1
|
He Z, Yan RG, Shang QB, Yang QE. Elevated Id2 expression causes defective meiosis and spermatogenesis in mice. Dev Dyn 2024; 253:593-605. [PMID: 38063258 DOI: 10.1002/dvdy.676] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/11/2023] [Accepted: 11/14/2023] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Inhibitors of DNA binding (ID) proteins mainly inhibit gene expression and regulate cell fate decisions by interacting with E-proteins. All four ID proteins (ID1-4) are present in the testis, and ID4 has a particularly important role in spermatogonial stem cell fate determination. Several lines of evidence indicate that ID proteins are involved in meiosis; however, functional experiments have not been conducted to validate this observation. RESULTS In this study, we report that ID2 is enriched in spermatocytes and that forced ID2 expression in germ cells causes defects in spermatogenesis. A detailed analysis demonstrated that Id2 overexpression (Id2 OE) decreased the total number of spermatogonia and changed the dynamics of meiosis progression. Specifically, spermatocytes were enriched in the zygotene stage, and the proportion of pachytene spermatocytes was significantly decreased, indicating defects in the zygotene-pachytene transition. The number of MLH1-positive foci per cell was decreased in pachytene spermatocytes from Id2 OE testes, suggesting abnormalities in recombination. Transcriptome analysis revealed that forced Id2 expression changed the expression of a list of genes mainly associated with meiosis and spermatid development. CONCLUSIONS ID2 protein is expressed in spermatocytes, and its genetic ablation in the germline does not affect spermatogenesis, likely due to genetic compensation of its family members. However, forced Id2 expression changes meiosis progression and causes defects in spermiogenesis. These data provide important evidence that ID proteins play pivotal roles in male meiosis and spermatid development.
Collapse
Affiliation(s)
- Zhen He
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Rong-Ge Yan
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qin-Bang Shang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China
| | - Qi-En Yang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Laboratory of Plateau Animal Breeding and Functional Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China
| |
Collapse
|
2
|
Wu Z, Chen X, Yan T, Yu L, Zhang L, Zheng M, Zhu H. Rreb1 is a key transcription factor in Sertoli cell maturation and function and spermatogenesis in mouse. ZYGOTE 2024; 32:130-138. [PMID: 38248872 DOI: 10.1017/s0967199423000655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Spermatogenesis is a developmental process driven by interactions between germ cells and Sertoli cells. This process depends on appropriate gene expression, which might be regulated by transcription factors. This study focused on Rreb1, a zinc finger transcription factor, and explored its function and molecular mechanisms in spermatogenesis in a mouse model. Our results showed that RREB1 was predominantly expressed in the Sertoli cells of the testis. The decreased expression of RREB1 following injection of siRNA caused impaired Sertoli cell development, which was characterized using a defective blood-testis barrier structure and decreased expression of Sertoli cell functional maturity markers; its essential trigger might be SMAD3 destabilization. The decreased expression of RREB1 in mature Sertoli cells influenced the cell structure and function, which resulted in abnormal spermatogenesis, manifested as oligoasthenoteratozoospermia, and we believe RREB1 plays this role by regulating the transcription of Fshr and Wt1. RREB1 has been reported to activate Fshr transcription, and we demonstrated that the knockdown of Rreb1 caused a reduction in follicle-stimulating hormone receptor (FSHR) in the testis, which could be the cause of the increased sperm malformation. Furthermore, we confirmed that RREB1 directly activates Wt1 promoter activity, and RREB1 downregulation induced the decreased expression of Wt1 and its downstream polarity-associated genes Par6b and E-cadherin, which caused increased germ-cell death and reduced sperm number and motility. In conclusion, RREB1 is a key transcription factor essential for Sertoli cell development and function and is required for normal spermatogenesis.
Collapse
Affiliation(s)
- Zhu Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Xu Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Tong Yan
- Department of Histology and Embryology, School of Basic Medical Sciences, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Li Yu
- Department of Histology and Embryology, School of Basic Medical Sciences, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Longsheng Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Meimei Zheng
- Reproductive Medicine Center of No. 960 Hospital of PLA, Jinan, China
| | - Hui Zhu
- Department of Histology and Embryology, School of Basic Medical Sciences, State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Gaggi G, Di Credico A, Barbagallo F, Ghinassi B, Di Baldassarre A. Bisphenols and perfluoroalkyls alter human stem cells integrity: A possible link with infertility. ENVIRONMENTAL RESEARCH 2023; 235:116487. [PMID: 37419196 DOI: 10.1016/j.envres.2023.116487] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/09/2023]
Abstract
Bisphenols and Perfluoroalkyls are chemical compounds widely used in industry known to be endocrine disruptors (EDs). Once ingested through contaminated aliments, they mimic the activity of endogenous hormones leading to a broad spectrum of diseases. Due to the extensive use of plastic in human life, particular attention should be paid to antenatal exposure to Bisphenols and Perfluoroalkyls since they cross the placental barrier and accumulates in developing embryo. Here we investigated the effects of Bisphenol-A (BPA), Bisphenol-S (BPS), perfluorooctane-sulfonate (PFOS) and perfluorooctanoic-acid (PFOA), alone or combined, on human-induced pluripotent stem cells (hiPSCs) that share several biological features with the stem cells of blastocysts. Our data show that these EDs affect hiPSC inducing a great mitotoxicity and dramatic changes in genes involved in the maintenance of pluripotency, germline specification, and epigenetic regulation. We also evidenced that these chemicals, when combined, may have additive, synergistic but also negative effects. All these data suggest that antenatal exposure to these EDs may affect the integrity of stem cells in the developing embryos, interfering with critical stages of early human development that might be determinant for fertility. The observation that the effects of exposure to a combination of these chemicals are not easily foreseeable further highlights the need for wider awareness of the complexity of the EDs effects on human health and of the social and economic burden attributable to these compounds.
Collapse
Affiliation(s)
- Giulia Gaggi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Medicine and Aging Sciences, "G. D'Annunzio" , University of Chieti-Pescara, 66100, Chieti, Italy; UdA -TechLab, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy
| | - Andrea Di Credico
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Medicine and Aging Sciences, "G. D'Annunzio" , University of Chieti-Pescara, 66100, Chieti, Italy; UdA -TechLab, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy
| | | | - Barbara Ghinassi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Medicine and Aging Sciences, "G. D'Annunzio" , University of Chieti-Pescara, 66100, Chieti, Italy; UdA -TechLab, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy.
| | - Angela Di Baldassarre
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100, Chieti, Italy; Department of Medicine and Aging Sciences, "G. D'Annunzio" , University of Chieti-Pescara, 66100, Chieti, Italy; UdA -TechLab, "G. D'Annunzio", University of Chieti-Pescara, 66100, Chieti, Italy
| |
Collapse
|
4
|
Di Credico A, Gaggi G, Bucci I, Ghinassi B, Di Baldassarre A. The Effects of Combined Exposure to Bisphenols and Perfluoroalkyls on Human Perinatal Stem Cells and the Potential Implications for Health Outcomes. Int J Mol Sci 2023; 24:15018. [PMID: 37834465 PMCID: PMC10573528 DOI: 10.3390/ijms241915018] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
The present study investigates the impact of two endocrine disruptors, namely Bisphenols (BPs) and Perfluoroalkyls (PFs), on human stem cells. These chemicals leach from plastic, and when ingested through contaminated food and water, they interfere with endogenous hormone signaling, causing various diseases. While the ability of BPs and PFs to cross the placental barrier and accumulate in fetal serum has been documented, the exact consequences for human development require further elucidation. The present research work explored the effects of combined exposure to BPs (BPA or BPS) and PFs (PFOS and PFOA) on human placenta (fetal membrane mesenchymal stromal cells, hFM-MSCs) and amniotic fluid (hAFSCs)-derived stem cells. The effects of the xenobiotics were assessed by analyzing cell proliferation, mitochondrial functionality, and the expression of genes involved in pluripotency and epigenetic regulation, which are crucial for early human development. Our findings demonstrate that antenatal exposure to BPs and/or PFs may alter the biological characteristics of perinatal stem cells and fetal epigenome, with potential implications for health outcomes at birth and in adulthood. Further research is necessary to comprehend the full extent of these effects and their long-term consequences.
Collapse
Affiliation(s)
- Andrea Di Credico
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy; (A.D.C.); (I.B.); (B.G.); (A.D.B.)
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- UdA TechLab Center (UdATech), 66100 Chieti, Italy
| | - Giulia Gaggi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy; (A.D.C.); (I.B.); (B.G.); (A.D.B.)
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- UdA TechLab Center (UdATech), 66100 Chieti, Italy
| | - Ines Bucci
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy; (A.D.C.); (I.B.); (B.G.); (A.D.B.)
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Barbara Ghinassi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy; (A.D.C.); (I.B.); (B.G.); (A.D.B.)
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- UdA TechLab Center (UdATech), 66100 Chieti, Italy
| | - Angela Di Baldassarre
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy; (A.D.C.); (I.B.); (B.G.); (A.D.B.)
- Department of Medicine and Aging Sciences, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- UdA TechLab Center (UdATech), 66100 Chieti, Italy
| |
Collapse
|
5
|
Chen C, Tang X, Yan S, Yang A, Xiang J, Deng Y, Yin Y, Chen B, Gu J. Comprehensive Analysis of the Transcriptome-Wide m 6A Methylome in Shaziling Pig Testicular Development. Int J Mol Sci 2023; 24:14475. [PMID: 37833923 PMCID: PMC10572705 DOI: 10.3390/ijms241914475] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
RNA N6-methyladenosine (m6A) modification is one of the principal post-transcriptional modifications and plays a dynamic role in testicular development and spermatogenesis. However, the role of m6A in porcine testis is understudied. Here, we performed a comprehensive analysis of the m6A transcriptome-wide profile in Shaziling pig testes at birth, puberty, and maturity. We analyzed the total transcriptome m6A profile and found that the m6A patterns were highly distinct in terms of the modification of the transcriptomes during porcine testis development. We found that key m6A methylated genes (AURKC, OVOL, SOX8, ACVR2A, and SPATA46) were highly enriched during spermatogenesis and identified in spermatogenesis-related KEGG pathways, including Wnt, cAMP, mTOR, AMPK, PI3K-Akt, and spliceosome. Our findings indicated that m6A methylations are involved in the complex yet well-organized post-transcriptional regulation of porcine testicular development and spermatogenesis. We found that the m6A eraser ALKBH5 negatively regulated the proliferation of immature porcine Sertoli cells. Furthermore, we proposed a novel mechanism of m6A modification during testicular development: ALKBH5 regulated the RNA methylation level and gene expression of SOX9 mRNA. In addition to serving as a potential target for improving boar reproduction, our findings contributed to the further understanding of the regulation of m6A modifications in male reproduction.
Collapse
Affiliation(s)
- Chujie Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (C.C.); (X.T.); (S.Y.); (A.Y.); (J.X.); (Y.D.); (Y.Y.)
- Hunan Provincial Key Laboratory for Genetic Improvement of Domestic Animal, Hunan Agricultural University, Changsha 410128, China
| | - Xiangwei Tang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (C.C.); (X.T.); (S.Y.); (A.Y.); (J.X.); (Y.D.); (Y.Y.)
- Hunan Provincial Key Laboratory for Genetic Improvement of Domestic Animal, Hunan Agricultural University, Changsha 410128, China
| | - Saina Yan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (C.C.); (X.T.); (S.Y.); (A.Y.); (J.X.); (Y.D.); (Y.Y.)
- Hunan Provincial Key Laboratory for Genetic Improvement of Domestic Animal, Hunan Agricultural University, Changsha 410128, China
| | - Anqi Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (C.C.); (X.T.); (S.Y.); (A.Y.); (J.X.); (Y.D.); (Y.Y.)
- Hunan Provincial Key Laboratory for Genetic Improvement of Domestic Animal, Hunan Agricultural University, Changsha 410128, China
| | - Jiaojiao Xiang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (C.C.); (X.T.); (S.Y.); (A.Y.); (J.X.); (Y.D.); (Y.Y.)
- Hunan Provincial Key Laboratory for Genetic Improvement of Domestic Animal, Hunan Agricultural University, Changsha 410128, China
| | - Yanhong Deng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (C.C.); (X.T.); (S.Y.); (A.Y.); (J.X.); (Y.D.); (Y.Y.)
- Hunan Provincial Key Laboratory for Genetic Improvement of Domestic Animal, Hunan Agricultural University, Changsha 410128, China
| | - Yulong Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (C.C.); (X.T.); (S.Y.); (A.Y.); (J.X.); (Y.D.); (Y.Y.)
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Bin Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (C.C.); (X.T.); (S.Y.); (A.Y.); (J.X.); (Y.D.); (Y.Y.)
- Hunan Provincial Key Laboratory for Genetic Improvement of Domestic Animal, Hunan Agricultural University, Changsha 410128, China
| | - Jingjing Gu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (C.C.); (X.T.); (S.Y.); (A.Y.); (J.X.); (Y.D.); (Y.Y.)
- Hunan Provincial Key Laboratory for Genetic Improvement of Domestic Animal, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
6
|
Transcriptome Analysis in High Temperature Inhibiting Spermatogonial Stem Cell Differentiation In Vitro. Reprod Sci 2022; 30:1938-1951. [DOI: 10.1007/s43032-022-01133-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022]
|
7
|
Werry N, Russell SJ, Gillis DJ, Miller S, Hickey K, Larmer S, Lohuis M, Librach C, LaMarre J. Characteristics of miRNAs Present in Bovine Sperm and Associations With Differences in Fertility. Front Endocrinol (Lausanne) 2022; 13:874371. [PMID: 35663333 PMCID: PMC9160602 DOI: 10.3389/fendo.2022.874371] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 04/14/2022] [Indexed: 12/23/2022] Open
Abstract
Small non-coding RNAs have been linked to different phenotypes in bovine sperm, however attempts to identify sperm-borne molecular biomarkers of male fertility have thus far failed to identify a robust profile of expressed miRNAs related to fertility. We hypothesized that some differences in bull fertility may be reflected in the levels of different miRNAs in sperm. To explore such differences in fertility that are not due to differences in visible metrics of sperm quality, we employed Next Generation Sequencing to compare the miRNA populations in Bos taurus sperm from bulls with comparable motility and morphology but varying Sire Conception Rates. We identified the most abundant miRNAs in both populations (miRs -34b-3p; -100-5p; -191-5p; -30d-4p; -21-5p) and evaluated differences in the overall levels and specific patterns of isomiR expression. We also explored correlations between specific pairs of miRNAs in each population and identified 10 distinct pairs of miRNAs that were positively correlated in bulls with higher fertility and negatively correlated in comparatively less fertile individuals. Furthermore, 8 additional miRNA pairs demonstrated the opposite trend; negatively correlated in high fertility animals and positively correlated in less fertile bulls. Finally, we performed pathway analysis to identify potential roles of miRNAs present in bull sperm in the regulation of specific genes that impact spermatogenesis and embryo development. Together, these results present a comprehensive picture of the bovine sperm miRNAome that suggests multiple potential roles in fertility.
Collapse
Affiliation(s)
- Nicholas Werry
- Department of Biomedical Sciences, The University of Guelph, Guelph, ON, Canada
| | | | - Daniel J. Gillis
- School of Computer Science, The University of Guelph, Guelph, ON, Canada
| | | | | | | | | | - Clifford Librach
- CReATe Fertility Centre, Toronto, ON, Canada
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Jonathan LaMarre
- Department of Biomedical Sciences, The University of Guelph, Guelph, ON, Canada
| |
Collapse
|
8
|
OVOL1 inhibits breast cancer cell invasion by enhancing the degradation of TGF-β type I receptor. Signal Transduct Target Ther 2022; 7:126. [PMID: 35484112 PMCID: PMC9050647 DOI: 10.1038/s41392-022-00944-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 02/16/2022] [Accepted: 02/24/2022] [Indexed: 11/09/2022] Open
Abstract
Ovo-like transcriptional repressor 1 (OVOL1) is a key mediator of epithelial lineage determination and mesenchymal-epithelial transition (MET). The cytokines transforming growth factor-β (TGF-β) and bone morphogenetic proteins (BMP) control the epithelial-mesenchymal plasticity (EMP) of cancer cells, but whether this occurs through interplay with OVOL1 is not known. Here, we show that OVOL1 is inversely correlated with the epithelial-mesenchymal transition (EMT) signature, and is an indicator of a favorable prognosis for breast cancer patients. OVOL1 suppresses EMT, migration, extravasation, and early metastatic events of breast cancer cells. Importantly, BMP strongly promotes the expression of OVOL1, which enhances BMP signaling in turn. This positive feedback loop is established through the inhibition of TGF-β receptor signaling by OVOL1. Mechanistically, OVOL1 interacts with and prevents the ubiquitination and degradation of SMAD family member 7 (SMAD7), which is a negative regulator of TGF-β type I receptor stability. Moreover, a small-molecule compound 6-formylindolo(3,2-b)carbazole (FICZ) was identified to activate OVOL1 expression and thereby antagonizing (at least in part) TGF-β-mediated EMT and migration in breast cancer cells. Our results uncover a novel mechanism by which OVOL1 attenuates TGF-β/SMAD signaling and maintains the epithelial identity of breast cancer cells.
Collapse
|
9
|
Adolfi MC, Du K, Kneitz S, Cabau C, Zahm M, Klopp C, Feron R, Paixão RV, Varela ES, de Almeida FL, de Oliveira MA, Nóbrega RH, Lopez-Roques C, Iampietro C, Lluch J, Kloas W, Wuertz S, Schaefer F, Stöck M, Guiguen Y, Schartl M. A duplicated copy of id2b is an unusual sex-determining candidate gene on the Y chromosome of arapaima (Arapaima gigas). Sci Rep 2021; 11:21544. [PMID: 34732792 PMCID: PMC8566520 DOI: 10.1038/s41598-021-01066-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022] Open
Abstract
Arapaima gigas is one of the largest freshwater fish species of high ecological and economic importance. Overfishing and habitat destruction are severe threats to the remaining wild populations. By incorporating a chromosomal Hi-C contact map, we improved the arapaima genome assembly to chromosome-level, revealing an unexpected high degree of chromosome rearrangements during evolution of the bonytongues (Osteoglossiformes). Combining this new assembly with pool-sequencing of male and female genomes, we identified id2bbY, a duplicated copy of the inhibitor of DNA binding 2b (id2b) gene on the Y chromosome as candidate male sex-determining gene. A PCR-test for id2bbY was developed, demonstrating that this gene is a reliable male-specific marker for genotyping. Expression analyses showed that this gene is expressed in juvenile male gonads. Its paralog, id2ba, exhibits a male-biased expression in immature gonads. Transcriptome analyses and protein structure predictions confirm id2bbY as a prime candidate for the master sex-determiner. Acting through the TGFβ signaling pathway, id2bbY from arapaima would provide the first evidence for a link of this family of transcriptional regulators to sex determination. Our study broadens our current understanding about the evolution of sex determination genetic networks and provide a tool for improving arapaima aquaculture for commercial and conservation purposes.
Collapse
Affiliation(s)
- Mateus C Adolfi
- Developmental Biochemistry, Biocenter, University of Wuerzburg, Am Hubland, 97074, Wuerzburg, Germany.
| | - Kang Du
- Developmental Biochemistry, Biocenter, University of Wuerzburg, Am Hubland, 97074, Wuerzburg, Germany
- The Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas, TX, 78666, USA
| | - Susanne Kneitz
- Biochemistry and Cell Biology, Biocenter, University of Wuerzburg, Am Hubland, 97074, Wuerzburg, Germany
| | - Cédric Cabau
- Sigenae, GenPhySE, INRAE, ENVT, Université de Toulouse, Castanet Tolosan, France
| | - Margot Zahm
- Sigenae, GenPhySE, INRAE, ENVT, Université de Toulouse, Castanet Tolosan, France
| | - Christophe Klopp
- MIAT, INRA, Université de Toulouse, Chemin de Borde Rouge, 31326, Castanet-Tolosan Cedex, France
| | - Romain Feron
- INRAE, LPGP, Rennes, France
- Department of Ecology and Evolution, University of Lausanne, and Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | | | | | | | - Marcos A de Oliveira
- Reproductive and Molecular Biology Group, Department of Morphology, Institute of Biosciences, UNESP, Botucatu, Brazil
| | - Rafael H Nóbrega
- Reproductive and Molecular Biology Group, Department of Morphology, Institute of Biosciences, UNESP, Botucatu, Brazil
| | | | | | - Jérôme Lluch
- GeT-PlaGe, INRAE, Genotoul, Castanet-Tolosan, France
| | - Werner Kloas
- Leibniz-Institute of Freshwater Ecology and Inland Fisheries, IGB, Müggelseedamm 301 & 310, 12587, Berlin, Germany
| | - Sven Wuertz
- Leibniz-Institute of Freshwater Ecology and Inland Fisheries, IGB, Müggelseedamm 301 & 310, 12587, Berlin, Germany
| | - Fabian Schaefer
- Leibniz-Institute of Freshwater Ecology and Inland Fisheries, IGB, Müggelseedamm 301 & 310, 12587, Berlin, Germany
| | - Matthias Stöck
- Leibniz-Institute of Freshwater Ecology and Inland Fisheries, IGB, Müggelseedamm 301 & 310, 12587, Berlin, Germany
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, 739-8526, Japan
| | | | - Manfred Schartl
- Developmental Biochemistry, Biocenter, University of Wuerzburg, Am Hubland, 97074, Wuerzburg, Germany
- The Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas, TX, 78666, USA
- Comprehensive Cancer Center Mainfranken, University Hospital, 97080, Würzburg, Germany
| |
Collapse
|
10
|
Chadourne M, Poumerol E, Jouneau L, Passet B, Castille J, Sellem E, Pailhoux E, Mandon-Pépin B. Structural and Functional Characterization of a Testicular Long Non-coding RNA (4930463O16Rik) Identified in the Meiotic Arrest of the Mouse Topaz1 -/- Testes. Front Cell Dev Biol 2021; 9:700290. [PMID: 34277642 PMCID: PMC8281061 DOI: 10.3389/fcell.2021.700290] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/14/2021] [Indexed: 12/23/2022] Open
Abstract
Spermatogenesis involves coordinated processes, including meiosis, to produce functional gametes. We previously reported Topaz1 as a germ cell-specific gene highly conserved in vertebrates. Topaz1 knockout males are sterile with testes that lack haploid germ cells because of meiotic arrest after prophase I. To better characterize Topaz1–/– testes, we used RNA-sequencing analyses at two different developmental stages (P16 and P18). The absence of TOPAZ1 disturbed the expression of genes involved in microtubule and/or cilium mobility, biological processes required for spermatogenesis. Moreover, a quarter of P18 dysregulated genes are long non-coding RNAs (lncRNAs), and three of them are testis-specific and located in spermatocytes, their expression starting between P11 and P15. The suppression of one of them, 4939463O16Rik, did not alter fertility although sperm parameters were disturbed and sperm concentration fell. The transcriptome of P18-4939463O16Rik–/– testes was altered and the molecular pathways affected included microtubule-based processes, the regulation of cilium movement and spermatogenesis. The absence of TOPAZ1 protein or 4930463O16Rik produced the same enrichment clusters in mutant testes despite a contrasted phenotype on male fertility. In conclusion, although Topaz1 is essential for the meiosis in male germ cells and regulate the expression of numerous lncRNAs, these studies have identified a Topaz1 regulated lncRNA (4930463O16Rik) that is key for both sperm production and motility.
Collapse
Affiliation(s)
- Manon Chadourne
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France
| | - Elodie Poumerol
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France
| | - Luc Jouneau
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France
| | - Bruno Passet
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | - Johan Castille
- INRAE, AgroParisTech, GABI, Université Paris-Saclay, Jouy-en-Josas, France
| | | | - Eric Pailhoux
- UVSQ, INRAE, BREED, Université Paris-Saclay, Jouy-en-Josas, France
| | | |
Collapse
|
11
|
Shadrina AS, Zlobin AS, Zaytseva OO, Klarić L, Sharapov SZ, D Pakhomov E, Perola M, Esko T, Hayward C, Wilson JF, Lauc G, Aulchenko YS, Tsepilov YA. Multivariate genome-wide analysis of immunoglobulin G N-glycosylation identifies new loci pleiotropic with immune function. Hum Mol Genet 2021; 30:1259-1270. [PMID: 33710309 DOI: 10.1093/hmg/ddab072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/19/2021] [Accepted: 03/05/2021] [Indexed: 12/15/2022] Open
Abstract
The N-glycosylation of immunoglobulin G (IgG) affects its structure and function. It has been demonstrated that IgG N-glycosylation patterns are inherited as complex quantitative traits. Genome-wide association studies identified loci harboring genes encoding enzymes directly involved in protein glycosylation as well as loci likely to be involved in regulation of glycosylation biochemical pathways. Many of these loci could be linked to immune functions and risk of inflammatory and autoimmune diseases. The aim of the present study was to discover and replicate new loci associated with IgG N-glycosylation and to investigate possible pleiotropic effects of these loci onto immune function and the risk of inflammatory and autoimmune diseases. We conducted a multivariate genome-wide association analysis of 23 IgG N-glycosylation traits measured in 8090 individuals of European ancestry. The discovery stage was followed up by replication in 3147 people and in silico functional analysis. Our study increased the total number of replicated loci from 22 to 29. For the discovered loci, we suggest a number of genes potentially involved in the control of IgG N-glycosylation. Among the new loci, two (near RNF168 and TNFRSF13B) were previously implicated in rare immune deficiencies and were associated with levels of circulating immunoglobulins. For one new locus (near AP5B1/OVOL1), we demonstrated a potential pleiotropic effect on the risk of asthma. Our findings underline an important link between IgG N-glycosylation and immune function and provide new clues to understanding their interplay.
Collapse
Affiliation(s)
- Alexandra S Shadrina
- Laboratory of Glycogenomics, Institute of Cytology and Genetics, Novosibirsk 630090, Russia
| | - Alexander S Zlobin
- Laboratory of Glycogenomics, Institute of Cytology and Genetics, Novosibirsk 630090, Russia
| | - Olga O Zaytseva
- Genos Glycoscience Research Laboratory, Zagreb 10000, Croatia
| | - Lucija Klarić
- Genos Glycoscience Research Laboratory, Zagreb 10000, Croatia.,MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Sodbo Z Sharapov
- Laboratory of Glycogenomics, Institute of Cytology and Genetics, Novosibirsk 630090, Russia
| | - Eugene D Pakhomov
- Laboratory of Glycogenomics, Institute of Cytology and Genetics, Novosibirsk 630090, Russia
| | - Marcus Perola
- Genomics and Biomarkers Unit, Department of Health, National Institute for Health and Welfare (THL), Helsinki, Finland
| | - Tonu Esko
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Caroline Hayward
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - James F Wilson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.,Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh EH8 9AG, Scotland
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Zagreb 10000, Croatia
| | - Yurii S Aulchenko
- Laboratory of Glycogenomics, Institute of Cytology and Genetics, Novosibirsk 630090, Russia.,PolyOmica, 's-Hertogenbosch 5237 PA, The Netherlands
| | - Yakov A Tsepilov
- Laboratory of Glycogenomics, Institute of Cytology and Genetics, Novosibirsk 630090, Russia.,Laboratory of Theoretical and Applied Functional Genomics, Novosibirsk State University, Novosibirsk 630090, Russia
| |
Collapse
|
12
|
Saxena K, Srikrishnan S, Celia-Terrassa T, Jolly MK. OVOL1/2: Drivers of Epithelial Differentiation in Development, Disease, and Reprogramming. Cells Tissues Organs 2020; 211:183-192. [PMID: 32932250 DOI: 10.1159/000511383] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 08/26/2020] [Indexed: 11/19/2022] Open
Abstract
OVOL proteins (OVOL1 and OVOL2), vertebrate homologs of Drosophila OVO, are critical regulators of epithelial lineage determination and differentiation during embryonic development in tissues such as kidney, skin, mammary epithelia, and testis. OVOL can inhibit epithelial-mesenchymal transition and/or can promote mesenchymal-epithelial transition. Moreover, they can regulate the stemness of cancer cells, thus playing an important role during cancer cell metastasis. Due to their central role in differentiation and maintenance of epithelial lineage, OVOL overexpression has been shown to be capable of reprogramming fibroblasts to epithelial cells. Here, we review the roles of OVOL-mediated epithelial differentiation across multiple contexts, including embryonic development, cancer progression, and cellular reprogramming.
Collapse
Affiliation(s)
- Kritika Saxena
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | | | - Toni Celia-Terrassa
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Mohit Kumar Jolly
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India,
| |
Collapse
|
13
|
Jeyarajah MJ, Jaju Bhattad G, Hillier DM, Renaud SJ. The Transcription Factor OVOL2 Represses ID2 and Drives Differentiation of Trophoblast Stem Cells and Placental Development in Mice. Cells 2020; 9:E840. [PMID: 32244352 PMCID: PMC7226816 DOI: 10.3390/cells9040840] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 12/14/2022] Open
Abstract
Trophoblasts are the first cell type to be specified during embryogenesis, and they are essential for placental morphogenesis and function. Trophoblast stem (TS) cells are the progenitor cells for all trophoblast lineages; control of TS cell differentiation into distinct trophoblast subtypes is not well understood. Mice lacking the transcription factor OVO-like 2 (OVOL2) fail to produce a functioning placenta, and die around embryonic day 10.5, suggesting that OVOL2 may be critical for trophoblast development. Therefore, our objective was to determine the role of OVOL2 in mouse TS cell fate. We found that OVOL2 was highly expressed in mouse placenta and differentiating TS cells. Placentas and TS cells lacking OVOL2 showed poor trophoblast differentiation potential, including increased expression of stem-state associated genes (Eomes, Esrrb, Id2) and decreased levels of differentiation-associated transcripts (Gcm1, Tpbpa, Prl3b1, Syna). Ectopic OVOL2 expression in TS cells elicited precocious differentiation. OVOL2 bound proximate to the gene encoding inhibitor of differentiation 2 (ID2), a dominant negative helix-loop-helix protein, and directly repressed its activity. Overexpression of ID2 was sufficient to reinforce the TS cell stem state. Our findings reveal a critical role of OVOL2 as a regulator of TS cell differentiation and placental development, in-part by coordinating repression of ID2.
Collapse
Affiliation(s)
- Mariyan J. Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A5C1, Canada; (M.J.J.)
| | - Gargi Jaju Bhattad
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A5C1, Canada; (M.J.J.)
| | - Dendra M. Hillier
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A5C1, Canada; (M.J.J.)
| | - Stephen J. Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A5C1, Canada; (M.J.J.)
- Children’s Health Research Institute, London, ON N6C2V5, Canada
- Lawson Health Research Institute, London, ON N6C2R5, Canada
| |
Collapse
|
14
|
Loss of Cx43 in Murine Sertoli Cells Leads to Altered Prepubertal Sertoli Cell Maturation and Impairment of the Mitosis-Meiosis Switch. Cells 2020; 9:cells9030676. [PMID: 32164318 PMCID: PMC7140672 DOI: 10.3390/cells9030676] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
Male factor infertility is a problem in today’s society but many underlying causes are still unknown. The generation of a conditional Sertoli cell (SC)-specific connexin 43 (Cx43) knockout mouse line (SCCx43KO) has provided a translational model. Expression of the gap junction protein Cx43 between adjacent SCs as well as between SCs and germ cells (GCs) is known to be essential for the initiation and maintenance of spermatogenesis in different species and men. Adult SCCx43KO males show altered spermatogenesis and are infertile. Thus, the present study aims to identify molecular mechanisms leading to testicular alterations in prepubertal SCCx43KO mice. Transcriptome analysis of 8-, 10- and 12-day-old mice was performed by next-generation sequencing (NGS). Additionally, candidate genes were examined by qRT-PCR and immunohistochemistry. NGS revealed many significantly differentially expressed genes in the SCCx43KO mice. For example, GC-specific genes were mostly downregulated and found to be involved in meiosis and spermatogonial differentiation (e.g., Dmrtb1, Sohlh1). In contrast, SC-specific genes implicated in SC maturation and proliferation were mostly upregulated (e.g., Amh, Fshr). In conclusion, Cx43 in SCs appears to be required for normal progression of the first wave of spermatogenesis, especially for the mitosis-meiosis switch, and also for the regulation of prepubertal SC maturation.
Collapse
|
15
|
Zhang J, Dong J, Qin W, Cao C, Wen Y, Tang Y, Yuan S. Ovol2, a zinc finger transcription factor, is dispensable for spermatogenesis in mice. Reprod Biol Endocrinol 2019; 17:98. [PMID: 31759386 PMCID: PMC6875160 DOI: 10.1186/s12958-019-0542-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/12/2019] [Indexed: 11/23/2022] Open
Abstract
Ovol2, a mouse homolog of Drosophila ovo, was identified as a zinc finger transcription factor predominantly expressed in testis. However, the function of Ovol2 in postnatal male germ cell development remains enigmatic. Here, we firstly examined the mRNA and protein levels of Ovol2 in developing mouse testes by RT-qPCR and western blot and found that both mRNA and protein of Ovol2 are continually expressed in postnatal developing testes from postnatal day 0 (P0) testes to adult testes (P56) and exhibits its higher level at adult testis. Further testicular immuno-staining revealed that OVOL2 is highly expressed in the spermatogonia, spermatocytes and round spermatids. Interestingly, our conditional ovol2 knockout mouse model show that loss of ovol2 in embryonic germ cells does not affect fecundity in mice. Our data also show that Ovol1 may have compensated for the loss of Ovol2 functions in germ cells. Overall, our data indicate that ovol2 is dispensable for germ cell development and spermatogenesis.
Collapse
Affiliation(s)
- Jin Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- College of Animal Science and Technology, Northwest A&F University, Yangling, People's Republic of China
| | - Juan Dong
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Weibing Qin
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, Guangzhou, People's Republic of China
| | - Congcong Cao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yujiao Wen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yunge Tang
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, Guangzhou, People's Republic of China.
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, People's Republic of China.
| |
Collapse
|
16
|
Zhang L, Huang Y, Wang M, Guo Y, Liang J, Yang X, Qi W, Wu Y, Si J, Zhu S, Li Z, Li R, Shi C, Wang S, Zhang Q, Tang Z, Wang L, Li K, Fei JF, Lan G. Development and Genome Sequencing of a Laboratory-Inbred Miniature Pig Facilitates Study of Human Diabetic Disease. iScience 2019; 19:162-176. [PMID: 31376679 PMCID: PMC6677790 DOI: 10.1016/j.isci.2019.07.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/11/2019] [Accepted: 07/13/2019] [Indexed: 01/10/2023] Open
Abstract
Pig has been proved to be a valuable large animal model used for research on diabetic disease. However, their translational value is limited given their distinct anatomy and physiology. For the last 30 years, we have been developing a laboratory Asian miniature pig inbred line (Bama miniature pig [BM]) from the primitive Bama xiang pig via long-term selective inbreeding. Here, we assembled a BM reference genome at full chromosome-scale resolution with a total length of 2.49 Gb. Comparative and evolutionary genomic analyses identified numerous variations between the BM and commercial pig (Duroc), particularly those in the genetic loci associated with the features advantageous to diabetes studies. Resequencing analyses revealed many differentiated gene loci associated with inbreeding and other selective forces. These together with transcriptome analyses of diabetic pig models provide a comprehensive genetic basis for resistance to diabetogenic environment, especially related to energy metabolism.
Collapse
Affiliation(s)
- Li Zhang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Yuemeng Huang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Meng Wang
- Novogene Bioinformatics Institute, Beijing 100083, China
| | - Yafen Guo
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Jing Liang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China.
| | - Xiurong Yang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Wenjing Qi
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Yanjun Wu
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Jinglei Si
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Siran Zhu
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Zhe Li
- Novogene Bioinformatics Institute, Beijing 100083, China
| | - Ruiqiang Li
- Novogene Bioinformatics Institute, Beijing 100083, China
| | - Chao Shi
- Shandong Provincial Key Laboratory of Biochemical Engineering, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| | - Shuo Wang
- Shandong Provincial Key Laboratory of Biochemical Engineering, College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Qunjie Zhang
- Institution of Genomics and Bioinformatics, South China Agricultural University, Guangzhou 510642, China
| | - Zhonglin Tang
- Research Centre for Animal Genome, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China; Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Lixian Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Kui Li
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ji-Feng Fei
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou 510631, China
| | - Ganqiu Lan
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China.
| |
Collapse
|
17
|
Conserved role of Ovo in germline development in mouse and Drosophila. Sci Rep 2017; 7:40056. [PMID: 28059165 PMCID: PMC5216385 DOI: 10.1038/srep40056] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/30/2016] [Indexed: 12/31/2022] Open
Abstract
Ovo, which encodes a transcription factor with Zn-finger domains, is evolutionarily conserved among animals. In Drosophila, in addition to its zygotic function for egg production, maternal ovo activity is required in primordial germ cells (PGCs) for expression of germline genes such as vasa and nanos. In this study, we found that maternal Ovo accumulates in PGC nuclei during embryogenesis. In these cells, ovo serves a dual function: activation of genes expressed predominantly in PGCs, and conversely suppression of somatic genes. Reduction of ovo activity in PGCs makes them unable to develop normally into germ cells of both sexes. In mice, knockout of the ovo ortholog, Ovol2, which is expressed in PGCs, decreases the number of PGCs during early embryogenesis. These data strongly suggest that ovo acts as part of an evolutionarily conserved mechanism that regulates germline development in animals.
Collapse
|
18
|
Cai X, Yu S, Mipam T, Yang F, Zhao W, Liu W, Cao S, Shen L, Zhao F, Sun L, Xu C, Wu S. Comparative analysis of testis transcriptomes associated with male infertility in cattleyak. Theriogenology 2017; 88:28-42. [DOI: 10.1016/j.theriogenology.2016.09.047] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 09/18/2016] [Accepted: 09/24/2016] [Indexed: 01/29/2023]
|
19
|
Ovol2 gene inhibits the Epithelial-to-Mesenchymal Transition in lung adenocarcinoma by transcriptionally repressing Twist1. Gene 2016; 600:1-8. [PMID: 27884772 DOI: 10.1016/j.gene.2016.11.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/07/2016] [Accepted: 11/17/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Associated with recent achievements in therapy for advanced lung adenocarcinoma, there will still be an unmet medical need for effective treatment of stage IIIb/IV, and the prognosis of lung cancer is not optimistic till now. OBJECTIVE In order to obtain some essential evidences for a potential targeted therapy in lung adenocarcinoma, the effects of Ovol2 gene on Epithelial-to-Mesenchymal Transition (EMT) was observed and the probable mechanisms were analyzed. METHODS Ovol2 expression was previously evaluated by immunochemistry in lung adenocarcinoma tissue, and Ovol2 was overexpressed by lentivirus infection in A549 cells. Subsequently, the migration and invasion ability of A549 cells was tested by Transwell and Wound healing experiments. The mRNA level of genes correlated to EMT was detected by Real-time PCR, and the expression of reasonable makers was probed by Western Blot. Finally, rescue experiment, Luciferase assay, and chromatin immunoprecipitation assay were performed to explore the probable mechanisms. RESULTS After treated with Ovol2 overexpression, the expression level of E-cadherin was increased, while the expression level of Vimentin and Twist1 was declined not only in the mRNA level but also in the protein level. Moreover, we found that Ovol2 represses transcription of Twist1 by binding to its promoter directly. Wound healing and Transwell assays indicate that the migration and invasion ability were downregulated by Ovol2 in A549 cells. CONCLUSION Ovol2 can suppress migration and invasion ability of A549 cells, and prevent EMT by inhibition of Twist1 transcription directly.
Collapse
|
20
|
Brouard V, Guénon I, Bouraima-Lelong H, Delalande C. Differential effects of bisphenol A and estradiol on rat spermatogenesis’ establishment. Reprod Toxicol 2016; 63:49-61. [DOI: 10.1016/j.reprotox.2016.05.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/22/2016] [Accepted: 05/07/2016] [Indexed: 11/26/2022]
|
21
|
Taniguchi H, Katano T, Nishida K, Yao I, Morimoto Y, Matsuda T, Ito S. Expression of hOvol2 in the XY body of human spermatocytes. Andrologia 2016; 49. [DOI: 10.1111/and.12599] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2016] [Indexed: 11/30/2022] Open
Affiliation(s)
- H. Taniguchi
- Department of Medical Chemistry; Kansai Medical University; Hirakata Osaka Japan
- Department of Urology and Andrology; Kansai Medical University; Hirakata Osaka Japan
| | - T. Katano
- Department of Medical Chemistry; Kansai Medical University; Hirakata Osaka Japan
| | - K. Nishida
- Department of Medical Chemistry; Kansai Medical University; Hirakata Osaka Japan
| | - I. Yao
- Medical Photonics Research Center; Hamamatsu University School of Medicine; Hamamatsu Shizuoka Japan
| | | | - T. Matsuda
- Department of Urology and Andrology; Kansai Medical University; Hirakata Osaka Japan
| | - S. Ito
- Department of Medical Chemistry; Kansai Medical University; Hirakata Osaka Japan
| |
Collapse
|
22
|
Browne JA, Yang R, Leir SH, Eggener SE, Harris A. Expression profiles of human epididymis epithelial cells reveal the functional diversity of caput, corpus and cauda regions. Mol Hum Reprod 2015; 22:69-82. [PMID: 26612782 DOI: 10.1093/molehr/gav066] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/20/2015] [Indexed: 01/01/2023] Open
Abstract
STUDY HYPOTHESIS Region-specific transcriptional profiling of tissues and cultured epithelial cells from the human epididymis will predict functional specialization along the duct. STUDY FINDING We identified the molecular signature driving functions of the caput, corpus and cauda epithelium, and determined how these differ to establish the regional differentiation of the organ. WHAT IS KNOWN ALREADY The epithelium lining the human male genital ducts has a critical role in fertility. In particular, it controls the luminal environment in the epididymis, which is required for normal sperm maturation and reproductive competence. Studies in many animal species have largely informed our understanding of the molecular basis of epididymis function. However, there are substantial differences between species. STUDY DESIGN, SAMPLES/MATERIALS, METHODS Using RNA sequencing on biological replicates, we described gene expression profiles for tissue from each region of the epididymis and cultured epithelial cells derived from these regions. Bioinformatic tools were then utilized to identify differentially expressed genes (DEGs) between tissues and cells from the caput, corpus and cauda. MAIN RESULTS AND THE ROLE OF CHANCE The data showed that the caput is functionally divergent from the corpus and cauda, which have very similar transcriptomes. Interrogation of DEGs using gene ontology process enrichment analyses showed that processes of ion transport, response to hormone stimulus and urogenital tract development are more evident in the caput, while defense response processes are more important in the corpus/cauda. Consistent with these regional differences in epididymis function, we observed differential expression of transcription factors in the caput and corpus/cauda. LIMITATIONS, REASONS FOR CAUTION Cultured caput, corpus and cauda cells may not faithfully represent the same cells in the intact organ, due to loss of hormonal signals from the testis and communication from other cell types. WIDER IMPLICATIONS OF THE FINDINGS Our data provide a molecular characterization that will facilitate advances in understanding human epididymis epithelium biology in health and disease. They may also reveal the mechanisms coordinating epididymis luminal environment and sperm maturation. LARGE SCALE DATA Data deposited at http://www.ncbi.nlm.nih.gov/geo/GSE72986. STUDY FUNDING AND COMPETING INTERESTS This work was supported by the National Institutes of Health: R01HD068901 (PI: A.H.). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- James A Browne
- Human Molecular Genetics Program, Lurie Children's Research Center, 2430 North Halsted Street, Box 211, Chicago, IL 60614, USA Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rui Yang
- Human Molecular Genetics Program, Lurie Children's Research Center, 2430 North Halsted Street, Box 211, Chicago, IL 60614, USA Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shih-Hsing Leir
- Human Molecular Genetics Program, Lurie Children's Research Center, 2430 North Halsted Street, Box 211, Chicago, IL 60614, USA Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Scott E Eggener
- Section of Urology, University of Chicago Medical Center, Chicago, IL, USA
| | - Ann Harris
- Human Molecular Genetics Program, Lurie Children's Research Center, 2430 North Halsted Street, Box 211, Chicago, IL 60614, USA Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
23
|
OVO-like 1 regulates progenitor cell fate in human trophoblast development. Proc Natl Acad Sci U S A 2015; 112:E6175-84. [PMID: 26504231 DOI: 10.1073/pnas.1507397112] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Epithelial barrier integrity is dependent on progenitor cells that either divide to replenish themselves or differentiate into a specialized epithelium. This paradigm exists in human placenta, where cytotrophoblast cells either propagate or undergo a unique differentiation program: fusion into an overlying syncytiotrophoblast. Syncytiotrophoblast is the primary barrier regulating the exchange of nutrients and gases between maternal and fetal blood and is the principal site for synthesizing hormones vital for human pregnancy. How trophoblast cells regulate their differentiation into a syncytium is not well understood. In this study, we show that the transcription factor OVO-like 1 (OVOL1), a homolog of Drosophila ovo, regulates the transition from progenitor to differentiated trophoblast cells. OVOL1 is expressed in human placenta and was robustly induced following stimulation of trophoblast differentiation. Disruption of OVOL1 abrogated cytotrophoblast fusion and inhibited the expression of a broad set of genes required for trophoblast cell fusion and hormonogenesis. OVOL1 was required to suppress genes that maintain cytotrophoblast cells in a progenitor state, including MYC, ID1, TP63, and ASCL2, and bound specifically to regions upstream of each of these genes. Our results reveal an important function of OVOL1 as a regulator of trophoblast progenitor cell fate during human trophoblast development.
Collapse
|
24
|
Luangpraseuth-Prosper A, Lesueur E, Jouneau L, Pailhoux E, Cotinot C, Mandon-Pépin B. TOPAZ1, a germ cell specific factor, is essential for male meiotic progression. Dev Biol 2015; 406:158-71. [PMID: 26358182 DOI: 10.1016/j.ydbio.2015.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/01/2015] [Accepted: 09/04/2015] [Indexed: 11/19/2022]
Abstract
Topaz1 (Testis and Ovary-specific PAZ domain gene 1) is a germ cell specific gene highly conserved in vertebrates. The putative protein TOPAZ1 contains a PAZ domain, specifically found in PIWI, Argonaute and Zwille proteins. Consequently, Topaz1 is supposed to have a role during gametogenesis and may be involved in the piRNA pathway and contribute to silencing of transposable elements and maintenance of genome integrity. Here we report Topaz1 inactivation in mouse. Female fertility was not affected, but male sterility appeared exclusively in homozygous mutants in accordance with the high expression of Topaz1 in male germ cells. Pachytene Topaz1--deficient spermatocytes progress through meiosis without either derepression of retrotransposons or MSCI dysfunction, but become arrested before the post-meiotic round spermatid stage with extensive apoptosis. Consequently, an absence of spermatids and spermatozoa was observed in Topaz1(-/-) testis. Histological analysis also revealed that disturbances of spermatogenesis take place between post natal days 15 and 20, during the first wave of male meiosis and before the generation of haploid germ cells. Transcriptomic analysis at these two stages showed that TOPAZ1 influences the expression of one hundred transcripts, most of which are up-regulated in mutant testis at post natal day 20. Our results also showed that 10% of these transcripts are long non-coding RNA. This suggests that a highly regulated balance of lncRNAs seems to be essential during spermatogenesis for induction of appropriate male gamete production.
Collapse
Affiliation(s)
| | - Elodie Lesueur
- INRA, UMR 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France.
| | - Luc Jouneau
- INRA, UMR 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France.
| | - Eric Pailhoux
- INRA, UMR 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France.
| | - Corinne Cotinot
- INRA, UMR 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France.
| | - Béatrice Mandon-Pépin
- INRA, UMR 1198 Biologie du Développement et Reproduction, F-78350 Jouy-en-Josas, France.
| |
Collapse
|
25
|
Kang Z, Li Q, Fu P, Yan S, Guan M, Xu J, Xu F. Correlation of KIF3A and OVOL1, but not ACTL9, with atopic dermatitis in Chinese pediatric patients. Gene 2015; 571:249-51. [PMID: 26127003 DOI: 10.1016/j.gene.2015.06.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 04/17/2015] [Accepted: 06/25/2015] [Indexed: 10/23/2022]
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease in Chinese pediatric patients. To date, the genetic susceptibility to AD in this population has not been fully clarified. Three single nucleotide polymorphisms have previously been associated with AD in Europeans, rs2897442 (KIF3A), rs479844 (OVOL1) and rs2164983 (ACTL9). To verify the correlation between AD and these three SNPs in the Chinese pediatric population, we conducted a case-control study including 235 pediatric patients with AD and 200 health controls. We confirmed the correlation between rs2897442 and rs479844 and AD in this population at both the genotype and allele levels. Statistical analysis showed that the C allele of rs2897442 is associated with an increased risk of developing AD, while the A allele of rs479844 is associated with a reduced risk. No correlation between rs2164983 and AD was identified. Our study indicates that KIF3A and OVOL1 are involved in the development of AD in the Chinese pediatric population.
Collapse
Affiliation(s)
- Zhihua Kang
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200040, PR China
| | - Qiao Li
- Department of Dermatology, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200040, PR China
| | - Pan Fu
- Department of Microbiology, Children's Hospital, Shanghai Medical School, Fudan University, Shanghai 200040, PR China
| | - Shuxian Yan
- Department of Dermatology, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200040, PR China
| | - Ming Guan
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200040, PR China; Central Laboratory, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200040, PR China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200040, PR China.
| | - Feng Xu
- Department of Dermatology, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai 200040, PR China.
| |
Collapse
|
26
|
Watanabe K, Villarreal-Ponce A, Sun P, Salmans ML, Fallahi M, Andersen B, Dai X. Mammary morphogenesis and regeneration require the inhibition of EMT at terminal end buds by Ovol2 transcriptional repressor. Dev Cell 2014; 29:59-74. [PMID: 24735879 DOI: 10.1016/j.devcel.2014.03.006] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 01/17/2014] [Accepted: 03/12/2014] [Indexed: 01/19/2023]
Abstract
Epithelial cells possess remarkable plasticity, having the ability to become mesenchymal cells through alterations in adhesion and motility (epithelial-to-mesenchymal transition [EMT]). However, how epithelial plasticity is kept in check in epithelial cells during tissue development and regeneration remains to be fully understood. Here we show that restricting the EMT of mammary epithelial cells by transcription factor Ovol2 is required for proper morphogenesis and regeneration. Deletion of Ovol2 blocks mammary ductal morphogenesis, depletes stem and progenitor cell reservoirs, and leads epithelial cells to undergo EMT in vivo to become nonepithelial cell types. Ovol2 directly represses myriad EMT inducers, and its absence switches response to TGF-β from growth arrest to EMT. Furthermore, forced expression of the repressor isoform of Ovol2 is able to reprogram metastatic breast cancer cells from a mesenchymal to an epithelial state. Our findings underscore the critical importance of exquisitely regulating epithelial plasticity in development and cancer.
Collapse
Affiliation(s)
- Kazuhide Watanabe
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Alvaro Villarreal-Ponce
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Peng Sun
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Michael L Salmans
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Magid Fallahi
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Bogi Andersen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Department of Medicine, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
27
|
Lee B, Villarreal-Ponce A, Fallahi M, Ovadia J, Sun P, Yu QC, Ito S, Sinha S, Nie Q, Dai X. Transcriptional mechanisms link epithelial plasticity to adhesion and differentiation of epidermal progenitor cells. Dev Cell 2014; 29:47-58. [PMID: 24735878 DOI: 10.1016/j.devcel.2014.03.005] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 01/17/2014] [Accepted: 03/12/2014] [Indexed: 01/05/2023]
Abstract
During epithelial tissue morphogenesis, developmental progenitor cells undergo dynamic adhesive and cytoskeletal remodeling to trigger proliferation and migration. Transcriptional mechanisms that restrict such a mild form of epithelial plasticity to maintain lineage-restricted differentiation in committed epithelial tissues are poorly understood. Here, we report that simultaneous ablation of transcriptional repressor-encoding Ovol1 and Ovol2 results in expansion and blocked terminal differentiation of embryonic epidermal progenitor cells. Conversely, mice overexpressing Ovol2 in their skin epithelia exhibit precocious differentiation accompanied by smaller progenitor cell compartments. We show that Ovol1/Ovol2-deficient epidermal cells fail to undertake α-catenin-driven actin cytoskeletal reorganization and adhesive maturation and exhibit changes that resemble epithelial-to-mesenchymal transition (EMT). Remarkably, these alterations and defective terminal differentiation are reversed upon depletion of EMT-promoting transcriptional factor Zeb1. Collectively, our findings reveal Ovol-Zeb1-α-catenin sequential repression and highlight Ovol1 and Ovol2 as gatekeepers of epithelial adhesion and differentiation by inhibiting progenitor-like traits and epithelial plasticity.
Collapse
Affiliation(s)
- Briana Lee
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine CA 92697, USA
| | - Alvaro Villarreal-Ponce
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine CA 92697, USA
| | - Magid Fallahi
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine CA 92697, USA
| | - Jeremy Ovadia
- Department of Mathematics, University of California, Irvine, Irvine CA 92697, USA
| | - Peng Sun
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine CA 92697, USA
| | - Qian-Chun Yu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seiji Ito
- Department of Medical Chemistry, Kansai Medical University, Moriguchi 570-8506, Japan
| | - Satrajit Sinha
- Department of Biochemistry, State University of New York, Buffalo, NY 14260, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine CA 92697, USA
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine CA 92697, USA.
| |
Collapse
|
28
|
Porto-Neto LR, Lee SH, Sonstegard TS, Van Tassell CP, Lee HK, Gibson JP, Gondro C. Genome-wide detection of signatures of selection in Korean Hanwoo cattle. Anim Genet 2014; 45:180-90. [PMID: 24494817 DOI: 10.1111/age.12119] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2013] [Indexed: 02/02/2023]
Abstract
The Korean Hanwoo cattle have been intensively selected for production traits, especially high intramuscular fat content. It is believed that ancient crossings between different breeds contributed to forming the Hanwoo, but little is known about the genomic differences and similarities between other cattle breeds and the Hanwoo. In this work, cattle breeds were grouped by origin into four types and used for comparisons: the Europeans (represented by six breeds), zebu (Nelore), African taurine (N'Dama) and Hanwoo. All animals had genotypes for around 680 000 SNPs after quality control of genotypes. Average heterozygosity was lower in Nelore and N'Dama (0.22 and 0.21 respectively) than in Europeans (0.26-0.31, with Shorthorn as outlier at 0.24) and Hanwoo (0.29). Pairwise FST analyses demonstrated that Hanwoo are more related to European cattle than to Nelore, with N'Dama in an intermediate position. This finding was corroborated by principal components and unsupervised hierarchical clustering. Using genome-wide smoothed FST , 55 genomic regions potentially under positive selection in Hanwoo were identified. Among these, 29 were regions also detected in previous studies. Twenty-four regions were exclusive to Hanwoo, and a number of other regions were shared with one or two of the other groups. These regions overlap a number of genes that are related to immune, reproduction and fatty acid metabolism pathways. Further analyses are needed to better characterize the ancestry of the Hanwoo cattle and to define the genes responsible to the identified selection peaks.
Collapse
Affiliation(s)
- L R Porto-Neto
- School of Environmental and Rural Science, University of New England, Armidale, NSW, 2351, Australia; School of Veterinary Science, The University of Queensland, Gatton, QLD, 4345, Australia
| | | | | | | | | | | | | |
Collapse
|
29
|
Bonnet A, Cabau C, Bouchez O, Sarry J, Marsaud N, Foissac S, Woloszyn F, Mulsant P, Mandon-Pepin B. An overview of gene expression dynamics during early ovarian folliculogenesis: specificity of follicular compartments and bi-directional dialog. BMC Genomics 2013; 14:904. [PMID: 24350644 PMCID: PMC3890531 DOI: 10.1186/1471-2164-14-904] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 12/10/2013] [Indexed: 12/03/2022] Open
Abstract
Background Successful early folliculogenesis is crucial for female reproductive function. It requires appropriate gene specific expression of the different types of ovarian cells at different developmental stages. To date, most gene expression studies on the ovary were conducted in rodents and did not distinguish the type of cell. In mono-ovulating species, few studies have addressed gene expression profiles and mainly concerned human oocytes. Results We used a laser capture microdissection method combined with RNA-seq technology to explore the transcriptome in oocytes and granulosa cells (GCs) during development of the sheep ovarian follicle. We first documented the expression profile of 15 349 genes, then focused on the 5 129 genes showing differential expression between oocytes and GCs. Enriched functional categories such as oocyte meiotic arrest and GC steroid synthesis reflect two distinct cell fates. We identified the implication of GC signal transduction pathways such as SHH, WNT and RHO GTPase. In addition, signaling pathways (VEGF, NOTCH, IGF1, etc.) and GC transzonal projections suggest the existence of complex cell-cell interactions. Finally, we highlighted several transcription regulators and specifically expressed genes that likely play an important role in early folliculogenesis. Conclusions To our knowledge, this is the first comprehensive exploration of transcriptomes derived from in vivo oocytes and GCs at key stages in early follicular development in sheep. Collectively, our data advance our understanding of early folliculogenesis in mono-ovulating species and will be a valuable resource for unraveling human ovarian dysfunction such as premature ovarian failure (POF).
Collapse
Affiliation(s)
- Agnes Bonnet
- INRA, UMR444 Génétique Cellulaire, Auzeville, BP52627, F-31326, Castanet-Tolosan, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
RanBPM is a multimodular scaffold protein that interacts with a great variety of molecules including nuclear, cytoplasmic, and membrane proteins. By building multiprotein complexes, RanBPM is thought to regulate various signaling pathways, especially in the immune and nervous system. However, the diversity of these interactions does not facilitate the identification of its precise mechanism of action, and therefore the physiological role of RanBPM still remains unclear. Recently, RanBPM has been shown to be critical for the fertility of both genders in mouse. Although mechanistically it is still unclear how RanBPM affects gametogenesis, the data collected so far suggest that it is a key player in this process. Here, we examine the RanBPM sterility phenotype in the context of other genetic mutations affecting mouse gametogenesis to investigate whether this scaffold protein affects the function of other known proteins whose deficiency results in similar sterility phenotypes.
Collapse
Affiliation(s)
- Sandrine Puverel
- Neural Development Section, Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, Maryland, USA.
| | | |
Collapse
|
31
|
Kumar A, Bhandari A, Sinha R, Sardar P, Sushma M, Goyal P, Goswami C, Grapputo A. Molecular phylogeny of OVOL genes illustrates a conserved C2H2 zinc finger domain coupled by hypervariable unstructured regions. PLoS One 2012; 7:e39399. [PMID: 22737237 PMCID: PMC3380836 DOI: 10.1371/journal.pone.0039399] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2011] [Accepted: 05/23/2012] [Indexed: 11/19/2022] Open
Abstract
OVO-like proteins (OVOL) are members of the zinc finger protein family and serve as transcription factors to regulate gene expression in various differentiation processes. Recent studies have shown that OVOL genes are involved in epithelial development and differentiation in a wide variety of organisms; yet there is a lack of comprehensive studies that describe OVOL proteins from an evolutionary perspective. Using comparative genomic analysis, we traced three different OVOL genes (OVOL1-3) in vertebrates. One gene, OVOL3, was duplicated during a whole-genome-duplication event in fish, but only the copy (OVOL3b) was retained. From early-branching metazoa to humans, we found that a core domain, comprising a tetrad of C2H2 zinc fingers, is conserved. By domain comparison of the OVOL proteins, we found that they evolved in different metazoan lineages by attaching intrinsically-disordered (ID) segments of N/C-terminal extensions of 100 to 1000 amino acids to this conserved core. These ID regions originated independently across different animal lineages giving rise to different types of OVOL genes over the course of metazoan evolution. We illustrated the molecular evolution of metazoan OVOL genes over a period of 700 million years (MY). This study both extends our current understanding of the structure/function relationship of metazoan OVOL genes, and assembles a good platform for further characterization of OVOL genes from diverged organisms.
Collapse
Affiliation(s)
- Abhishek Kumar
- Department of Biology, University of Padua, Padova, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Giese S, Hossain H, Markmann M, Chakraborty T, Tchatalbachev S, Guillou F, Bergmann M, Failing K, Weider K, Brehm R. Sertoli-cell-specific knockout of connexin 43 leads to multiple alterations in testicular gene expression in prepubertal mice. Dis Model Mech 2012; 5:895-913. [PMID: 22699423 PMCID: PMC3484871 DOI: 10.1242/dmm.008649] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A significant decline in human male reproductive function has been reported for the past 20 years but the molecular mechanisms remain poorly understood. However, recent studies showed that the gap junction protein connexin-43 (CX43; also known as GJA1) might be involved. CX43 is the predominant testicular connexin (CX) in most species, including in humans. Alterations of its expression are associated with different forms of spermatogenic disorders and infertility. Men with impaired spermatogenesis often exhibit a reduction or loss of CX43 expression in germ cells (GCs) and Sertoli cells (SCs). Adult male transgenic mice with a conditional knockout (KO) of the Gja1 gene [referred to here as connexin-43 (Cx43)] in SCs (SCCx43KO) show a comparable testicular phenotype to humans and are infertile. To detect possible signaling pathways and molecular mechanisms leading to the testicular phenotype in adult SCCx43KO mice and to their failure to initiate spermatogenesis, the testicular gene expression of 8-day-old SCCx43KO and wild-type (WT) mice was compared. Microarray analysis revealed that 658 genes were significantly regulated in testes of SCCx43KO mice. Of these genes, 135 were upregulated, whereas 523 genes were downregulated. For selected genes the results of the microarray analysis were confirmed using quantitative real-time PCR and immunostaining. The majority of the downregulated genes are GC-specific and are essential for mitotic and meiotic progression of spermatogenesis, including Stra8, Dazl and members of the DM (dsx and map-3) gene family. Other altered genes can be associated with transcription, metabolism, cell migration and cytoskeleton organization. Our data show that deletion of Cx43 in SCs leads to multiple alterations of gene expression in prepubertal mice and primarily affects GCs. The candidate genes could represent helpful markers for investigators exploring human testicular biopsies from patients showing corresponding spermatogenic deficiencies and for studying the molecular mechanisms of human male sterility.
Collapse
Affiliation(s)
- Sarah Giese
- Institute of Veterinary Anatomy, Histology and Embryology, University of Giessen, 35392 Giessen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
[Regulation of gene expression during spermatogenesis at transcriptional level]. YI CHUAN = HEREDITAS 2012; 33:1300-7. [PMID: 22207375 DOI: 10.3724/sp.j.1005.2011.01300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Mammalian spermatogenesis is a highly complex cell division and differentiation process occurring in the seminiferous tubules of the testis. This processes are regulated at both transcriptional and post-transcriptional levels, any mistake in this process can lead to infertility. Unveiling the molecular mechanisms of spermatogenesis has important implications for exploring novel contraceptive approach and treatment of infertility. This review addresses recent progress towards understanding the regulation of androgen, estrogen and their receptors, transcription factors and chromatin-associated factors for spermatogenesis at transcriptional level.
Collapse
|
34
|
Cao Y, Fu YL, Ge CH, Xu WX, Zhan YQ, Li CY, Li W, Wang XH, Wang ZD, Yu M, Yang XM. Mice overexpression of human augmenter of liver regeneration (hALR) in male germ cells shows abnormal spermatogenesis and reduced fertility. Endocr J 2012; 59:989-99. [PMID: 22863717 DOI: 10.1507/endocrj.ej12-0160] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Human augmenter of liver regeneration (hALR) is a sulfhydryl oxidase that is highly expressed in spermatogonia and early spermatocytes. To investigate the physiological effects of hALR in spermatogenesis, we generated a hALR transgenic mouse model driven by the human TSPY (testis-specific protein, Y-encoded) promoter that allows the transgene to be specifically activated in the testes. hALR content was found to be increased in both germ cells. The histological and TUNEL analysis of transgenic testes revealed a number of spermatogenetic defects including primary spermatocyte overpopulation followed by depletion through apoptosis, degenerating and detached nucleated germ cells, haploid cell loss and intraepithelial vacuoles of varying sizes. In line with these features, adult transgenic male mice also displayed a reduction in fertility. Our data suggest that regulated spatial and temporal expression of hALR is required for normal testicular development and spermatogenesis, and overexpression of hALR results in influencing the sperm morphology and quantity and the eventual reduction in male fertility. Present findings in the mouse may be of interest to human male fertility.
Collapse
Affiliation(s)
- Yan Cao
- Beijing Institute of Radiation Medicine, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Paternoster L, Standl M, Chen CM, Ramasamy A, Bønnelykke K, Duijts L, Ferreira MA, Alves AC, Thyssen JP, Albrecht E, Baurecht H, Feenstra B, Sleiman PMA, Hysi P, Warrington NM, Curjuric I, Myhre R, Curtin JA, Groen-Blokhuis MM, Kerkhof M, Sääf A, Franke A, Ellinghaus D, Fölster-Holst R, Dermitzakis E, Montgomery SB, Prokisch H, Heim K, Hartikainen AL, Pouta A, Pekkanen J, Blakemore AIF, Buxton JL, Kaakinen M, Duffy DL, Madden PA, Heath AC, Montgomery GW, Thompson PJ, Matheson MC, Le Souëf P, AAGC collaborators, Pourcain BS, Smith GD, Henderson J, Kemp JP, Timpson NJ, Deloukas P, Ring SM, Wichmann HE, Müller-Nurasyid M, Novak N, Klopp N, Rodríguez E, McArdle W, Linneberg A, Menné T, Nohr EA, Hofman A, Uitterlinden AG, van Duijn CM, Rivadeneira F, de Jongste JC, van der Valk RJP, Wjst M, Jogi R, Geller F, Boyd HA, Murray JC, Kim C, Mentch F, March M, Mangino M, Spector TD, Bataille V, Pennell CE, Holt PG, Sly P, Tiesler CMT, Thiering E, Illig T, Imboden M, Nystad W, Simpson A, Hottenga JJ, Postma D, Koppelman GH, Smit HA, Söderhäll C, Chawes B, Kreiner-Møller E, Bisgaard H, Melén E, Boomsma DI, Custovic A, Jacobsson B, Probst-Hensch NM, Palmer LJ, Glass D, Hakonarson H, et alPaternoster L, Standl M, Chen CM, Ramasamy A, Bønnelykke K, Duijts L, Ferreira MA, Alves AC, Thyssen JP, Albrecht E, Baurecht H, Feenstra B, Sleiman PMA, Hysi P, Warrington NM, Curjuric I, Myhre R, Curtin JA, Groen-Blokhuis MM, Kerkhof M, Sääf A, Franke A, Ellinghaus D, Fölster-Holst R, Dermitzakis E, Montgomery SB, Prokisch H, Heim K, Hartikainen AL, Pouta A, Pekkanen J, Blakemore AIF, Buxton JL, Kaakinen M, Duffy DL, Madden PA, Heath AC, Montgomery GW, Thompson PJ, Matheson MC, Le Souëf P, AAGC collaborators, Pourcain BS, Smith GD, Henderson J, Kemp JP, Timpson NJ, Deloukas P, Ring SM, Wichmann HE, Müller-Nurasyid M, Novak N, Klopp N, Rodríguez E, McArdle W, Linneberg A, Menné T, Nohr EA, Hofman A, Uitterlinden AG, van Duijn CM, Rivadeneira F, de Jongste JC, van der Valk RJP, Wjst M, Jogi R, Geller F, Boyd HA, Murray JC, Kim C, Mentch F, March M, Mangino M, Spector TD, Bataille V, Pennell CE, Holt PG, Sly P, Tiesler CMT, Thiering E, Illig T, Imboden M, Nystad W, Simpson A, Hottenga JJ, Postma D, Koppelman GH, Smit HA, Söderhäll C, Chawes B, Kreiner-Møller E, Bisgaard H, Melén E, Boomsma DI, Custovic A, Jacobsson B, Probst-Hensch NM, Palmer LJ, Glass D, Hakonarson H, Melbye M, Jarvis DL, Jaddoe VWV, Gieger C, The GOYA consortium, Strachan DP, Martin NG, Jarvelin MR, Heinrich J, Evans DM, Weidinger S. Meta-analysis of genome-wide association studies identifies three new risk loci for atopic dermatitis. Nat Genet 2011; 44:187-92. [PMID: 22197932 PMCID: PMC3272375 DOI: 10.1038/ng.1017] [Show More Authors] [Citation(s) in RCA: 261] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 11/01/2011] [Indexed: 02/07/2023]
Abstract
Atopic dermatitis (AD) is a commonly occurring chronic skin disease with high heritability. Apart from filaggrin (FLG), the genes influencing atopic dermatitis are largely unknown. We conducted a genome-wide association meta-analysis of 5,606 affected individuals and 20,565 controls from 16 population-based cohorts and then examined the ten most strongly associated new susceptibility loci in an additional 5,419 affected individuals and 19,833 controls from 14 studies. Three SNPs reached genome-wide significance in the discovery and replication cohorts combined, including rs479844 upstream of OVOL1 (odds ratio (OR) = 0.88, P = 1.1 × 10(-13)) and rs2164983 near ACTL9 (OR = 1.16, P = 7.1 × 10(-9)), both of which are near genes that have been implicated in epidermal proliferation and differentiation, as well as rs2897442 in KIF3A within the cytokine cluster at 5q31.1 (OR = 1.11, P = 3.8 × 10(-8)). We also replicated association with the FLG locus and with two recently identified association signals at 11q13.5 (rs7927894; P = 0.008) and 20q13.33 (rs6010620; P = 0.002). Our results underline the importance of both epidermal barrier function and immune dysregulation in atopic dermatitis pathogenesis.
Collapse
Affiliation(s)
- Lavinia Paternoster
- MRC CAiTE centre, School of Social & Community Medicine, University of Bristol, Bristol, UK
| | - Marie Standl
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Chih-Mei Chen
- Hannover Medical School, Department for Paediatric Pneumology, Allergy and Neonatology, Hannover, Germany
| | - Adaikalavan Ramasamy
- Respiratory Epidemiology and Public Health, Imperial College London, United Kingdom
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
- Department of Medical and Molecular Genetics, Kings College London, Guy’s Hospital, London, United Kingdom
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood; Health Sciences, University of Copenhagen & Copenhagen University Hospital, Gentofte, Denmark
| | - Liesbeth Duijts
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Pediatrics, Division of Respiratory Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Alexessander Couto Alves
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
| | - Jacob P Thyssen
- National Allergy Research Centre, Department of Dermato-Allergology, Gentofte Hospital, University of Copenhagen, Denmark
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Hansjörg Baurecht
- Department of Dermatology and Allergy, Technische Universität München, Munich, Germany
- ZAUM-Center for Allergy and Environment, Helmholtz-Zentrum and Technische Universität, Munich, Germany
- Graduate School of Information Science in Health, Technische Universität München, Munich, Germany
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Patrick MA Sleiman
- The Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Pirro Hysi
- Department of Twin Research and Genetic Epidemiology, King’s College London
| | - Nicole M Warrington
- School of Women’s and Infants’ Health, The University of Western Australia, Western Australia, Australia
| | - Ivan Curjuric
- Swiss Tropical and Public Health Institute (SwissTPH), Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Ronny Myhre
- Norwegian Institute of Public Health, Department of Genes and Environment, Division of Epidemiology, Oslo, Norway
| | - John A Curtin
- The University of Manchester, Manchester Academic Health Science Centre, NIHR Translational Research Facility in Respiratory Medicine, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | | | - Marjan Kerkhof
- Department of Pediatric Pulmonology and Allergology, University Medical Center Groningen, University of Groningen, GRIAC research institute, Groningen, The Netherlands
| | - Annika Sääf
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Regina Fölster-Holst
- Department of Dermatology, Allergology, and Venerology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Emmanouil Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, 1211 Switzerland
- Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Stephen B Montgomery
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, 1211 Switzerland
- Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Holger Prokisch
- Institute of Human Genetics, Technische Universität München, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Katharina Heim
- Institute of Human Genetics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Anneli Pouta
- Department of Obstetrics and Gynaecology, University of Oulu
- Department of Children, Young People and Families, National Institute for Health and Welfare, Finland
| | - Juha Pekkanen
- Department of Environmental Health, National Institute for Health and Welfare (THL), Kuopio, Finland
| | | | | | - Marika Kaakinen
- Institute of Health Sciences, University of Oulu, Oulu, Finland Biocenter Oulu, University of Oulu, Oulu, Finland
| | - David L Duffy
- Queensland Institute of Medical Research, Brisbane, Australia
| | - Pamela A Madden
- Washington University School of Medicine, St Louis, United States
| | - Andrew C Heath
- Washington University School of Medicine, St Louis, United States
| | | | - Philip J Thompson
- Lung Institute of Western Australia (WA) and Centre for Asthma, Allergy and Respiratory Research, University of WA, Perth, Australia
| | - Melanie C Matheson
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, University of Melbourne, Melbourne, Australia
| | - Peter Le Souëf
- School of Paediatrics and Child Health, Princess Margaret Hospital for Children, Perth, Australia
| | - AAGC collaborators
- Australian Asthma Genetics Consortium, a full list of collaborators is included in the Supplementary Note
| | - Beate St Pourcain
- The School of Social & Community Medicine, University of Bristol, Bristol, UK
| | - George Davey Smith
- MRC CAiTE centre, School of Social & Community Medicine, University of Bristol, Bristol, UK
| | - John Henderson
- The School of Social & Community Medicine, University of Bristol, Bristol, UK
| | - John P Kemp
- MRC CAiTE centre, School of Social & Community Medicine, University of Bristol, Bristol, UK
| | - Nicholas J Timpson
- MRC CAiTE centre, School of Social & Community Medicine, University of Bristol, Bristol, UK
| | - Panos Deloukas
- Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Susan M Ring
- The School of Social & Community Medicine, University of Bristol, Bristol, UK
| | - H-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Klinikum Grosshadern, Munich, Germany
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology and Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, Munich, Germany
| | - Natalija Novak
- Department of Dermatology and Allergy, University of Bonn Medical Center, Bonn, Germany
| | - Norman Klopp
- Unit for Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Elke Rodríguez
- Department of Dermatology and Allergy, Technische Universität München, Munich, Germany
- ZAUM-Center for Allergy and Environment, Helmholtz-Zentrum and Technische Universität, Munich, Germany
| | - Wendy McArdle
- University of Bristol, ALSPAC Laboratory, School of Social & Community Medicine, University of Bristol, Bristol, UK
| | - Allan Linneberg
- Research Center for Prevention and Health, Glostrup University Hospital, Denmark
| | - Torkil Menné
- National Allergy Research Centre, Department of Dermato-Allergology, Gentofte Hospital, University of Copenhagen, Denmark
| | - Ellen A Nohr
- Institute of Public Health, Aarhus University, Denmark
| | - Albert Hofman
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - André G Uitterlinden
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Fernando Rivadeneira
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Johan C de Jongste
- Department of Pediatrics, Division of Respiratory Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ralf JP van der Valk
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Pediatrics, Division of Respiratory Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Matthias Wjst
- Comprehensive Pneumology Center and Institute of Lung Biology and Disease, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Rain Jogi
- Lung Clinic, Tartu University, Tartu, Estonia
| | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Heather A Boyd
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | | | - Cecilia Kim
- The Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Frank Mentch
- The Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michael March
- The Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King’s College London
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King’s College London
| | - Veronique Bataille
- Department of Twin Research and Genetic Epidemiology, King’s College London
| | - Craig E Pennell
- School of Women’s and Infants’ Health, The University of Western Australia, Western Australia, Australia
| | - Patrick G Holt
- Telethon Institute for Child Health Research and Centre for Child Health Research, The University of Western Australia, Western Australia, Australia
| | - Peter Sly
- Queensland Children’s Medical Research Institute; University of Queensland; WHO Collaborating Centre for Research on Children’s Environmental Health, Queensland, Australia
| | - Carla MT Tiesler
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Division of Metabolic Diseases and Nutritional Medicine, Dr von Hauner Children’s Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Elisabeth Thiering
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Illig
- Unit for Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Medea Imboden
- Swiss Tropical and Public Health Institute (SwissTPH), Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Wenche Nystad
- Norwegian Institute of Public Health, Division of Epidemiology, Oslo, Norway
| | - Angela Simpson
- The University of Manchester, Manchester Academic Health Science Centre, NIHR Translational Research Facility in Respiratory Medicine, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Dirkje Postma
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerard H Koppelman
- Dpt of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children ’s Hospital, GRIAC research institute, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Henriette A Smit
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Cilla Söderhäll
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Bo Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood; Health Sciences, University of Copenhagen & Copenhagen University Hospital, Gentofte, Denmark
| | - Eskil Kreiner-Møller
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood; Health Sciences, University of Copenhagen & Copenhagen University Hospital, Gentofte, Denmark
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood; Health Sciences, University of Copenhagen & Copenhagen University Hospital, Gentofte, Denmark
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Astrid Lindgren Children’s Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Dorret I Boomsma
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Adnan Custovic
- The University of Manchester, Manchester Academic Health Science Centre, NIHR Translational Research Facility in Respiratory Medicine, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
| | - Bo Jacobsson
- Norwegian Institute of Public Health, Department of Genes and Environment, Division of Epidemiology, Oslo, Norway
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Sahlgrenska Academy, Göteborg University, Sweden
| | - Nicole M Probst-Hensch
- Swiss Tropical and Public Health Institute (SwissTPH), Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Lyle J Palmer
- Ontario Institute for Cancer Research, Toronto; University of Toronto, Toronto, Canada
| | - Daniel Glass
- Department of Twin Research and Genetic Epidemiology, King’s College London
| | - Hakon Hakonarson
- The Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatric, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Deborah L Jarvis
- Respiratory Epidemiology and Public Health, Imperial College London, United Kingdom
| | - Vincent WV Jaddoe
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - The GOYA consortium
- The Genetics of Overweight Young Adults consortium, list of members in Supplementary Note
| | - David P Strachan
- Division of Population Health Sciences and Education, St George’s, University of London, London, UK
| | | | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom
- Institute of Health Sciences, Biocenter, University of Oulu, Finland
- National Institute of Health and Welfare, Finland
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - David M Evans
- MRC CAiTE centre, School of Social & Community Medicine, University of Bristol, Bristol, UK
| | - Stephan Weidinger
- Department of Dermatology, Allergology, and Venerology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
36
|
Bettegowda A, Wilkinson MF. Transcription and post-transcriptional regulation of spermatogenesis. Philos Trans R Soc Lond B Biol Sci 2010; 365:1637-51. [PMID: 20403875 DOI: 10.1098/rstb.2009.0196] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Spermatogenesis in mammals is achieved by multiple players that pursue a common goal of generating mature spermatozoa. The developmental processes acting on male germ cells that culminate in the production of the functional spermatozoa are regulated at both the transcription and post-transcriptional levels. This review addresses recent progress towards understanding such regulatory mechanisms and identifies future challenges to be addressed in this field. We focus on transcription factors, chromatin-associated factors and RNA-binding proteins necessary for spermatogenesis and/or sperm maturation. Understanding the molecular mechanisms that govern spermatogenesis has enormous implications for new contraceptive approaches and treatments for infertility.
Collapse
Affiliation(s)
- Anilkumar Bettegowda
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, MC 0864, La Jolla, CA 92093-0864, USA
| | | |
Collapse
|
37
|
Zhou H, Liu LH, Zhang H, Lei Z, Lan ZJ. Expression of zinc finger protein 105 in the testis and its role in male fertility. Mol Reprod Dev 2010; 77:511-20. [PMID: 20186958 PMCID: PMC3062163 DOI: 10.1002/mrd.21171] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Using an in silico approach, we identified a putative zinc finger domain-containing transcription factor (zinc finger protein 105, ZFP105) enriched in the adult mouse testis. RT-PCR analyses showed that Zfp105 was indeed highly expressed in adult mouse testis and that its expression was regulated during postnatal development. To further characterize Zfp105 expression, we generated a Zfp105:beta-galactosidase (LacZ) knock-in reporter mouse line (Zfp105(LacZ/+)) in which a Zfp105:LacZ fusion gene was expressed. Whole-mount LacZ analyses of adult Zfp105(LacZ/+) tissues showed robust LacZ staining in the testis, very weak staining in the ovary, and no staining in the spleen, liver, kidney, heart, lung, thymus, adrenal gland, uterus, or oviduct. Sectional LacZ staining showed that ZFP105 was highly expressed in pachytene spermatocytes. ZNF35, the human ortholog of Zfp105, was also highly expressed in human testis. Immunofluorescence analysis showed that ZNF35 was located primarily in the cytoplasm of male germ cells. More importantly, reduced male fertility was observed in adult Zfp105(LacZ/LacZ) mice. Histological studies showed the presence of undifferentiated spermatogenic cells in the lumen of seminiferous tubules at stage VII and in the epididymal lumen of adult Zfp105(LacZ/LacZ) mice. Taken together, our results suggest that ZFP105 is a male germ-cell factor and plays a role in male reproduction.
Collapse
Affiliation(s)
- Huaxin Zhou
- Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA.
| | | | | | | | | |
Collapse
|
38
|
Rashid S, Grzmil P, Drenckhahn JD, Meinhardt A, Adham I, Engel W, Neesen J. Disruption of the murine dynein light chain gene Tcte3-3 results in asthenozoospermia. Reproduction 2010; 139:99-111. [PMID: 19778998 DOI: 10.1530/rep-09-0243] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To elucidate the role of the mouse gene Tcte3 (Tctex2), which encodes a putative light chain of the outer dynein arm of cilia and sperm flagella, we have inactivated this gene in mice using targeted disruption. Breeding of heterozygous males and females resulted in normal litter size; however, we were not able to detect homozygous Tcte3-deficent mice using standard genotype techniques. In fact, our results indicate the presence of at least three highly similar copies of the Tcte3 gene (Tcte3-1, Tcte3-2, and Tcte3-3) in the murine genome. Therefore, quantitative real-time PCR was established to differentiate between mice having one or two targeted Tcte3-3 alleles. By this approach, Tcte3-3(-/-) animals were identified, which were viable and revealed no obvious malformation. Interestingly, some homozygous Tcte3-3-deficient male mice bred with wild-type female produced no offspring while other Tcte3-3-deficient males revealed decreased sperm motility but were fertile. In infertile Tcte3-3(-/-) males, spermatogenesis was affected and sperm motility was reduced, too, resulting in decreased ability of Tcte3-3-deficient spermatozoa to move from the uterus into the oviduct. Impaired flagellar motility is not correlated with any gross defects in the axonemal structure, since outer dynein arms are detectable in sperm of Tcte3-3(-/-) males. However, in infertile males, deficient Tcte3-3 function is correlated with increased apoptosis during male germ cell development, resulting in a reduction of sperm number. Moreover, multiple malformations in developing haploid germ cells are present. Our results support a role of Tcte3-3 in generation of sperm motility as well as in male germ cell differentiation.
Collapse
Affiliation(s)
- Sajid Rashid
- Institute of Human Genetics, University of Goettingen, 37073 Goettingen, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Vetere A, Li WC, Paroni F, Juhl K, Guo L, Nishimura W, Dai X, Bonner-Weir S, Sharma A. OVO homologue-like 1 (Ovol1) transcription factor: a novel target of neurogenin-3 in rodent pancreas. Diabetologia 2010; 53:115-22. [PMID: 19882138 PMCID: PMC3066144 DOI: 10.1007/s00125-009-1567-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Accepted: 09/14/2009] [Indexed: 10/20/2022]
Abstract
AIMS/HYPOTHESIS The basic helix-loop-helix transcription factor neurogenin-3 (NGN3) commits the fates of pancreatic progenitors to endocrine cell types, but knowledge of the mechanisms regulating the choice between proliferation and differentiation of these progenitors is limited. METHODS Using a chromatin immunoprecipitation cloning approach, we searched for direct targets of NGN3 and identified a zinc-finger transcription factor, OVO homologue-like 1 (OVOL1). Transactivation experiments were carried out to elucidate the functional role of NGN3 in Ovol1 gene expression. Embryonic and adult rodents pancreases were immunostained for OVOL1, Ki67 and NGN3. RESULTS We showed that NGN3 negatively regulates transcription of Ovol1 in an E-box-dependent fashion. The presence of either NGN3 or NEUROD1, but not MYOD, reduced endogenous Ovol1 mRNA. OVOL1 was detected in pancreatic tissue around embryonic day 15.5, after which OVOL1 levels dramatically increased. In embryonic pancreas, OVOL1 protein levels were low in NGN3(+) or Ki67(+) cells, but high in quiescent differentiated cells. OVOL1 presence was maintained in adult pancreas, where it was detected in islets, pancreatic ducts and some acinar cells. Additionally OVOL1 presence was lacking in proliferating ductules in regenerating pancreas and induced in cells as they began to acquire their differentiated phenotype. CONCLUSIONS/INTERPRETATION The timing of OVOL1 appearance in pancreas and its increased levels in differentiated cells suggest that OVOL1 promotes the transition of cells from a proliferating, less-differentiated state to a quiescent more-differentiated state. We conclude that OVOL1, a downstream target of NGN3, may play an important role in regulating the balance between proliferation and differentiation of pancreatic cells.
Collapse
Affiliation(s)
- A Vetere
- Section of Islet Transplantation and Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wells J, Lee B, Cai AQ, Karapetyan A, Lee WJ, Rugg E, Sinha S, Nie Q, Dai X. Ovol2 suppresses cell cycling and terminal differentiation of keratinocytes by directly repressing c-Myc and Notch1. J Biol Chem 2009; 284:29125-35. [PMID: 19700410 DOI: 10.1074/jbc.m109.008847] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ovol2 belongs to the Ovo family of evolutionarily conserved zinc finger transcription factors that act downstream of key developmental signaling pathways including Wg/Wnt and BMP/TGF-beta. We previously reported Ovol2 expression in the basal layer of epidermis, where epidermal stem/progenitor cells reside. In this work, we use HaCaT human keratinocytes to investigate the cellular and molecular functions of Ovol2. We show that depletion of Ovol2 leads to transient cell expansion but a loss of cells with long term proliferation potential. Mathematical modeling and experimental findings suggest that both faster cycling and precocious withdrawal from the cell cycle underlie this phenotype. Ovol2 depletion also accelerates extracellular signal-induced terminal differentiation in two- and three-dimensional culture models. By chromatin immunoprecipitation, luciferase reporter, and functional rescue assays, we demonstrate that Ovol2 directly represses two critical downstream targets, c-Myc and Notch1, thereby suppressing keratinocyte transient proliferation and terminal differentiation, respectively. These findings shed light on how an epidermal cell maintains a proliferation-competent and differentiation-resistant state.
Collapse
Affiliation(s)
- Julie Wells
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, California 92697, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Gu B, Sun P, Yuan Y, Moraes RC, Li A, Teng A, Agrawal A, Rhéaume C, Bilanchone V, Veltmaat JM, Takemaru KI, Millar S, Lee EYHP, Lewis MT, Li B, Dai X. Pygo2 expands mammary progenitor cells by facilitating histone H3 K4 methylation. ACTA ACUST UNITED AC 2009; 185:811-26. [PMID: 19487454 PMCID: PMC2711593 DOI: 10.1083/jcb.200810133] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent studies have unequivocally identified multipotent stem/progenitor cells in mammary glands, offering a tractable model system to unravel genetic and epigenetic regulation of epithelial stem/progenitor cell development and homeostasis. In this study, we show that Pygo2, a member of an evolutionarily conserved family of plant homeo domain–containing proteins, is expressed in embryonic and postnatal mammary progenitor cells. Pygo2 deficiency, which is achieved by complete or epithelia-specific gene ablation in mice, results in defective mammary morphogenesis and regeneration accompanied by severely compromised expansive self-renewal of epithelial progenitor cells. Pygo2 converges with Wnt/β-catenin signaling on progenitor cell regulation and cell cycle gene expression, and loss of epithelial Pygo2 completely rescues β-catenin–induced mammary outgrowth. We further describe a novel molecular function of Pygo2 that is required for mammary progenitor cell expansion, which is to facilitate K4 trimethylation of histone H3, both globally and at Wnt/β-catenin target loci, via direct binding to K4-methyl histone H3 and recruiting histone H3 K4 methyltransferase complexes.
Collapse
Affiliation(s)
- Bingnan Gu
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Haerizadeh F, Wong CE, Bhalla PL, Gresshoff PM, Singh MB. Genomic expression profiling of mature soybean (Glycine max) pollen. BMC PLANT BIOLOGY 2009; 9:25. [PMID: 19265555 PMCID: PMC2660330 DOI: 10.1186/1471-2229-9-25] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Accepted: 03/06/2009] [Indexed: 05/19/2023]
Abstract
BACKGROUND Pollen, the male partner in the reproduction of flowering plants, comprises either two or three cells at maturity. The current knowledge of the pollen transcriptome is limited to the model plant systems Arabidopsis thaliana and Oryza sativa which have tri-cellular pollen grains at maturity. Comparative studies on pollen of other genera, particularly crop plants, are needed to understand the pollen gene networks that are subject to functional and evolutionary conservation. In this study, we used the Affymetrix Soybean GeneChip to perform transcriptional profiling on mature bi-cellular soybean pollen. RESULTS Compared to the sporophyte transcriptome, the soybean pollen transcriptome revealed a restricted and unique repertoire of genes, with a significantly greater proportion of specifically expressed genes than is found in the sporophyte tissue. Comparative analysis shows that, among the 37,500 soybean transcripts addressed in this study, 10,299 transcripts (27.46%) are expressed in pollen. Of the pollen-expressed sequences, about 9,489 (92.13%) are also expressed in sporophytic tissues, and 810 (7.87%) are selectively expressed in pollen. Overall, the soybean pollen transcriptome shows an enrichment of transcription factors (mostly zinc finger family proteins), signal recognition receptors, transporters, heat shock-related proteins and members of the ubiquitin proteasome proteolytic pathway. CONCLUSION This is the first report of a soybean pollen transcriptional profile. These data extend our current knowledge regarding regulatory pathways that govern the gene regulation and development of pollen. A comparison between transcription factors up-regulated in soybean and those in Arabidopsis revealed some divergence in the numbers and kinds of regulatory proteins expressed in both species.
Collapse
Affiliation(s)
- Farzad Haerizadeh
- Plant Molecular Biology and Biotechnology Laboratory, ARC Centre of Excellence for Integrative Legume Research, Faculty of Land and Food resources, The University of Melbourne, Parkville 3010, Australia
| | | | - Prem L Bhalla
- Plant Molecular Biology and Biotechnology Laboratory, ARC Centre of Excellence for Integrative Legume Research, Faculty of Land and Food resources, The University of Melbourne, Parkville 3010, Australia
| | - Peter M Gresshoff
- ARC Centre of Excellence for Integrative Legume Research, The University of Queensland, Brisbane, Australia
| | - Mohan B Singh
- Plant Molecular Biology and Biotechnology Laboratory, ARC Centre of Excellence for Integrative Legume Research, Faculty of Land and Food resources, The University of Melbourne, Parkville 3010, Australia
| |
Collapse
|
43
|
Lenartowicz M, Brzek B, Polański Z, Sierpniowska A, Galas J, Bilińska B. Changes in germ cells population and in vitro oocyte maturation in female mice with imperforated vagina. ACTA ACUST UNITED AC 2008; 309:269-77. [PMID: 18412125 DOI: 10.1002/jez.457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
This study characterized female mice with a congenital defect in their reproductive tract. In females derived from an outbred colony maintained in the Department of Genetics and Evolution, the frequency of the imperforated vaginae was approximately 2%. A consequence of this defect is infertility. Affected animals developed hydrometrocolpos (the uterus and vagina were greatly distended by fluid). Morphology of the ovary and oviduct was normal and similar to that of control mice. In the females with an imperforated vagina numerous corpora lutea were observed. Accordingly, we have found that both spontaneous as well as exogenous gonadotrophin-induced ovulation occurs in such females. Nevertheless, oocytes derived from ovaries of occluded females exhibited a partial block during in vitro maturation. Histological analysis of ovarian tissue revealed an increase in the number of primary follicles (type I follicles) and a decrease in the number of secondary and antral follicles (type IV and type VI follicles) when compared with control mice. The concentration of androgens in the ovarian tissue was higher in the affected females. Our data show that females with an imperforated vagina can be a useful model for studying the mechanism of genetic control of the development of the urogenital tract in mammals.
Collapse
Affiliation(s)
- Małgorzata Lenartowicz
- Department of Genetics and Evolution, Institute of Zoology, Jagiellonian University, Kraków, Poland.
| | | | | | | | | | | |
Collapse
|
44
|
Rossi P, Lolicato F, Grimaldi P, Dolci S, Di Sauro A, Filipponi D, Geremia R. Transcriptome analysis of differentiating spermatogonia stimulated with kit ligand. Gene Expr Patterns 2007; 8:58-70. [PMID: 18036996 DOI: 10.1016/j.modgep.2007.10.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 10/02/2007] [Accepted: 10/17/2007] [Indexed: 01/15/2023]
Abstract
Kit ligand (KL) is a survival factor and a mitogenic stimulus for differentiating spermatogonia. However, it is not known whether KL also plays a role in the differentiative events that lead to meiotic entry of these cells. We performed a wide genome analysis of difference in gene expression induced by treatment with KL of spermatogonia from 7-day-old mice, using gene chips spanning the whole mouse genome. The analysis revealed that the pattern of RNA expression induced by KL is compatible with the qualitative changes of the cell cycle that occur during the subsequent cell divisions in type A and B spermatogonia, i.e. the progressive lengthening of the S phase and the shortening of the G2/M transition. Moreover, KL up-regulates in differentiating spermatogonia the expression of early meiotic genes (for instance: Lhx8, Nek1, Rnf141, Xrcc3, Tpo1, Tbca, Xrcc2, Mesp1, Phf7, Rtel1), whereas it down-regulates typical spermatogonial markers (for instance: Pole, Ptgs2, Zfpm2, Egr2, Egr3, Gsk3b, Hnrpa1, Fst, Ptch2). Since KL modifies the expression of several genes known to be up-regulated or down-regulated in spermatogonia during the transition from the mitotic to the meiotic cell cycle, these results are consistent with a role of the KL/kit interaction in the induction of their meiotic differentiation.
Collapse
Affiliation(s)
- Pellegrino Rossi
- Dipartimento di Sanita' Pubblica e Biologia Cellulare, Universita' degli Studi di Roma Tor Vergata, via Montpellier 1, 00133 Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
45
|
Unezaki S, Horai R, Sudo K, Iwakura Y, Ito S. Ovol2/Movo, a homologue of Drosophila ovo, is required for angiogenesis, heart formation and placental development in mice. Genes Cells 2007; 12:773-85. [PMID: 17573777 DOI: 10.1111/j.1365-2443.2007.01084.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The zinc-finger transcription factor Ovol2 (Movo, Movo2) is a mouse homologue of Drosophila ovo, which is essential for the survival and differentiation of female germ line cells. To elucidate OVOL2 function in mammals, we generated Ovol2-deficient mice by gene targeting. The Ovol2 mutants died at embryonic days 9.5-10.5 (E9.5-E10.5), as a result of defects in extraembryonic and embryonic vascularization, and in heart formation. Although the Ovol2 expression was weak, severe defects were detected in extraembryonic and embryonic vascularization, and in heart formation at E8.5-E9.5. In Ovol2(-/-) placentas, allantoic blood vessel expansion and development of the labyrinthine layer were impaired at E10.5. In an endothelial cell line, siRNAs for Ovol2 reduced the expression of Ovol2 and inhibited the capillary-like network formation on Matrigel in vitro. These results demonstrate that Ovol2 may play a critical role in vascular angiogenesis during early embryogenesis.
Collapse
Affiliation(s)
- Sawako Unezaki
- Department of Medical Chemistry, Kansai Medical University, Moriguchi, 570-8506, Japan
| | | | | | | | | |
Collapse
|
46
|
Li B, Rhéaume C, Teng A, Bilanchone V, Munguia JE, Hu M, Jessen S, Piccolo S, Waterman ML, Dai X. Developmental phenotypes and reduced Wnt signaling in mice deficient for pygopus 2. Genesis 2007; 45:318-25. [PMID: 17458864 DOI: 10.1002/dvg.20299] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Canonical Wnt signaling involves complex intracellular events culminating in the stabilization of beta-catenin, which enters the nucleus and binds to LEF/TCF transcription factors to stimulate gene expression. Pygopus was identified as a genetic modifier of Wg (Wnt homolog) signaling in Drosophila, and encodes a PHD domain protein that associates with the beta-catenin/LEF/TCF complex. Two murine pygopus paralogs, mpygo1 and mpygo2, have been identified, but their roles in development and Wnt signaling remain elusive. In this study, we report that ablation of mpygo2 expression in mice causes defects in morphogenesis of both ectodermally and endodermally derived tissues, including brain, eyes, hair follicles, and lung. However, no gross abnormality was observed in embryonic intestine. Using a BAT-gal reporter, we found Wnt signaling at most body sites to be reduced in the absence of mpygo2. Taken together, our studies show for the first time that mpygo2 deletion affects embryonic development of some but not all Wnt-requiring tissues.
Collapse
Affiliation(s)
- Boan Li
- Department of Biological Chemistry, University of California, Irvine, California 92697-1700, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Nair M, Bilanchone V, Ortt K, Sinha S, Dai X. Ovol1 represses its own transcription by competing with transcription activator c-Myb and by recruiting histone deacetylase activity. Nucleic Acids Res 2007; 35:1687-97. [PMID: 17311813 PMCID: PMC1865076 DOI: 10.1093/nar/gkl1141] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Ovol1 belongs to a family of evolutionarily conserved zinc finger proteins that act downstream of key developmental signaling pathways such as Wnt and TGF-β/BMP. It plays important roles in epithelial and germ cell development, particularly by repressing c-Myc and Id2 genes and modulating the balance between proliferation and differentiation of progenitor cells. In this study, we show that Ovol1 negatively regulates its own expression by binding to and repressing the activity of its promoter. We further demonstrate that Ovol1 uses both passive and active repression mechanisms to auto-repress: (1) it antagonizes transcriptional activation of c-Myb, a known positive regulator of proliferation, by competing for DNA binding; (2) it recruits histone deacetylase activity to the promoter via an N-terminal SNAG repressor domain. At Ovol1 cognate sites in the endogenous Ovol1 promoter, c-Myb binding correlates with increased histone acetylation, whereas the expression of Ovol1 correlates with a displacement of c-Myb from the DNA and decreased histone acetylation. Collectively, our data suggest that Ovol1 restricts its own expression by counteracting c-Myb activation and histone acetylation of the Ovol1 promoter.
Collapse
Affiliation(s)
- Mahalakshmi Nair
- Department of Biological Chemistry, School of Medicine, Developmental Biology Center, University of California, Irvine, CA 92697, USA and Department of Biochemistry, State University of New York at Buffalo, New York, USA
| | - Virginia Bilanchone
- Department of Biological Chemistry, School of Medicine, Developmental Biology Center, University of California, Irvine, CA 92697, USA and Department of Biochemistry, State University of New York at Buffalo, New York, USA
| | - Kori Ortt
- Department of Biological Chemistry, School of Medicine, Developmental Biology Center, University of California, Irvine, CA 92697, USA and Department of Biochemistry, State University of New York at Buffalo, New York, USA
| | - Satrajit Sinha
- Department of Biological Chemistry, School of Medicine, Developmental Biology Center, University of California, Irvine, CA 92697, USA and Department of Biochemistry, State University of New York at Buffalo, New York, USA
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, Developmental Biology Center, University of California, Irvine, CA 92697, USA and Department of Biochemistry, State University of New York at Buffalo, New York, USA
- *To whom correspondence should be addressed. +1 949 824 3101+1 949 824 2688
| |
Collapse
|
48
|
Teng A, Wells MN, Segre JA, Dai X. Strain-dependent perinatal lethality of Ovol1-deficient mice and identification of Ovol2 as a downstream target of Ovol1 in skin epidermis. Biochim Biophys Acta Mol Basis Dis 2006; 1772:89-95. [PMID: 17049212 PMCID: PMC1773004 DOI: 10.1016/j.bbadis.2006.08.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2006] [Accepted: 08/15/2006] [Indexed: 01/29/2023]
Abstract
Ovol1 encodes a zinc finger transcriptional repressor that is downstream of the LEF1/beta-catenin complex, nuclear effectors of canonical Wnt signaling. Previous gene knockout studies performed in a 129SvxC57BL/6 mixed genetic background revealed that Ovol1-deficient mice survive to adulthood but display multiple tissue defects. In this study, we describe a C57BL/6 strain-specific reduction in perinatal survival of Ovol1 mutant mice. The perinatal lethality is accompanied by kidney epithelial cysts of embryonic onset and delayed skin barrier acquisition. Genetic analysis suggests a partial functional compensation by Ovol2 for the loss of Ovol1. The expression of Ovol2 was up-regulated in Ovol1-deficient epidermis, and Ovol1 represses the activity of Ovol2 promoter in a DNA binding-dependent manner. Collectively, these studies uncover novel functions of Ovol1 in mouse development and identify Ovol2 as a downstream target of Ovol1.
Collapse
Affiliation(s)
| | | | - Julia A. Segre
- National Human Genome Research Institute, NIH, 49 Convent
Drive, Bethesda, MD 20892
| | - Xing Dai
- Department of Biological Chemistry
- Developmental Biology Center University of California, Irvine,
CA 92697
- *To whom correspondence should be addressed:
Department of Biological Chemistry College of Medicine D250 Med Sci I University
of California Irvine, CA 92697-1700 Tel: 949-824-3101 Fax: 949-824-2688 E-mail:
| |
Collapse
|
49
|
Gomis RR, Alarcón C, He W, Wang Q, Seoane J, Lash A, Massagué J. A FoxO-Smad synexpression group in human keratinocytes. Proc Natl Acad Sci U S A 2006; 103:12747-52. [PMID: 16908841 PMCID: PMC1568919 DOI: 10.1073/pnas.0605333103] [Citation(s) in RCA: 196] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Transforming growth factor beta (TGF-beta) signals through activation of Smad transcription factors. Activated Smad proteins associate with different DNA-binding cofactors for the recognition and regulation of specific target genes. Members of the forkhead box O family (FoxO1, FoxO3, and FoxO4) play such a role in the induction of the cyclin-dependent kinase inhibitors p15Ink4b and p21Cip1. To delineate the organization of the TGF-beta response in human keratinocytes, we defined the set of genes whose activation by TGF-beta requires both FoxO and Smad functions. FoxO factors are shown to be essential for 11 of the 115 immediate gene activation responses to TGF-beta in these cells. FoxO1, FoxO3, and FoxO4 act redundantly as mediators of these effects. Smad4, which functions as a partner of receptor-phosphorylated Smad2/3, is required for all of these responses. These results define a FoxO-Smad synexpression group or group of genes that are jointly induced by a common mechanism in response to TGF-beta. In addition to p15INK4b and p21CIP1, these genes include mediators of stress responses (GADD45A, GADD45B, and IER1) and adaptive cell signaling responses (CTGF, JAG1, LEMD3, SGK, CDC42EP3, and OVOL1). Bioinformatic analysis of the promoter region of these genes reveals diverse configurations of Smad and FoxO binding elements, implying differences in the regulatory properties of this group of genes. Indeed, a subset of FoxO/Smad-dependent TGF-beta gene responses additionally require the transcription factor CCAAT/enhancer-binding protein beta. The composition of the FoxO-Smad synexpression group suggests that stress reactions and adaptive functions accompany the cytostatic response of keratinocytes to TGF-beta.
Collapse
Affiliation(s)
- Roger R. Gomis
- *Cancer Biology and Genetics Program, Howard Hughes Medical Institute, and
| | - Claudio Alarcón
- *Cancer Biology and Genetics Program, Howard Hughes Medical Institute, and
| | - Wei He
- *Cancer Biology and Genetics Program, Howard Hughes Medical Institute, and
| | - Qiongqing Wang
- *Cancer Biology and Genetics Program, Howard Hughes Medical Institute, and
| | - Joan Seoane
- *Cancer Biology and Genetics Program, Howard Hughes Medical Institute, and
| | - Alex Lash
- Bioinformatics Core Facility, Memorial Sloan–Kettering Cancer Center, New York, NY 10021
| | - Joan Massagué
- *Cancer Biology and Genetics Program, Howard Hughes Medical Institute, and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
50
|
Nair M, Teng A, Bilanchone V, Agrawal A, Li B, Dai X. Ovol1 regulates the growth arrest of embryonic epidermal progenitor cells and represses c-myc transcription. ACTA ACUST UNITED AC 2006; 173:253-64. [PMID: 16636146 PMCID: PMC2063816 DOI: 10.1083/jcb.200508196] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Transcriptional control plays a key role in regulating epidermal proliferation and differentiation. Although ample information has been obtained on how epidermal homeostasis is controlled in adult skin, less is known about the control of proliferation/differentiation of epidermal stem/progenitor cells in the developing embryo. Ovol1, encoding a zinc finger protein homologous to Drosophila melanogaster Ovo, is expressed in embryonic epidermal progenitor cells that are transiting from proliferation to terminal differentiation. In this study, we demonstrate a function for Ovol1 in interfollicular epidermal development. In its absence, developing epidermis fails to properly restrict the proliferative potential of progenitor cells, and cultured keratinocytes fail to efficiently undergo growth arrest in response to extrinsic growth-inhibitory signals. We present molecular evidence that c-myc expression is up-regulated in Ovol1-deficient suprabasal cells and that Ovol1 represses c-myc transcription by directly binding to its promoter. Collectively, our findings indicate that Ovol1 is required for proliferation exit of committed epidermal progenitor cells and identify c-myc as an Ovol1 target.
Collapse
Affiliation(s)
- Mahalakshmi Nair
- Department of Biological Chemistry, University of California, Irvine, 92697, USA
| | | | | | | | | | | |
Collapse
|