- Afshan Dean
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Sander van den Driesche
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Yili Wang
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Chris McKinnell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Sheila Macpherson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Sharon L Eddie
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Hazel Kinnell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Pablo Hurtado-Gonzalez
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Tom J Chambers
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Kerrie Stevenson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Elke Wolfinger
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Lenka Hrabalkova
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Ana Calarrao
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Rosey Al Bayne
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Casper P Hagen
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Richard M Sharpe
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
27
|
MicroRNAs: From Female Fertility, Germ Cells, and Stem Cells to Cancer in Humans. Stem Cells Int 2015; 2016:3984937. [PMID: 26664407 PMCID: PMC4655303 DOI: 10.1155/2016/3984937] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/19/2015] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs are a family of naturally occurring small noncoding RNA molecules that play an important regulatory role in gene expression. They are suggested to regulate a large proportion of protein encoding genes by mediating the translational suppression and posttranscriptional control of gene expression. Recent findings show that microRNAs are emerging as important regulators of cellular differentiation and dedifferentiation, and are deeply involved in developmental processes including human preimplantation development. They keep a balance between pluripotency and differentiation in the embryo and embryonic stem cells. Moreover, it became evident that dysregulation of microRNA expression may play a fundamental role in progression and dissemination of different cancers including ovarian cancer. The interest is still increased by the discovery of exosomes, that is, cell-derived vesicles, which can carry different proteins but also microRNAs between different cells and are involved in cell-to-cell communication. MicroRNAs, together with exosomes, have a great potential to be used for prognosis, therapy, and biomarkers of different diseases including infertility. The aim of this review paper is to summarize the existent knowledge on microRNAs related to female fertility and cancer: from primordial germ cells and ovarian function, germinal stem cells, oocytes, and embryos to embryonic stem cells.
Collapse
|
28
|
Findlay JK, Hutt KJ, Hickey M, Anderson RA. How Is the Number of Primordial Follicles in the Ovarian Reserve Established? Biol Reprod 2015; 93:111. [PMID: 26423124 DOI: 10.1095/biolreprod.115.133652] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/22/2015] [Indexed: 12/15/2022] Open
Abstract
The number of primordial follicles in the ovarian reserve is an important determinant of the length of the ovarian lifespan, and therefore the fertility of an individual. This reserve contains all of the oocytes potentially available for fertilization throughout the fertile lifespan. The maximum number is set during pregnancy or just after birth in most mammalian species; current evidence does not support neofolliculogenesis after the ovarian reserve is established, although this is increasingly being reexamined. Under physiological circumstances, this number will be influenced by the number of primordial germ cells initially specified in the epiblast of the developing embryo, their proliferation during and after migration to the developing gonads, and their death during oogenesis and formation of primordial follicles at nest breakdown. Death of germ cells during the establishment of the ovarian reserve occurs principally by autophagy or apoptosis, although the triggers that initiate these remain elusive. This review outlines the regulatory steps that determine the number of primordial follicles and thus the number of oocytes in the ovarian reserve at birth, using the mouse as the model, interspersed with human data where available. This information has application for understanding the variability in duration of fertility that occurs between normal individuals and with age, in premature ovarian insufficiency, and after chemotherapy or radiotherapy.
Collapse
Affiliation(s)
- John K Findlay
- Centre for Reproductive Biology, Hudson Institute of Medical Research, Clayton, Victoria, Australia Department of Obstetrics & Gynaecology, Monash University, Clayton, Victoria, Australia Department of Obstetrics & Gynaecology, University of Melbourne, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Karla J Hutt
- Centre for Reproductive Biology, Hudson Institute of Medical Research, Clayton, Victoria, Australia Department of Anatomy & Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Martha Hickey
- Department of Obstetrics & Gynaecology, University of Melbourne, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Richard A Anderson
- Medical Research Council Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
29
|
Gonadal development and germ cell tumors in mouse and humans. Semin Cell Dev Biol 2015; 45:114-23. [DOI: 10.1016/j.semcdb.2015.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/01/2015] [Indexed: 12/12/2022]
|
30
|
Ren Y, Suzuki H, Jagarlamudi K, Golnoski K, McGuire M, Lopes R, Pachnis V, Rajkovic A. Lhx8 regulates primordial follicle activation and postnatal folliculogenesis. BMC Biol 2015; 13:39. [PMID: 26076587 PMCID: PMC4487509 DOI: 10.1186/s12915-015-0151-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/10/2015] [Indexed: 12/23/2022] Open
Abstract
Background The early stages of ovarian follicle formation—beginning with the breakdown of germ cell cysts and continuing with the formation of primordial follicles and transition to primary and secondary follicles—are critical in determining reproductive life span and fertility. Previously, we discovered that global knockouts of germ cell-specific transcriptional co-regulators Sohlh1, Sohlh2, Lhx8, and Nobox, cause rapid oocyte loss and ovarian failure. Also factors such as Nobox and Sohlh1 are associated with human premature ovarian failure. In this study, we developed a conditional knockout of Lhx8 to study oocyte-specific pathways in postnatal folliculogenesis. Results The conditional deficiency of Lhx8 in the oocytes of primordial follicles leads to massive primordial oocyte activation, in part, by indirectly interacting with the PI3K-AKT pathway, as shown by synergistic effects on FOXO3 nucleocytoplasmic translocation and rpS6 activation. However, LHX8 does not directly regulate members of the PI3K-AKT pathway; instead, we show that LHX8 represses Lin28a expression, a known regulator of mammalian metabolism and of the AKT/mTOR pathway. LHX8 can bind to the Lin28a promoter, and the depletion of Lin28a in Lhx8-deficient oocytes partially suppresses primordial oocyte activation. Moreover, unlike the PI3K-AKT pathway, LHX8 is critical beyond primordial follicle activation, and blocks the primary to secondary follicle transition. Conclusions Our results indicate that the LHX8-LIN28A pathway is essential in the earliest stages of primordial follicle activation, and LHX8 is an important oocyte-specific transcription factor in the ovary for regulating postnatal folliculogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12915-015-0151-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu Ren
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Hitomi Suzuki
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Krishna Jagarlamudi
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Kayla Golnoski
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Megan McGuire
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Rita Lopes
- Division of Molecular Neurobiology, MRC National Institute of Medical Research, London, NW7 1AA, UK.
| | - Vassilis Pachnis
- Division of Molecular Neurobiology, MRC National Institute of Medical Research, London, NW7 1AA, UK.
| | - Aleksandar Rajkovic
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
31
|
Bayne RAL, Kinnell HL, Coutts SM, He J, Childs AJ, Anderson RA. GDF9 is transiently expressed in oocytes before follicle formation in the human fetal ovary and is regulated by a novel NOBOX transcript. PLoS One 2015; 10:e0119819. [PMID: 25790371 PMCID: PMC4366263 DOI: 10.1371/journal.pone.0119819] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/16/2015] [Indexed: 12/05/2022] Open
Abstract
During human fetal ovary development, the process of primordial follicle formation is immediately preceded by a highly dynamic period of germ cell and somatic cell reorganisation. This is regulated by germ-cell specific transcription regulators, by the conserved RNA binding proteins DAZL and BOLL and by secreted growth factors of the TGFβ family, including activin βA: these all show changing patterns of expression preceding follicle formation. In mice, the transcription factor Nobox is essential for follicle formation and oocyte survival, and NOBOX regulates the expression of GDF9 in humans. We have therefore characterised the expression of GDF9 in relation to these known key factors during follicle formation in the human fetal ovary. mRNA levels of GDF9, BMP15 and NOBOX were quantified by qRT-PCR and showed dramatic increases across gestation. GDF9 protein expression was localised by immunohistochemistry to the same population of germ cells as those expressing activin βA prior to follicle formation but did not co-localise with either BOLL or DAZL. A novel NOBOX isoform was identified in fetal ovary that was shown to be capable of up-regulating the GDF9 promoter in reporter assays. Thus, during oogenesis in humans, oocytes go through a dynamic and very sharply demarcated sequence of changes in expression of these various proteins, even within individual germ cell nests, likely to be of major functional significance in determining selective germ cell survival at this key stage in ovarian development. Transcriptional variation may contribute to the range of age of onset of POI in women with NOBOX mutations.
Collapse
Affiliation(s)
- Rosemary A. L. Bayne
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
- * E-mail:
| | - Hazel L. Kinnell
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Shiona M. Coutts
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Jing He
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Andrew J. Childs
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, United Kingdom
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| |
Collapse
|
32
|
Sugawa F, Araúzo-Bravo MJ, Yoon J, Kim KP, Aramaki S, Wu G, Stehling M, Psathaki OE, Hübner K, Schöler HR. Human primordial germ cell commitment in vitro associates with a unique PRDM14 expression profile. EMBO J 2015; 34:1009-24. [PMID: 25750208 PMCID: PMC4406649 DOI: 10.15252/embj.201488049] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 02/18/2015] [Indexed: 01/30/2023] Open
Abstract
Primordial germ cells (PGCs) develop only into sperm and oocytes in vivo. The molecular mechanisms underlying human PGC specification are poorly understood due to inaccessibility of cell materials and lack of in vitro models for tracking the earliest stages of germ cell development. Here, we describe a defined and stepwise differentiation system for inducing pre-migratory PGC-like cells (PGCLCs) from human pluripotent stem cells (PSCs). In response to cytokines, PSCs differentiate first into a heterogeneous mesoderm-like cell population and then into PGCLCs, which exhibit minimal PRDM14 expression. PGC specification in humans is similar to the murine process, with the sequential activation of mesodermal and PGC genes, and the suppression of neural induction and of de novo DNA methylation, suggesting that human PGC formation is induced via epigenesis, the process of germ cell specification via inductive signals from surrounding somatic cells. This study demonstrates that PGC commitment in humans shares key features with that of the mouse, but also highlights key differences, including transcriptional regulation during the early stage of human PGC development (3–6 weeks). A more comprehensive understanding of human germ cell development may lead to methodology for successfully generating PSC-derived gametes for reproductive medicine.
Collapse
Affiliation(s)
- Fumihiro Sugawa
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Marcos J Araúzo-Bravo
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany Group of Computational Biology and Bioinformatics, Biodonostia Health Research Institute, San Sebastián, Spain IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Juyong Yoon
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Kee-Pyo Kim
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Shinya Aramaki
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Guangming Wu
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Martin Stehling
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Olympia E Psathaki
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Karin Hübner
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany Medical Faculty, University of Münster, Münster, Germany
| |
Collapse
|
33
|
Aeckerle N, Drummer C, Debowski K, Viebahn C, Behr R. Primordial germ cell development in the marmoset monkey as revealed by pluripotency factor expression: suggestion of a novel model of embryonic germ cell translocation. Mol Hum Reprod 2014; 21:66-80. [PMID: 25237007 PMCID: PMC4275041 DOI: 10.1093/molehr/gau088] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Primordial germ cells (PGCs) are the embryonic progenitors of sperm and egg cells. Mammalian PGCs are thought to actively migrate from the yolk sac endoderm over long distances across the embryo to reach the somatic genital ridges. The general principles of mammalian PGC development were discovered in mice. In contrast, little is known about PGC development in primates due to extremely limited access to primate embryos. Here, we analyzed 12 well preserved marmoset monkey (Callithrix jacchus) embryos covering the phase from PGC emergence in the endoderm to the formation of the sexually differentiated gonad (embryonic day (E) 50 to E95). We show using immunohistochemistry that the pluripotency factors OCT4A and NANOG specifically mark PGCs throughout the period studied. In contrast, SALL4 and LIN28 were first expressed ubiquitously and only later down-regulated in somatic tissues. We further show, for the first time, that PGCs are located in the endoderm in E50 embryos in close spatial proximity to the prospective genital ridge, making a long-range migration of PGCs dispensable. At E65, PGCs are already present in the primitive gonad, while significantly later embryonic stages still exhibit PGCs at their original endodermal site, revealing a wide spatio-temporal window of PGC distribution. Our findings challenge the ‘dogma’ of active long-range PGC migration from the endoderm to the gonads. We therefore favor an alternative model based primarily on passive translocation of PGCs from the mesenchyme that surrounds the gut to the prospective gonad through the intercalar expansion of mesenchymal tissue which contains the PGCs. In summary, we (i) show differential pluripotency factor expression during primate embryo development and (ii) provide a schematic model for embryonic PGC translocation.
Collapse
Affiliation(s)
- N Aeckerle
- Stem Cell Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - C Drummer
- Stem Cell Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - K Debowski
- Stem Cell Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - C Viebahn
- Department of Anatomy and Embryology, Center of Anatomy, University of Göttingen, Kreuzbergring 36, 37075 Göttingen, Germany
| | - R Behr
- Stem Cell Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| |
Collapse
|
34
|
Yuan J, Zhang D, Wang L, Liu M, Mao J, Yin Y, Ye X, Liu N, Han J, Gao Y, Cheng T, Keefe DL, Liu L. No evidence for neo-oogenesis may link to ovarian senescence in adult monkey. Stem Cells 2014; 31:2538-50. [PMID: 23897655 DOI: 10.1002/stem.1480] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 06/18/2013] [Accepted: 07/05/2013] [Indexed: 12/15/2022]
Abstract
Female germline or oogonial stem cells transiently residing in fetal ovaries are analogous to the spermatogonial stem cells or germline stem cells (GSCs) in adult testes where GSCs and meiosis continuously renew. Oocytes can be generated in vitro from embryonic stem cells and induced pluripotent stem cells, but the existence of GSCs and neo-oogenesis in adult mammalian ovaries is less clear. Preliminary findings of GSCs and neo-oogenesis in mice and humans have not been consistently reproducible. Monkeys provide the most relevant model of human ovarian biology. We searched for GSCs and neo-meiosis in ovaries of adult monkeys at various ages, and compared them with GSCs from adult monkey testis, which are characterized by cytoplasmic staining for the germ cell marker DAZL and nuclear expression of the proliferative markers PCNA and KI67, and pluripotency-associated genes LIN28 and SOX2, and lack of nuclear LAMIN A, a marker for cell differentiation. Early meiocytes undergo homologous pairing at prophase I distinguished by synaptonemal complex lateral filaments with telomere perinuclear distribution. By exhaustive searching using comprehensive experimental approaches, we show that proliferative GSCs and neo-meiocytes by these specific criteria were undetectable in adult mouse and monkey ovaries. However, we found proliferative nongermline somatic stem cells that do not express LAMIN A and germ cell markers in the adult ovaries, notably in the cortex and granulosa cells of growing follicles. These data support the paradigm that adult ovaries do not undergo germ cell renewal, which may contribute significantly to ovarian senescence that occurs with age.
Collapse
Affiliation(s)
- Jihong Yuan
- State Key Laboratory of Medicinal Chemical Biology, The 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics and College of Life Sciences, Nankai University, Tianjin, China; Key Laboratory of Ministry of Health on Hormones and Development, Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Jørgensen A, Rajpert-De Meyts E. Regulation of meiotic entry and gonadal sex differentiation in the human: normal and disrupted signaling. Biomol Concepts 2014; 5:331-41. [DOI: 10.1515/bmc-2014-0014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 05/28/2014] [Indexed: 11/15/2022] Open
Abstract
AbstractMeiosis is a unique type of cell division that is performed only by germ cells to form haploid gametes. The switch from mitosis to meiosis exhibits a distinct sex-specific difference in timing, with female germ cells entering meiosis during fetal development and male germ cells at puberty when spermatogenesis is initiated. During early fetal development, bipotential primordial germ cells migrate to the forming gonad where they remain sexually indifferent until the sex-specific differentiation of germ cells is initiated by cues from the somatic cells. This irreversible step in gonadal sex differentiation involves the initiation of meiosis in fetal ovaries and prevention of meiosis in the germ cells of fetal testes. During the last decade, major advances in the understanding of meiosis regulation have been accomplished, with the discovery of retinoic acid as an inducer of meiosis being the most prominent finding. Knowledge about the molecular mechanisms regulating meiosis signaling has mainly been established by studies in rodents, while this has not yet been extensively investigated in humans. In this review, the current knowledge about the regulation of meiosis signaling is summarized and placed in the context of fetal gonad development and germ cell differentiation, with emphasis on results obtained in humans. Furthermore, the consequences of dysregulated meiosis signaling in humans are briefly discussed in the context of selected pathologies, including testicular germ cell cancer and some forms of male infertility.
Collapse
Affiliation(s)
- Anne Jørgensen
- 1Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- 1Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| |
Collapse
|
36
|
Hu Q, Peng J, Liu W, He X, Cui L, Chen X, Yang M, Liu H, Liu S, Wang H. Lin28B is a novel prognostic marker in gastric adenocarcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:5083-5092. [PMID: 25197381 PMCID: PMC4152071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/12/2014] [Indexed: 06/03/2023]
Abstract
Lin28B, a homologue of Lin28, represses biogenesis of let-7 microRNAs with a role in tumorigenesis and is considered a potential therapeutic target for various human cancers. The aim of the study was to identify the clinical significance of Lin28B in gastric adenocarcinoma (GAC). We examined the expression of Lin28B in 97 human gastric cancer samples with 32 samples of non-dysplastic tissues by immunohistochemistry. In the 97 GAC cases, 42 were with high Lin28B expression. The expression levels of Lin28B proteins in GAC were higher than in corresponding adjacent normal tissues (P=0.001). Significant correlations were noted between Lin28B expression and lymph node status (P=0.005), TNM stage (P < 0.001), tumor invasion (P=0.036), and differentiation (P=0.001) of GAC patients. The Kaplan-Meier estimates showed a negative correlation of overall 5-year survival rate with Lin28B expression where higher expression resulted in poorer prognosis in GAC. In univariate analysis, lymph node metastasis, TNM stage, serosal invasion, Lin28B expression as well as differentiation grade could predict the prognosis of GAC patients (P=0.002, P < 0.001, P=0.003, P < 0.001, P=0.001, respectively). Multivariate analysis revealed that the expression level of Lin28B (P < 0.001), TNM stage (P < 0.001) as well as differentiation grade (P < 0.001) were the three potential independent prognostic factors in our study [Hazard ratio (HR)=2.108 and P=0.017, HR=1.994 and P=0.018, HR=1.939 and P=0.046, respectively]. Our findings point to the prognostic role of Lin28B in GAC, and indicate Lin28B as a potential therapeutic target of GAC patients.
Collapse
Affiliation(s)
- Qian Hu
- Laboratory of Genetics, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Laboratory of Cell and Gene Therapy, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationChengdu 610041, China
| | - Jing Peng
- Laboratory of Genetics, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Laboratory of Cell and Gene Therapy, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationChengdu 610041, China
| | - Weiping Liu
- Department of Pathology, West China Hospital of Sichuan University37 Guoxue Street, Chengdu, Sichuan 610041, China
| | - Xiaoli He
- Department of Obstetric and Gynecologic, Henan Provincial People’s HospitalZhengzhou, Henan 450003, China
| | - Ling Cui
- Department of Gynecological Oncology, Second People’s Hospital of Sichuan (Sichuan Cancer Hospital)Chengdu, Sichuan 610041, China
| | - Xinlian Chen
- Laboratory of Genetics, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Laboratory of Cell and Gene Therapy, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationChengdu 610041, China
| | - Mei Yang
- Laboratory of Genetics, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Laboratory of Cell and Gene Therapy, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationChengdu 610041, China
| | - Hongqian Liu
- Laboratory of Genetics, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Laboratory of Cell and Gene Therapy, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationChengdu 610041, China
| | - Shanling Liu
- Laboratory of Cell and Gene Therapy, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationChengdu 610041, China
| | - He Wang
- Laboratory of Genetics, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Department of Obstetric and Gynecologic, West China Second University Hospital, Sichuan UniversityChengdu 610041, China
- Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of EducationChengdu 610041, China
| |
Collapse
|
37
|
Stratoulias V, Heino TI, Michon F. Lin-28 regulates oogenesis and muscle formation in Drosophila melanogaster. PLoS One 2014; 9:e101141. [PMID: 24963666 PMCID: PMC4071072 DOI: 10.1371/journal.pone.0101141] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 06/03/2014] [Indexed: 01/07/2023] Open
Abstract
Understanding the control of stem cell (SC) differentiation is important to comprehend developmental processes as well as to develop clinical applications. Lin28 is a conserved molecule that is involved in SC maintenance and differentiation by regulating let-7 miRNA maturation. However, little is known about the in vivo function of Lin28. Here, we report critical roles for lin-28 during oogenesis. We found that let-7 maturation was increased in lin-28 null mutant fly ovaries. We showed that lin-28 null mutant female flies displayed reduced fecundity, due to defects in egg chamber formation. More specifically, we demonstrated that in mutant ovaries, the egg chambers fuse during early oogenesis resulting in abnormal late egg chambers. We also showed that this phenotype is the combined result of impaired germline SC differentiation and follicle SC differentiation. We suggest a model in which these multiple oogenesis defects result from a misregulation of the ecdysone signaling network, through the fine-tuning of Abrupt and Fasciclin2 expression. Our results give a better understanding of the evolutionarily conserved role of lin-28 on GSC maintenance and differentiation.
Collapse
Affiliation(s)
| | - Tapio I. Heino
- Department of Biosciences, University of Helsinki, Helsinki, Finland
- * E-mail: (FM); (TH)
| | - Frederic Michon
- Institute of Biotechnology, Developmental Biology Program, University of Helsinki, Helsinki, Finland
- * E-mail: (FM); (TH)
| |
Collapse
|
38
|
Fereydouni B, Drummer C, Aeckerle N, Schlatt S, Behr R. The neonatal marmoset monkey ovary is very primitive exhibiting many oogonia. Reproduction 2014; 148:237-47. [PMID: 24840529 PMCID: PMC4086814 DOI: 10.1530/rep-14-0068] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oogonia are characterized by diploidy and mitotic proliferation. Human and mouse oogonia express several factors such as OCT4, which are characteristic of pluripotent cells. In human, almost all oogonia enter meiosis between weeks 9 and 22 of prenatal development or undergo mitotic arrest and subsequent elimination from the ovary. As a consequence, neonatal human ovaries generally lack oogonia. The same was found in neonatal ovaries of the rhesus monkey, a representative of the old world monkeys (Catarrhini). By contrast, proliferating oogonia were found in adult prosimians (now called Strepsirrhini), which is a group of ‘lower’ primates. The common marmoset monkey (Callithrix jacchus) belongs to the new world monkeys (Platyrrhini) and is increasingly used in reproductive biology and stem cell research. However, ovarian development in the marmoset monkey has not been widely investigated. Herein, we show that the neonatal marmoset ovary has an extremely immature histological appearance compared with the human ovary. It contains numerous oogonia expressing the pluripotency factors OCT4A, SALL4, and LIN28A (LIN28). The pluripotency factor-positive germ cells also express the proliferation marker MKI67 (Ki-67), which has previously been shown in the human ovary to be restricted to premeiotic germ cells. Together, the data demonstrate the primitiveness of the neonatal marmoset ovary compared with human. This study may introduce the marmoset monkey as a non-human primate model to experimentally study the aspects of primate primitive gonad development, follicle assembly, and germ cell biology in vivo.
Collapse
Affiliation(s)
- B Fereydouni
- Stem Cell Biology UnitGerman Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, GermanyCentre of Reproductive Medicine and AndrologyUniversity of Münster, Domagkstraße 11, 48149 Münster, Germany
| | - C Drummer
- Stem Cell Biology UnitGerman Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, GermanyCentre of Reproductive Medicine and AndrologyUniversity of Münster, Domagkstraße 11, 48149 Münster, Germany
| | - N Aeckerle
- Stem Cell Biology UnitGerman Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, GermanyCentre of Reproductive Medicine and AndrologyUniversity of Münster, Domagkstraße 11, 48149 Münster, Germany
| | - S Schlatt
- Stem Cell Biology UnitGerman Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, GermanyCentre of Reproductive Medicine and AndrologyUniversity of Münster, Domagkstraße 11, 48149 Münster, Germany
| | - R Behr
- Stem Cell Biology UnitGerman Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, GermanyCentre of Reproductive Medicine and AndrologyUniversity of Münster, Domagkstraße 11, 48149 Münster, Germany
| |
Collapse
|
39
|
Leitch HG, Tang WWC, Surani MA. Primordial germ-cell development and epigenetic reprogramming in mammals. Curr Top Dev Biol 2014; 104:149-87. [PMID: 23587241 DOI: 10.1016/b978-0-12-416027-9.00005-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Primordial germ cells (PGCs) are the embryonic precursors of the gametes and represent the founder cells of the germline. Specification of PGCs is a critical divergent point during embryogenesis. Whereas the somatic lineages will ultimately perish, cells of the germline have the potential to form a new individual and hence progress to the next generation. It is therefore critical that the genome emerges intact and carrying the appropriate epigenetic information during its passage through the germline. To ensure this fidelity of transmission, PGC development encompasses extensive epigenetic reprogramming. The low cell numbers and relative inaccessibility of PGCs present a challenge to those seeking mechanistic understanding of the crucial developmental and epigenetic processes in this most fascinating of lineages. Here, we present an overview of PGC development in the mouse and compare this with the limited information available for other mammalian species. We believe that a comparative approach will be increasingly important to uncover the extent to which mechanisms are conserved and reveal the critical steps during PGC development in humans.
Collapse
Affiliation(s)
- Harry G Leitch
- Wellcome Trust/Cancer Research UK Gurdon Institute of Cancer and Developmental Biology, University of Cambridge, Cambridge, United Kingdom
| | | | | |
Collapse
|
40
|
Increased expression of Lin28B associates with poor prognosis in patients with oral squamous cell carcinoma. PLoS One 2013; 8:e83869. [PMID: 24386298 PMCID: PMC3875533 DOI: 10.1371/journal.pone.0083869] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 11/09/2013] [Indexed: 02/02/2023] Open
Abstract
Recent studies showed that incomplete cell reprogramming can transform cells into tumour-like cells. Lin28A is associated with fibroblast and sarcoma cell reprogramming, whereas its homologue Lin28B is associated with hematopoietic cell reprogramming. This study aimed to investigate the expression and prognostic difference between Lin28A and Lin28B in oral squamous cell carcinoma (OSCC). Expression level was assessed by immunohistochemistry and staining location was confirmed by immunofluorescence. Prognostic values were analysed and compared by the Kaplan-Meier analysis and uni and multivariate Cox regression models. Besides, in vitro cell assays and in vivo nude mice xenograft were used to demonstrate the influence of increased Lin28B expression in OSCC. Lin28A and Lin28B expression increased in OSCC, and co-expression of Lin28A and Lin28B showed no significant association with patient prognosis. Kaplan-Meier analysis showed that patients with high Lin28B but not Lin28A expression had lower overall survival (OS) rates than those with low Lin28B expression. Further Univariate analysis showed that patients with increased Lin28B expression had shorter disease-free survival (DFS) and shorter OS, while multivariate analysis showed Lin28B overexpression with TNM stage predicted poor prognosis in patients with OSCC. Besides, stable expressing Lin28B in oral cancer cells promoted cell migration, invasion, colony formation, in vivo proliferation and increased the expression of cancer suppressor miRNA let-7 targeted genes IL-6, HMGA2, the EMT markers Snail and Twist, the angiogenesis inducer VEGF, and the apoptosis inhibitor Survivin. These combined results indicate that Lin28B is a novel marker for predicting prognosis in patients with OSCC and may be a therapeutic target.
Collapse
|
41
|
He J, Stewart K, Kinnell HL, Anderson RA, Childs AJ. A developmental stage-specific switch from DAZL to BOLL occurs during fetal oogenesis in humans, but not mice. PLoS One 2013; 8:e73996. [PMID: 24086306 PMCID: PMC3783425 DOI: 10.1371/journal.pone.0073996] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 07/29/2013] [Indexed: 01/24/2023] Open
Abstract
The Deleted in Azoospermia gene family encodes three germ cell-specific RNA-binding proteins (DAZ, DAZL and BOLL) that are essential for gametogenesis in diverse species. Targeted disruption of Boll in mice causes male-specific spermiogenic defects, but females are apparently fertile. Overexpression of human BOLL promotes the derivation of germ cell-like cells from genetically female (XX), but not male (XY) human ES cells however, suggesting a functional role for BOLL in regulating female gametogenesis in humans. Whether BOLL is expressed during oogenesis in mammals also remains unclear. We have therefore investigated the expression of BOLL during fetal oogenesis in humans and mice. We demonstrate that BOLL protein is expressed in the germ cells of the human fetal ovary, at a later developmental stage than, and almost mutually-exclusive to, the expression of DAZL. Strikingly, BOLL is downregulated, and DAZL re-expressed, as primordial follicles form, revealing BOLL expression to be restricted to a narrow window during fetal oogenesis. By quantifying the extent of co-expression of DAZL and BOLL with markers of meiosis, we show that this window likely corresponds to the later stages of meiotic prophase I. Finally, we demonstrate that Boll is also transiently expressed during oogenesis in the fetal mouse ovary, but is simultaneously co-expressed within the same germ cells as Dazl. These data reveal significant similarities and differences between the expression of BOLL homologues during oogenesis in humans and mice, and raise questions as to the validity of the Boll(-/-) mouse as a model for understanding BOLL function during human oogenesis.
Collapse
Affiliation(s)
- Jing He
- MRC Centre for Reproductive Health, the Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Kayleigh Stewart
- MRC Centre for Reproductive Health, the Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Hazel L. Kinnell
- MRC Centre for Reproductive Health, the Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, the Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew J. Childs
- MRC Centre for Reproductive Health, the Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Comparative Biomedical Sciences, the Royal Veterinary College, University of London, Camden, London, United Kingdom
- * E-mail:
| |
Collapse
|
42
|
Anderson RA, McIlwain L, Coutts S, Kinnell HL, Fowler PA, Childs AJ. Activation of the aryl hydrocarbon receptor by a component of cigarette smoke reduces germ cell proliferation in the human fetal ovary. Mol Hum Reprod 2013; 20:42-8. [PMID: 23979962 PMCID: PMC3867980 DOI: 10.1093/molehr/gat059] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Fetal life is a critical time for female fertility, when germ cells complete proliferation, initiate meiosis and ultimately form the lifetime stock of primordial follicles. Female fertility may be reduced by in utero exposure to cigarette smoke, which contains ligands for the aryl hydrocarbon receptor (AhR). The AhR is a critical regulator of ovarian germ cell survival in mice; thus activation of this receptor in the ovaries of fetuses exposed to maternal cigarette smoke in utero may provide a mechanism by which female fertility is reduced in later life. We have therefore investigated AhR expression in the human fetal ovary, and examined the effects of an AhR ligand present in cigarette smoke, on germ cells in human fetal ovaries cultured in vitro. The results showed that AHR mRNA expression increased 2-fold between first and late second trimester (P = 0.008). AhR protein was confined to germ cells at all gestations, but varied from expression in most germ cells during the first trimester, to only patchy expression by clusters of germ cells at later gestations. Culture of human fetal ovaries with the AhR ligand 9,10-dimethyl-1,2-benzanthracene-3,4-dihydrodiol (DMBA-DHD; a component of cigarette smoke) did not affect germ cell number in vitro, but significantly reduced the proportion of proliferating germ cells by 29% (as assessed by phospho-histone H3 staining (P = 0.04)). Germ cell apoptosis was not significantly affected. These results reveal that germ cells in the human fetal ovary express AhR from the proliferative stage of development through entry into meiosis and beyond, and demonstrate that AhR ligands found in cigarette smoke have the capacity to impair human fetal ovarian germ cell proliferation.
Collapse
Affiliation(s)
- Richard A Anderson
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK and
| | | | | | | | | | | |
Collapse
|
43
|
Assou S, Al-edani T, Haouzi D, Philippe N, Lecellier CH, Piquemal D, Commes T, Aït-Ahmed O, Dechaud H, Hamamah S. MicroRNAs: new candidates for the regulation of the human cumulus-oocyte complex. Hum Reprod 2013; 28:3038-49. [PMID: 23904466 DOI: 10.1093/humrep/det321] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
STUDY QUESTION What is the expression pattern of microRNAs (miRNAs) in human cumulus-oocyte complexes (COCs)? SUMMARY ANSWER Several miRNAs are enriched in cumulus cells (CCs) or oocytes, and are predicted to target genes involved in biological functions of the COC. WHAT IS KNOWN ALREADY The transcriptional profiles of human MII oocytes and the surrounding CCs are known. However, very limited data are available about post-transcriptional regulators, such as miRNAs. This is the first study focussing on the identification and quantification of small RNAs, including miRNAs, in human oocytes and CCs using a deep-sequencing approach. STUDY DESIGN, SIZE, DURATION MII oocytes and CCs were collected from women who underwent IVF. PARTICIPANTS/MATERIALS, SETTING, METHODS Using the Illumina/deep-sequencing technology, we analyzed the small RNAome of pooled MII oocytes (n = 24) and CC samples (n = 20). The mRNA targets of CC and MII oocyte miRNAs were identified using in silico prediction algorithms. Using oligonucleotide microarrays, genome-wide gene expression was studied in oocytes (10 pools of 19 ± 3 oocytes/each) and 10 individual CC samples. TaqMan miRNA assays were used to confirm the sequencing results in independent pools of MII oocytes (3 pools of 8 ± 3 oocytes/each) and CC samples (3 pools of 7 ± 3 CCs/each). The functional role of one miRNA, MIR23a, was assessed in primary cultures of human CCs. MAIN RESULTS AND THE ROLE OF CHANCE Deep sequencing of small RNAs yielded more than 1 million raw reads. By mapping reads with a single location to the human genome, known miRNAs that were abundant in MII oocytes (MIR184, MIR100 and MIR10A) or CCs (MIR29a, MIR30d, MIR21, MIR93, MIR320a, MIR125a and the LET7 family) were identified. Predicted target genes of the oocyte miRNAs were associated with the regulation of transcription and cell cycle, whereas genes targeted by CC miRNAs were involved in extracellular matrix and apoptosis. Comparison of the predicted miRNA target genes and mRNA microarray data resulted in a list of 224 target genes that were differentially expressed in MII oocytes and CCs, including PTGS2, CTGF and BMPR1B that are important for cumulus-oocyte communication. Functional analysis using primary CC cultures revealed that BCL2 and CYP19A1 mRNA levels were decreased upon MIR23a overexpression. LIMITATIONS, REASONS FOR CAUTION Only known miRNAs were investigated in the present study on COCs. Moreover, the source of the material is MII oocytes that failed to fertilize. WIDER IMPLICATIONS OF THE FINDINGS The present findings suggest that miRNA could play a role in the regulation of the oocyte and CC crosstalk. STUDY FUNDING/COMPETING INTEREST(S) This work was partially supported by a grant from Ferring Pharmaceuticals. The authors of the study have no conflict of interest to report. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- S Assou
- Université Montpellier 1, UFR de Médecine, Montpellier, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Seervai RNH, Wessel GM. Lessons for inductive germline determination. Mol Reprod Dev 2013; 80:590-609. [PMID: 23450642 DOI: 10.1002/mrd.22151] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 01/03/2013] [Indexed: 12/25/2022]
Abstract
Formation of the germline in an embryo marks a fresh round of reproductive potential, yet the developmental stage and location within the embryo where the primordial germ cells (PGCs) form differs wildly among species. In most animals, the germline is formed either by an inherited mechanism, in which maternal provisions within the oocyte drive localized germ-cell fate once acquired in the embryo, or an inductive mechanism that involves signaling between cells that directs germ-cell fate. The inherited mechanism has been widely studied in model organisms such as Drosophila melanogaster, Caenorhabditis elegans, Xenopus laevis, and Danio rerio. Given the rapid generation time and the effective adaptation for laboratory research of these organisms, it is not coincidental that research on these organisms has led the field in elucidating mechanisms for germline specification. The inductive mechanism, however, is less well understood and is studied primarily in the mouse (Mus musculus). In this review, we compare and contrast these two fundamental mechanisms for germline determination, beginning with the key molecular determinants that play a role in the formation of germ cells across all animal taxa. We next explore the current understanding of the inductive mechanism of germ-cell determination in mice, and evaluate the hypotheses for selective pressures on these contrasting mechanisms. We then discuss the hypothesis that the transition between these determination mechanisms, which has happened many times in phylogeny, is more of a continuum than a binary change. Finally, we propose an analogy between germline determination and sex determination in vertebrates-two of the milestones of reproduction and development-in which animals use contrasting strategies to activate similar pathways.
Collapse
Affiliation(s)
- Riyad N H Seervai
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, 02192, USA
| | | |
Collapse
|
45
|
Abstract
Primordial germ cell (PGC) development in the human fetus remains relatively uncharted. A new study suggests that epigenetic reprogramming and sex differentiation in human PGCs occur asynchronously over an extended time period. This finding raises questions and implications for in vitro PGC differentiation.
Collapse
|
46
|
Jørgensen A, Nielsen JE, Almstrup K, Toft BG, Petersen BL, Rajpert-De Meyts E. Dysregulation of the mitosis-meiosis switch in testicular carcinoma in situ. J Pathol 2013; 229:588-98. [PMID: 23303528 DOI: 10.1002/path.4154] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 11/15/2012] [Accepted: 11/27/2012] [Indexed: 12/17/2022]
Abstract
Testicular germ cell tumours (TGCT) of young adults arise from the intratubular precursor, carcinoma in situ (CIS). CIS cells are thought to be developmentally arrested and transformed fetal germ cells that survive through childhood and gain invasive capacity after puberty. Given that germ cell neoplasms arise frequently in undervirilized and dysgenetic gonads and the striking physiological difference between meiotic entry in ovaries (fetal life) versus testes (at puberty), this study aimed to investigate whether errors in regulation of meiosis may be implicated in the pathogenesis of CIS or its invasive progression to TGCT. The main focus was on a key sex differentiation and meiosis regulator, DMRT1, which has also been linked to TGCT risk in recent genetic association studies. Expression patterns of DMRT1 and other meiosis regulators (SCP3, DMC1, STRA8, CYP26B1, NANOS2, NANOS3) were investigated in pre- and post-pubertal CIS samples and TGCT by quantitative RT-PCR and immunohistochemistry. The results demonstrated that meiosis markers and meiosis inhibitors were simultaneously expressed in CIS cells, in both pre- and post-pubertal testis samples. DMRT1 was present in a restricted subset of CIS cells, which was relatively greater in pre-pubertal (27%) compared to adult (2.6%) samples. In contrast to the majority of CIS cells, DMRT1-positive CIS cells in adult testes were not proliferating. DMRT1 and most of the other meiosis regulators were absent or expressed at low levels in invasive TGCT, except in spermatocytic seminoma (not derived from CIS). In conclusion, this study indicates that meiosis signalling is dysregulated in CIS cells and that a key regulator of the mitosis-meiosis switch, DMRT1, is expressed in 'early-stage' CIS cells but is down-regulated with further invasive transformation. Whether this mixed meiosis signalling in CIS cells is caused by insufficient virilization of the fetal somatic niche or a partial post-pubertal maturation remains uncertain and requires further study.
Collapse
Affiliation(s)
- Anne Jørgensen
- Department of Growth and Reproduction, Copenhagen University Hospital, Denmark
| | | | | | | | | | | |
Collapse
|