1
|
Yu C, Zhong B, Zhang Y, Zhao H, Wu J, Yu H, Yu H, Li H. Combining ATAC-seq and RNA-seq reveals key genes for gonadal abnormalities in one-month-old XX-DSD pigs. BMC Genomics 2025; 26:447. [PMID: 40329180 PMCID: PMC12057259 DOI: 10.1186/s12864-025-11613-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 04/18/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Disorders of Sex Development (DSD) are caused by congenital abnormalities in the chromosomes, and subsequent development of gonads or sexual anatomy. XX-DSD pigs exhibit a series of adverse symptoms such as sterility, genital infections, and decline in meat quality, leading to significant economic losses in the breeding industry. However, the understanding of the etiology and pathogenesis of XX-DSD in pigs remains limited. To investigate the molecular mechanisms underlying abnormal gonadal development in XX-DSD pigs, we analyzed the gonads of 1-month-old XX-DSD pigs, normal females, and normal males using RNA-seq and ATAC-seq techniques. RESULTS From RNA-seq, we identified potential genes involved in gonadal development in XX-DSD pigs, including SOX9, HSD3B1, CYP19A1, CCNB2, CYP11A1, DMRT1, and MGP. Following this, we analyzed ATAC-seq data and identified 14,820 differential accessible chromatin peaks. Then, by integrating the ATAC-seq and RNA-seq analysis results, we identified several candidate genes (SOX9, COL1A1, COL1A2, FDX1, COL6A1, HSD3B1, FSHR, and CYP17A1) that might be associated with sex development. Through PPI (Protein-Protein Interaction Networks) analysis, we found that SOX9 gene was the top hub gene. Furthermore, we confirmed the effect of the open chromatin region on SOX9 gene expression by a dual-luciferase reporter assay, thus further validating the critical role of this open region in regulating SOX9 expression. CONCLUSIONS This study elucidates the critical regulatory role of specific open chromatin structures in the SOX9 gene promoter region (8647563-8648475) in gonadal development of XX-DSD pigs. Additionally, we identify that genes such as SOX9, HSD3B1, and CYP19A1 act in concert to participate in gonadal development. These findings provide molecular evidence for the dynamic chromatin regulatory network underlying gonadal dysgenesis in XX-DSD and lay the foundation for subsequent mechanistic studies.
Collapse
Affiliation(s)
- Congying Yu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Animal Science and Technology College, Foshan University, Foshan, Guangdong, 528225, China
| | - Bingzhou Zhong
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Animal Science and Technology College, Foshan University, Foshan, Guangdong, 528225, China
| | - Yuqiao Zhang
- Zhongshan Baishi Pig Farm Co., Ltd.r, Zhongshan, 528463, China
| | - Haiquan Zhao
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Animal Science and Technology College, Foshan University, Foshan, Guangdong, 528225, China
| | - Jinhua Wu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Animal Science and Technology College, Foshan University, Foshan, Guangdong, 528225, China
| | - Haiyi Yu
- School of Biological Sciences, Crawley (Perth), The University of Western Australia, 35 Stirling Highway, Perth, WA, 6009, Australia
| | - Hui Yu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Animal Science and Technology College, Foshan University, Foshan, Guangdong, 528225, China.
| | - Hua Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Animal Science and Technology College, Foshan University, Foshan, Guangdong, 528225, China.
| |
Collapse
|
2
|
Kumar S, Pandit R, Sarathi V, Memon SS, Lila AR, Thakkar H, Arya S, Karlekar M, Dodamani MH, Barnabas R, Patil VA, Shah NS, Bandgar TR. 46, XY under-virilization and NR5A1 variants: Monocentric Indian experience and systematic review. ANNALES D'ENDOCRINOLOGIE 2025; 86:101731. [PMID: 40280302 DOI: 10.1016/j.ando.2025.101731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
PURPOSE NR5A1 variants are rare causes of 46,XY DSD with scarce literature from India. A systematic review of genotype-phenotype correlation is lacking. We aim to describe clinical, biochemical, histological, and genotype-phenotype correlation in 46,XY DSD with NR5A1 variants. METHODS Retrospective monocentric review of 11 genetically-proven probands and systematic review including these and 288 from the literature. RESULTS Eleven probands of 46,XY DSD with NR5A1 variants from our centre exhibited phenotypic variability, female-to-male social-gender change in ∼two-thirds (4/7), primary adrenal insufficiency (PAI) in one, and five novel variants. Systematic review included 299 probands (age: 4.0 [0.3-13] years; Sinnecker score: 4 [3-4]) with 218 different NR5A1 variants. Systematic review reported female-to-male gender-change (27/166, 16.3%), spontaneous puberty/pubertal virilization (37/86, 43%), DSD in siblings (25/299, 8.3%), paternal hypospadias (7/299, 2.3%), maternal premature ovarian insufficiency (19/299, 6.4%), bilateral labio-scrotal gonads (71/214, 33.2%), absent Mullerian structure (187/232, 80.6%), PAI (5/222, 2.2%) and gonadal malignancy (2/111, 1.8%) in probands. Serum LH was elevated in mini-puberty, pre-puberty, and peri/post-puberty in 35.4% (17/48), 46.2% (18/39), and 63.6% (49/77) patients, respectively. Germ cells were present in 55.6% (5/9) in mini-pubertal age and absent in 96.8% (61/63) at later age. Sertoli cells were reported normal in 100% (7/7), and 70.4% (38/54) in mini-pubertal and later ages, respectively. Presence of Mullerian structures and Sinnecker score of 4/5 were associated with LBD variants (54.5%vs. 30.6%, P=0.003) and protein start-lost/deletion (7.3% vs. nil, P=0.004), respectively. CONCLUSIONS 46,XY DSD with NR5A1 variants is characterized by progressive decline in Sertoli and Leydig cell function, pubertal virilization, frequent partial gonadal dysgenesis and probably lower gonadal malignancy risk than gonadal dysgenesis of other origin. Further studies are warranted to validate these observations.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Endocrinology, Seth G.S. Medical College, KEM Hospital, Parel, Mumbai 400012, Maharashtra, India
| | - Reshma Pandit
- Department of Endocrinology, Seth G.S. Medical College, KEM Hospital, Parel, Mumbai 400012, Maharashtra, India
| | - Vijaya Sarathi
- Department of Endocrinology, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, India
| | - Saba Samad Memon
- Department of Endocrinology, Seth G.S. Medical College, KEM Hospital, Parel, Mumbai 400012, Maharashtra, India
| | - Anurag Ranjan Lila
- Department of Endocrinology, Seth G.S. Medical College, KEM Hospital, Parel, Mumbai 400012, Maharashtra, India.
| | - Hemangini Thakkar
- Department of Radiology, Seth G.S. Medical College, KEM Hospital, Mumbai, India
| | - Sneha Arya
- Department of Endocrinology, Seth G.S. Medical College, KEM Hospital, Parel, Mumbai 400012, Maharashtra, India
| | - Manjiri Karlekar
- Department of Endocrinology, Seth G.S. Medical College, KEM Hospital, Parel, Mumbai 400012, Maharashtra, India
| | | | - Rohit Barnabas
- Department of Endocrinology, Seth G.S. Medical College, KEM Hospital, Parel, Mumbai 400012, Maharashtra, India
| | - Virendra A Patil
- Department of Endocrinology, Seth G.S. Medical College, KEM Hospital, Parel, Mumbai 400012, Maharashtra, India
| | - Nalini S Shah
- Department of Endocrinology, Seth G.S. Medical College, KEM Hospital, Parel, Mumbai 400012, Maharashtra, India
| | - Tushar R Bandgar
- Department of Endocrinology, Seth G.S. Medical College, KEM Hospital, Parel, Mumbai 400012, Maharashtra, India
| |
Collapse
|
3
|
Li J, Zhang X, Wang X, Wang Z, Li X, Zheng J, Li J, Xu G, Sun C, Yi G, Yang N. Single-nucleus transcriptional and chromatin accessible profiles reveal critical cell types and molecular architecture underlying chicken sex determination. J Adv Res 2025; 70:29-43. [PMID: 38734369 PMCID: PMC11976570 DOI: 10.1016/j.jare.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/23/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
INTRODUCTION Understanding the sex determination mechanisms in birds has great significance for the biological sciences and production in the poultry industry. Sex determination in chickens is a complex process that involves fate decisions of supporting cells such as granulosa or Sertoli cells. However, a systematic understanding of the genetic regulation and cell commitment process underlying sex determination in chickens is still lacking. OBJECTIVES We aimed to dissect the molecular characteristics associated with sex determination in the gonads of chicken embryos. METHODS Single-nucleus RNA-seq (snRNA-seq) and ATAC-seq (snATAC-seq) analysis were conducted on the gonads of female and male chickens at embryonic day 3.5 (E3.5), E4.5, and E5.5. RESULTS Here, we provided a time-course transcriptional and chromatin accessible profiling of gonads during chicken sex determination at single-cell resolution. We uncovered differences in cell composition and developmental trajectories between female and male gonads and found that the divergence of transcription and accessibility in gonadal cells first emerged at E5.5. Furthermore, we revealed key cell-type-specific transcription factors (TFs) and regulatory networks that drive lineage commitment. Sex determination signaling pathways, dominated by BMP signaling, are preferentially activated in males during gonadal development. Further pseudotime analysis of the supporting cells indicated that granulosa cells were regulated mainly by the TEAD gene family and that Sertoli cells were driven by the DMRT1 regulons. Cross-species analysis suggested high conservation of both cell types and cell-lineage-specific TFs across the six vertebrates. CONCLUSIONS Overall, our study will contribute to accelerating the development of sex manipulation technology in the poultry industry and the application of chickens as a unique model for studying cell fate decisions.
Collapse
Affiliation(s)
- Jianbo Li
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Xiuan Zhang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Xiqiong Wang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Zhen Wang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Xingzheng Li
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Jiangxia Zheng
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Junying Li
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Guiyun Xu
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China
| | - Congjiao Sun
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China.
| | - Guoqiang Yi
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Kunpeng Institute of Modern Agriculture at Foshan, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China.
| | - Ning Yang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
4
|
Gao Y, Wang Z, Long Y, Yang L, Jiang Y, Ding D, Teng B, Chen M, Yuan J, Gao F. Unveiling the roles of Sertoli cells lineage differentiation in reproductive development and disorders: a review. Front Endocrinol (Lausanne) 2024; 15:1357594. [PMID: 38699384 PMCID: PMC11063913 DOI: 10.3389/fendo.2024.1357594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/07/2024] [Indexed: 05/05/2024] Open
Abstract
In mammals, gonadal somatic cell lineage differentiation determines the development of the bipotential gonad into either the ovary or testis. Sertoli cells, the only somatic cells in the spermatogenic tubules, support spermatogenesis during gonadal development. During embryonic Sertoli cell lineage differentiation, relevant genes, including WT1, GATA4, SRY, SOX9, AMH, PTGDS, SF1, and DMRT1, are expressed at specific times and in specific locations to ensure the correct differentiation of the embryo toward the male phenotype. The dysregulated development of Sertoli cells leads to gonadal malformations and male fertility disorders. Nevertheless, the molecular pathways underlying the embryonic origin of Sertoli cells remain elusive. By reviewing recent advances in research on embryonic Sertoli cell genesis and its key regulators, this review provides novel insights into sex determination in male mammals as well as the molecular mechanisms underlying the genealogical differentiation of Sertoli cells in the male reproductive ridge.
Collapse
Affiliation(s)
- Yang Gao
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Zican Wang
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Yue Long
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Lici Yang
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Yongjian Jiang
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Dongyu Ding
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Baojian Teng
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Min Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, China
- Lin He’s Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, China
- Lin He’s Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
5
|
Tao Q, Zhang L, Zhang Y, Liu M, Wang J, Zhang Q, Wu J, Wang A, Jin Y, Tang K. The miR-34b/MEK/ERK pathway is regulated by NR5A1 and promotes differentiation in primary bovine Sertoli cells. Theriogenology 2024; 215:224-233. [PMID: 38100994 DOI: 10.1016/j.theriogenology.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023]
Abstract
Sertoli cells play a key role in testicular development and spermatogenesis. It has been suggested that Sertoli cells differentiate after their proliferation ceases. Our previous study showed that miR-34b inhibits proliferation by targeting MAP2K1 mediated MEK/ERK signaling pathway in bovine immature Sertoli cells. Subsequent studies have revealed that the differentiation marker androgen receptor is upregulated during this process. However, the effect of the miR-34b/MEK/ERK pathway on immature bovine Sertoli cell differentiation and the underlying molecular mechanisms are yet to be explored. In this study, we determined that the miR-34b/MEK/ERK pathway was involved in the differentiation of primary Sertoli cells (PSCs) in response to retinoic acid. Transfection of an miR-34b mimic into PSCs promoted cell differentiation, whereas transfection of an miR-34b inhibitor into PSCs delayed it. Pharmacological inhibition of MEK/ERK signaling by AZD6244 promoted PSCs differentiation. Mechanistically, miR-34b promoted PSCs differentiation by inhibiting the MEK/ERK signaling pathway. Through a combination of bioinformatics analysis, dual-luciferase reporter assay, quantitative real-time PCR, and western blotting, nuclear receptor subfamily 5 group A member 1 (NR5A1) was identified as an upstream negative transcription factor of miR-34b. Furthermore, NR5A1 knockdown promoted Sertoli cell differentiation, whereas NR5A1 overexpression had the opposite effect. Together, this study revealed a new NR5A1/miR-34b/MEK/ERK axis that plays a significant role in Sertoli cell differentiation and provides a theoretical and experimental framework for further clarifying the regulation of cell differentiation in bovine PSCs.
Collapse
Affiliation(s)
- Qibing Tao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Linlin Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Yun Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Mingming Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Jie Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Qian Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiancheng Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, China
| | - Aihua Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, China.
| | - Yaping Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, China.
| | - Keqiong Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
6
|
Zhang P, Wang M, Chen X, Jing K, Li Y, Liu X, Ran H, Qin J, Zhong J, Cai X. Dysregulated genes in undifferentiated spermatogonia and Sertoli cells are associated with the spermatogenic arrest in cattleyak. Mol Reprod Dev 2022; 89:632-645. [PMID: 36409004 DOI: 10.1002/mrd.23653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/23/2022]
Abstract
Hybrid male sterility (HMS) is a reproductive isolation mechanism limiting the formation of fertile offspring through interspecific fertilization. Cattleyak is the interspecific hybrid presenting significant heterosis in several economic traits, but HMS restricted its wide reproduction in cettleyak breeding. In this study, we detected the specifically expressed genes of a variety of cells (undifferentiated spermatogonia, primary spermatocytes, secondary spermatocytes, haploid spermatids, sperm, Sertoli cells, Leydig cells, and macrophages) in the testis of yak and cattleyak, and found that the spermatogenesis of cattleyak might be blocked at meiosis I, and the expression of niche factors (NR5A1, GATA4, STAR, CYP11A1, CD68, TNF, and CX3CR1) in undifferentiated spermatogonia niche was abnormal. Then we isolated the undifferentiated spermatogonia and Sertoli cells from yak and cattleyak by enzyme digestion, and detected the specific genes in the two bovid testicular cells as well as the proliferation capacity of the undifferentiated spermatogonia. These results indicated that weak proliferation ability and scarce number of undifferentiated spermatogonia and abnormal gene expressions in Sertoli cells may contribute to male sterility of cattleyak.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Mingxiu Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xuemei Chen
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Kemin Jing
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yuqian Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinrui Liu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hongbiao Ran
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jie Qin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jincheng Zhong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province and Ministry of Education, Institute of Qinghai-Tibetan Plateau, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Different fixatives influence morphology, antigen preservation, and TUNEL staining in chicken (Gallus gallus) testis. Acta Histochem 2021; 123:151822. [PMID: 34861475 DOI: 10.1016/j.acthis.2021.151822] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022]
Abstract
The optimized fixative for testis is still controversial. This study investigated the effects of Modified Davidson's Fluid (mDF), 4% Paraformaldehyde (4% PFA), and Bouin's Fluid (BF) fixatives on chicken testes in normal/cadmium (Cd) feeding groups using hematoxylin and eosin (HE), immunohistochemistry (IHC), and Terminal Transferase dUTP Nick End Labeling (TUNEL) staining. Compared to the mDF, we established that the testes fixed with 4% PFA and BF in the normal group had severe shrinkage in tubular and interstitial compartments. Moreover, compared with 4% PFA, the number of GATA4-positive Sertoli cells/mm2 reduced by 67.61% in mDF and 80.57% in BF for one seminiferous tubule. The TUNEL assay illustrated that more positive cells/mm2 in mDF group (28.47 ± 11.38) than in 4% PFA (10.49 ± 7.89). In Cd-treated testes, mDF showed more morphological details than 4% PFA and BF. In contrast, the number of GATA4-positive Sertoli cells/mm2 of 4% PFA was higher than that of mDF by 65.78% and BF by 64.80% in a seminiferous tubule. The number of TUNEL positive cells/mm2 in mDF (272.60 ± 34.41) were higher than in 4% PFA (175.91 ± 19.87). These results suggest that mDF fixative is suitable for normal and Cd-treated testis fixation for HE and TUNEL staining in chicken, whereas 4% PFA fixative is better for IHC examination.
Collapse
|
8
|
The conditional deletion of steroidogenic factor 1 (Nr5a1) in Sox9-Cre mice compromises testis differentiation. Sci Rep 2021; 11:4486. [PMID: 33627800 PMCID: PMC7904858 DOI: 10.1038/s41598-021-84095-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 02/12/2021] [Indexed: 12/25/2022] Open
Abstract
Steroidogenic factor 1 (NR5A1) is essential for gonadal development. To study the importance of NR5A1 during early gonadal sex differentiation, we generated Sox9-Cre-Nr5a1 conditional knockout (cKO) mice: Sox9-Cre;Nr5a1flox/flox and Sox9-Cre;Nr5a1flox/− mice. Double-immunostaining for NR5A1 and AMH revealed silenced NR5A1 in Sertoli cells and reduced AMH+ cells in the gonads of XY Sox9-Cre-Nr5a1 cKO mice between embryonic days 12.5 (E12.5) and E14.5. Double-immunostaining for SOX9 and FOXL2 further indicated an early block in Sertoli cells and ectopic granulosa cell differentiation. The number of cells expressing the Leydig cell marker 3βHSD obviously reduced in the gonads of XY Sox9-Cre;Nr5a1flox/− but not Sox9-Cre;Nr5a1flox/flox mice at E15.5. The presence of STRA8+ cells indicated that germ cells entered meiosis in the gonads of XY Sox9-Cre-Nr5a1 cKO mice. The results of qRT-PCR revealed remarkably reduced and elevated levels of testis and ovary markers, respectively, in the gonads of XY Sox9-Cre-Nr5a1 cKO mice at E12.5‒E13.5. These data suggested that the loss of Nr5a1 abrogates the testicular pathway and induces the ectopic ovarian pathway, resulting in postnatal partial/complete male-to-female gonadal sex reversal. Our findings provide evidence for the critical role of NR5A1 in murine gonadal sex determination in vivo.
Collapse
|
9
|
Yin S, Qin W, Wang B, Zhou J, Yang L, Xiong X, Li J. Absence of Sirtuin 1 impairs the testicular development in cattleyak by inactivating SF-1. Reprod Domest Anim 2020; 55:1054-1060. [PMID: 32497285 DOI: 10.1111/rda.13737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/20/2020] [Indexed: 12/30/2022]
Abstract
Cattleyak, which are interspecific hybrids between cattle and yak, display much higher growth performances than yak. However, F1 male cattleyak are infertile due to defective testicular development. Sirtuin 1 (SIRT1) is a histone deacetylase that is essential for various biological processes, while the roles of testicular SIRT1 in yak and cattleyak are still poorly understood. Here, we found that SIRT1 was localized in various kinds of yak testicular cells except elongated spermatids while it was deficient in cattleyak testis. Further studies indicated that cattleyak testis exhibited decreased histone acetylation levels on H3 and H4. One of SIRT1 co-factors, steroidogenic factor-1 (SF-1), was lost in cattleyak testis at protein level. Expressions of several SF-1 target genes responsible for Sertoli cell development and steroidogenesis, including STAR, CYP11A1, CYP26B1, FDX1 and HSD3B, decreased significantly in cattleyak testis. In addition, SIRT1-mediated P53 acetylation was not responsible for the cell apoptosis in cattleyak testis. Taken together, our results suggested the deficiency of SIRT1 in yak testis caused inactivation of SF-1 and the impairment of testicular development. This research provides theoretical bases for understanding the mechanism of cattleyak sterility and gives new insights in revealing the roles of SIRT1 in regulating yak testicular development.
Collapse
Affiliation(s)
- Shi Yin
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Chengdu, China.,Key Laboratory of Modem Technology (Southwest Minzu University), State Ethnic Affairs Commission, Chengdu, China
| | - Wenchang Qin
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Bin Wang
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Jingwen Zhou
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Liuqing Yang
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Xianrong Xiong
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China
| | - Jian Li
- College of Life Science and Technology, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
10
|
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
|
11
|
Cui L, Gu Y, Liu S, Li M, Ye J, Zhang F, Luo X, Chang WL, Gui Y. TBC1D20 Is Essential for Mouse Blood-Testis Barrier Integrity Through Maintaining the Epithelial Phenotype and Modulating the Maturation of Sertoli Cells. Reprod Sci 2020; 27:1443-1454. [PMID: 31994000 DOI: 10.1007/s43032-020-00156-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/10/2019] [Indexed: 11/30/2022]
Abstract
Sertoli cells are important for spermatogenesis not only by directly interacting with germ line cells in the seminiferous epithelium but also by constituting the blood-testis barrier (BTB) structure to create a favorable environment for spermatogenesis. Blind sterile (bs) male mice are infertile, with excessive germ cell apoptosis and spermatogenesis arrest. TBC1D20 (TBC1 domain family member 20) deficiency has been identified as the causative mutation in bs mice. However, whether TBC1D20 loss of function also impairs BTB integrity, which further contributes to the failed spermatogenesis of bs male mice, remains unclear. In the present study, biotin tracer assay and transmission electron microscopy showed severely disrupted BTB integrity in bs testes. Compared to the wild-type Sertoli cells, BTB components of cultured bs Sertoli cells in vitro was perturbed with downregulation of E-cadherin, ZO-1, β-catenin, and Claudin 11. The obvious rearrangement of F-actin indicated disrupted epithelial-mesenchymal balance in TBC1D20-deficient Sertoli cells. The ability of bs Sertoli cells to maintain the clone formation of spermatogonia stem cells was also obviously limited. Furthermore, the decreasing of SOX9 (sex-determining region Y box 9) and WT1 (Wilms' tumor 1) and increasing of vimentin in bs Sertoli cells indicated that TBC1D20 loss of function attenuated the differentiation progression of bs Sertoli cells. In summary, TBC1D20 loss of function impedes the maturation of adult Sertoli cells and resulted in impaired BTB integrity, which is further implicated in the infertile phenotype of bs male mice.
Collapse
Affiliation(s)
- Lina Cui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Yanli Gu
- Department of Obstetrics, the People's Hospital of Longhua, Shenzhen, 518109, China
| | - Shuo Liu
- College of Biological Sciences and Technology, Beijing Forestry University, Beijing, 10083, China
| | - Minghua Li
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Jing Ye
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Fanting Zhang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Xiaomin Luo
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Wen-Lin Chang
- Department of Obstetrics, the People's Hospital of Longhua, Shenzhen, 518109, China.
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China.
| |
Collapse
|
12
|
Singh S, Singh SK. Effect of gestational exposure to perfluorononanoic acid on neonatal mice testes. J Appl Toxicol 2019; 39:1663-1671. [PMID: 31389053 DOI: 10.1002/jat.3883] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/24/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022]
Abstract
Perfluoroalkyl acids (PFAAs) are widely used in commercial products and are found in many goods of daily use. Perfluorononanoic acid (PFNA) is one of the PFAAs that possesses endocrine disrupting properties and we have recently shown that PFNA affects testicular functions in Parkes mice. Exposure to environmental endocrine disruptors during fetal life is believed to affect gonadal development and they might produce reproductive abnormalities in males. Therefore, the present study examined the effect of gestational exposure to PFNA on the testes of neonatal mice offspring. Pregnant Parkes mice were orally administered PFNA (2 and 5 mg/kg body weight) or distilled water from gestational day 12 until parturition. Male pups were killed on postnatal day 3. PFNA treatment decreased testosterone biosynthesis by inhibiting expression of steroidogenic acute regulatory protein, cytochrome P450scc, and 3β- and 17β-hydroxysteroid dehydrogenase; proliferation of testicular cells was also affected in treated mice. Furthermore, a marked decrease in expression of Wilms tumor 1, steroidogenic factor 1 and insulin-like factor 3 was noted in neonatal mice testes, indicating that the PFNA treatment may affect the development of the testis. Moreover, observation of the dose-related expression of anti-müllerian hormone and c-Kit in neonatal mice testes is also suggestive of an interference with gonadal development by PFNA exposure. In conclusion, the results suggest that the gestational exposure to PFNA decreased testosterone biosynthesis and altered the expression of critical factors involved in the development of the testis, thereby advocating a potential risk of PFNA to male reproductive health.
Collapse
Affiliation(s)
- Shilpi Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Shio Kumar Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
13
|
Anamthathmakula P, Miryala CSJ, Moreci RS, Kyathanahalli C, Hassan SS, Condon JC, Jeyasuria P. Steroidogenic Factor 1 (Nr5a1) is Required for Sertoli Cell Survival Post Sex Determination. Sci Rep 2019; 9:4452. [PMID: 30872705 PMCID: PMC6418149 DOI: 10.1038/s41598-019-41051-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 01/31/2019] [Indexed: 12/25/2022] Open
Abstract
The elevated level of Steroidogenic Factor 1 (Nr5a1, Sf-1) expression in the male gonadal development pathway, post sex determination, implies a vital role in testis gonadal differentiation. In this study we generated Sertoli cell-specific Nr5a1 KO mice (SC-SF-1-/-) at E14.5, which coincides with testis development post sex determination, using the Amh-Cre mouse model. Analysis of SC-SF-1-/- (Sertoli cell specific Nr5a1 knockout) testes demonstrated apoptosis as early as E15. Further analysis revealed that SC-SF-1-/- gonads displayed lower MDM2 levels resulting in elevated TP53 levels, which we believe may lead to apoptosis of the Sertoli cell population, inferring the possibility that NR5A1 directly regulates MDM2 expression. By E15.5, the Sertoli cell and germ cell population declined in SC-SF-1-/- mice resulting in the disruption of seminiferous cords with limited cord structure remaining at E18.5. Due to the loss of Sertoli and germ cells, the testis weights of SC-SF-1-/- mice at 6-weeks were much reduced; however, SC-SF-1-/- seminal vesicles weights were comparable suggesting intact Leydig cell androgen production. We conclude that NR5A1 regulates the TP53 pathway during development, is essential for fetal Sertoli cell survival and controls the cell cycle of Sertoli cells during differentiation.
Collapse
Affiliation(s)
- Prashanth Anamthathmakula
- Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Mott Center for Growth and Development, Detroit, MI, USA
| | - Chandra Suma Johnson Miryala
- Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Mott Center for Growth and Development, Detroit, MI, USA
| | - Rebecca S Moreci
- Department of Cell Biology, Duke University, Durham, NC, 27708, USA
| | - Chandrashekara Kyathanahalli
- Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Mott Center for Growth and Development, Detroit, MI, USA
| | - Sonia S Hassan
- Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Mott Center for Growth and Development, Detroit, MI, USA
| | - Jennifer C Condon
- Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Mott Center for Growth and Development, Detroit, MI, USA
| | - Pancharatnam Jeyasuria
- Department of Obstetrics and Gynecology, Wayne State University Perinatal Initiative, School of Medicine, Wayne State University, Mott Center for Growth and Development, Detroit, MI, USA.
| |
Collapse
|
14
|
Mandal K, Bader SL, Kumar P, Malakar D, Campbell DS, Pradhan BS, Sarkar RK, Wadhwa N, Sensharma S, Jain V, Moritz RL, Majumdar SS. An integrated transcriptomics-guided genome-wide promoter analysis and next-generation proteomics approach to mine factor(s) regulating cellular differentiation. DNA Res 2018; 24:143-157. [PMID: 28065881 PMCID: PMC5397609 DOI: 10.1093/dnares/dsw057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 11/24/2016] [Indexed: 12/19/2022] Open
Abstract
Differential next-generation-omics approaches aid in the visualization of biological processes and pave the way for divulging important events and/or interactions leading to a functional output at cellular or systems level. To this end, we undertook an integrated Nextgen transcriptomics and proteomics approach to divulge differential gene expression of infant and pubertal rat Sertoli cells (Sc).Unlike, pubertal Sc, infant Sc are immature and fail to support spermatogenesis. We found exclusive association of 14 and 19 transcription factor binding sites to infantile and pubertal states of Sc, respectively, using differential transcriptomics-guided genome-wide computational analysis of relevant promoters employing 220 Positional Weight Matrices from the TRANSFAC database. Proteomic SWATH-MS analysis provided extensive quantification of nuclear and cytoplasmic protein fractions revealing 1,670 proteins differentially located between the nucleus and cytoplasm of infant Sc and 890 proteins differentially located within those of pubertal Sc. Based on our multi-omics approach, the transcription factor YY1 was identified as one of the lead candidates regulating differentiation of Sc.YY1 was found to have abundant binding sites on promoters of genes upregulated during puberty. To determine its significance, we generated transgenic rats with Sc specific knockdown of YY1 that led to compromised spermatogenesis.
Collapse
Affiliation(s)
- Kamal Mandal
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, India
| | | | - Pankaj Kumar
- G.N.R. Knowledge Centre for Genome Informatics, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | | | | | - Bhola Shankar Pradhan
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, India
| | - Rajesh K Sarkar
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, India
| | - Neerja Wadhwa
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, India
| | - Souvik Sensharma
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, India
| | - Vaibhav Jain
- Next-Generation Sequencing Facility, National Institute of Immunology, New Delhi, India
| | | | - Subeer S Majumdar
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, India.,National Institute of Animal Biotechnology, Miyapur, Hyderabad, India
| |
Collapse
|
15
|
Chen H, Wang Y, Ge R, Zirkin BR. Leydig cell stem cells: Identification, proliferation and differentiation. Mol Cell Endocrinol 2017; 445:65-73. [PMID: 27743991 PMCID: PMC5346484 DOI: 10.1016/j.mce.2016.10.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 10/03/2016] [Accepted: 10/11/2016] [Indexed: 01/21/2023]
Abstract
Adult Leydig cells develop from undifferentiated mesenchymal-like stem cells (stem Leydig cells, SLCs) present in the interstitial compartment of the early postnatal testis. Putative SLCs also have been identified in peritubular and perivascular locations of the adult testis. The latter cells, which normally are quiescent, are capable of regenerating new Leydig cells upon the loss of the adult cells. Recent studies have identified several protein markers to identify these cells, including nestin, PDGFRα, COUP-TFII, CD51 and CD90. We have shown that the proliferation of the SLCs is stimulated by DHH, FGF2, PDGFBB, activin and PDGFAA. Suppression of proliferation occurred with TGFβ, androgen and PKA signaling. The differentiation of the SLCs into testosterone-producing Leydig cells was found to be regulated positively by DHH (Desert hedgehog), lithium-induced signaling and activin; and negatively by TGFβ, PDGFBB, FGF2, Notch and Wnt signaling. DHH, by itself, was found to induce SLC differentiation into LH-responsive steroidogenic cells, suggesting that DHH plays a critical role in the commitment of SLC into the Leydig lineage. These studies, taken together, address the function and regulation of low turnover stem cells in a complex, adult organ, and also have potential application to the treatment of androgen deficiency.
Collapse
Affiliation(s)
- Haolin Chen
- Center for Scientific Research, Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | - Yiyan Wang
- Center for Scientific Research, Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Renshan Ge
- Center for Scientific Research, Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Barry R Zirkin
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
16
|
Murugananthkumar R, Senthilkumaran B. Expression analysis and localization of wt1, ad4bp/sf-1 and gata4 in the testis of catfish, Clarias batrachus: Impact of wt1-esiRNA silencing. Mol Cell Endocrinol 2016; 431:164-76. [PMID: 27173028 DOI: 10.1016/j.mce.2016.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/06/2016] [Accepted: 05/08/2016] [Indexed: 11/23/2022]
Abstract
In teleosts, a comprehensive role or interaction of wt1, ad4bp/sf-1 and gata4 genes in relation to gonadal development and/or recrudescence was never attempted. Present study aimed to identify the involvement of these genes during testicular development of catfish, Clarias batrachus. Dominant expression of wt1 and gata4 was observed in developing and adult testis, while ad4bp/sf-1 showed steady expression. Localization of these genes in adult testis revealed their presence in spermatogonia, spermatocytes and interstitial/Leydig cells. Significant high expression during pre-spawning and spawning phases, and upregulated levels of these genes after hCG induction authenticated gonadotropic regulation. Transient silencing of wt1-esiRNA displayed decrease in wt1 expression, which further downregulated the expression of ad4bp/sf-1 and gata4, and certain steroidogenic enzyme genes related to androgen production. These results suggest that wt1 might target ad4bp/sf-1 and gata4 expression, and also have regulatory influence either indirectly or directly on the steroidogenic enzyme genes of catfish.
Collapse
Affiliation(s)
- Raju Murugananthkumar
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, P.O. Central University, Hyderabad 500046, Telangana, India
| | - Balasubramanian Senthilkumaran
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, P.O. Central University, Hyderabad 500046, Telangana, India.
| |
Collapse
|
17
|
Boyer A, Girard M, Thimmanahalli DS, Levasseur A, Céleste C, Paquet M, Duggavathi R, Boerboom D. mTOR Regulates Gap Junction Alpha-1 Protein Trafficking in Sertoli Cells and Is Required for the Maintenance of Spermatogenesis in Mice. Biol Reprod 2016; 95:13. [PMID: 27281705 PMCID: PMC5029431 DOI: 10.1095/biolreprod.115.138016] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/14/2016] [Accepted: 05/17/2016] [Indexed: 12/29/2022] Open
Abstract
The mammalian target of rapamycin (Mtor) gene encodes a serine/threonine kinase that acts as a master regulator of processes as diverse as cell growth, protein synthesis, cytoskeleton reorganization, and cell survival. In the testis, physiological roles for Mtor have been proposed in perinatal Sertoli cell proliferation and blood-testis barrier (BTB) remodeling during spermatogenesis, but no in vivo studies of Mtor function have been reported. Here, we used a conditional knockout approach to target Mtor in Sertoli cells. The resulting Mtor(flox/flox); Amhr2(cre/+) mice were characterized by progressive, adult-onset testicular atrophy associated with disorganization of the seminiferous epithelium, loss of Sertoli cell polarity, increased germ cell apoptosis, premature release of germ cells, decreased epididymal sperm counts, increased sperm abnormalities, and infertility. Histopathologic analysis and quantification of the expression of stage-specific markers showed a specific loss of pachytene spermatocytes and spermatids. Although the BTB and the ectoplasmic specializations did not appear to be altered in Mtor(flox/flox);Amhr2(cre/+) mice, a dramatic redistribution of gap junction alpha-1 (GJA1) was detected in their Sertoli cells. Phosphorylation of GJA1 at Ser373, which is associated with its internalization, was increased in the testes of Mtor(flox/flox); Amhr2(cre/+) mice, as was the expression and phosphorylation of AKT, which phosphorylates GJA1 at this site. Together, these results indicate that Mtor expression in Sertoli cells is required for the maintenance of spermatogenesis and the progression of germ cell development through the pachytene spermatocyte stage. One mechanism of mTOR action may be to regulate gap junction dynamics by inhibiting AKT, thereby decreasing GJA1 phosphorylation and internalization. mTOR regulates gap junction alpha-1 protein distribution in Sertoli cells and is necessary for progression through the pachytene spermatocyte stage.
Collapse
Affiliation(s)
- Alexandre Boyer
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Montréal, Québec, Canada
| | - Meggie Girard
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Montréal, Québec, Canada
| | | | - Adrien Levasseur
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Montréal, Québec, Canada
| | - Christophe Céleste
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Montréal, Québec, Canada
| | - Marilène Paquet
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Montréal, Québec, Canada
| | - Rajesha Duggavathi
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, Québec, Canada
| | - Derek Boerboom
- Centre de Recherche en Reproduction Animale, Faculté de Médecine Vétérinaire, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
18
|
Ferlin A, Rocca MS, Vinanzi C, Ghezzi M, Di Nisio A, Foresta C. Mutational screening of NR5A1 gene encoding steroidogenic factor 1 in cryptorchidism and male factor infertility and functional analysis of seven undescribed mutations. Fertil Steril 2015; 104:163-9.e1. [PMID: 25989977 DOI: 10.1016/j.fertnstert.2015.04.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/01/2015] [Accepted: 04/11/2015] [Indexed: 01/27/2023]
Abstract
OBJECTIVE To study the role of NR5A1 in cryptorchidism and male factor infertility. Mutations in NR5A1 have been initially associated with primary adrenal insufficiency and 46,XY gonadal dysgenesis and more recently with less severe phenotypes, including preliminary descriptions in severe forms of male factor infertility. Far less clear is the possible involvement of NR5A1 mutations in cryptorchidism. DESIGN Retrospective cross-sectional cohort study and functional analysis of mutant proteins. SETTING University department. PATIENT(S) Nine hundred fifty-nine subjects, including children with cryptorchidism and adults with different semen phenotypes associated or not associated with a history of cryptorchidism. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Mutation screening of NR5A1 by sequencing all exons. Functional analysis of mutant proteins by transactivation assays of CYP11A1 and CYP17A1 promoters. RESULT(S) We identified seven undescribed and one previously described missense mutation in subjects with severe spermatogenic impairment, without (4/236, 1.7%) and with (3/85, 3.5%) a history of cryptorchidism. Newborns with cryptorchidism carry NR5A1 mutations at low frequency (0.7%), whereas no mutations were found in milder forms of infertility and normozoospermia, irrespective of the presence of cryptorchidism. The mutant proteins showed impaired transactivation of gonadal promoters. A single nucleotide polymorphism (rs1110061; c.437 G→C; p.Gly146Ala) was also associated with more severe forms of spermatogenic impairment with cryptorchidism. CONCLUSION(S) This study, combined with what is already known about NR5A1-associated phenotypes, suggests considering mutations in this gene as a novel genetic cause of more severe forms of male factor infertility, especially when associated with a history of cryptorchidism.
Collapse
Affiliation(s)
- Alberto Ferlin
- Department of Medicine, Unit of Andrology and Human Reproductive Medicine, University of Padova, Padova, Italy.
| | - Maria Santa Rocca
- Department of Medicine, Unit of Andrology and Human Reproductive Medicine, University of Padova, Padova, Italy
| | - Cinzia Vinanzi
- Department of Medicine, Unit of Andrology and Human Reproductive Medicine, University of Padova, Padova, Italy
| | - Marco Ghezzi
- Department of Medicine, Unit of Andrology and Human Reproductive Medicine, University of Padova, Padova, Italy
| | - Andrea Di Nisio
- Department of Medicine, Unit of Andrology and Human Reproductive Medicine, University of Padova, Padova, Italy
| | - Carlo Foresta
- Department of Medicine, Unit of Andrology and Human Reproductive Medicine, University of Padova, Padova, Italy
| |
Collapse
|
19
|
Bao J, Tang C, Yuan S, Porse BT, Yan W. UPF2, a nonsense-mediated mRNA decay factor, is required for prepubertal Sertoli cell development and male fertility by ensuring fidelity of the transcriptome. Development 2014; 142:352-62. [PMID: 25503407 DOI: 10.1242/dev.115642] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Nonsense-mediated mRNA decay (NMD) represents a highly conserved RNA surveillance mechanism through which mRNA transcripts bearing premature termination codons (PTCs) are selectively degraded to maintain transcriptomic fidelity in the cell. Numerous in vitro studies have demonstrated the importance of the NMD pathway; however, evidence supporting its physiological necessity has only just started to emerge. Here, we report that ablation of Upf2, which encodes a core NMD factor, in murine embryonic Sertoli cells (SCs) leads to severe testicular atrophy and male sterility owing to rapid depletion of both SCs and germ cells during prepubertal testicular development. RNA-Seq and bioinformatic analyses revealed impaired transcriptomic homeostasis in SC-specific Upf2 knockout testes, characterized by an accumulation of PTC-containing transcripts and the transcriptome-wide dysregulation of genes encoding splicing factors and key proteins essential for SC fate control. Our data demonstrate an essential role of UPF2-mediated NMD in prepubertal SC development and male fertility.
Collapse
Affiliation(s)
- Jianqiang Bao
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, 1664 North Virginia Street, MS575, Reno, NV 89557, USA
| | - Chong Tang
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, 1664 North Virginia Street, MS575, Reno, NV 89557, USA
| | - Shuiqiao Yuan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, 1664 North Virginia Street, MS575, Reno, NV 89557, USA
| | - Bo T Porse
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Jagtvej 124, Copenhagen, DK-2200, Denmark Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Jagtvej 124, Copenhagen, DK-2200, Denmark Danish Stem Cell Centre (DanStem), Faculty of Health Sciences, University of Copenhagen, Ole Maaløes Vej 5, Copenhagen N DK2200, Denmark
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, 1664 North Virginia Street, MS575, Reno, NV 89557, USA
| |
Collapse
|
20
|
Bachelard E, Raucci F, Montillet G, Pain B. Identification of side population cells in chicken embryonic gonads. Theriogenology 2014; 83:377-84. [PMID: 25447150 DOI: 10.1016/j.theriogenology.2014.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/23/2014] [Accepted: 09/27/2014] [Indexed: 10/24/2022]
Abstract
The side population (SP) phenotype, defined by the ability of a cell to efflux fluorescent dyes such as Hoechst, is common to several stem/progenitor cell types. In avian species, SP phenotype has been identified in pubertal and adult testes, but nothing is known about its expression during prenatal development of a male gonad. In this study, we characterized the Hoechst SP phenotype via the cytofluorimetric analysis of disaggregated testes on different days of chicken embryonic development. Male prenatal gonads contained a fraction of SP cells at each stage analyzed. At least two main SP fractions, named P3 and P4, were identified. The percentage of P3 fraction decreased as development proceeds, whereas P4 cell number was not affected by gonad growth. Functional inhibition of BCRP1 channel membrane using Verapamil and/or Ko143 showed that P3, but not P4 phenotype, was dependent on BCRP1 activity. Molecular analysis of both P3- and P4-sorted fractions revealed a differential RNA expression pattern, indicating that P3 cells mainly contained germinal stem cell markers, whereas P4 was preferentially composed of both Sertoli and Leydig cell progenitor markers. Finally, these findings provided evidence that the SP phenotype is a common feature of both germ and somatic cells detected in chicken developing testis.
Collapse
Affiliation(s)
- Elodie Bachelard
- INSERM, U846, Stem Cell and Brain Research Institute, Bron, France; INRA, USC1361, Bron, France; Université de Lyon, Lyon 1, UMR S 846, Lyon, France
| | - Franca Raucci
- INSERM, U846, Stem Cell and Brain Research Institute, Bron, France; INRA, USC1361, Bron, France; Université de Lyon, Lyon 1, UMR S 846, Lyon, France
| | - Guillaume Montillet
- INSERM, U846, Stem Cell and Brain Research Institute, Bron, France; INRA, USC1361, Bron, France; Université de Lyon, Lyon 1, UMR S 846, Lyon, France
| | - Bertrand Pain
- INSERM, U846, Stem Cell and Brain Research Institute, Bron, France; INRA, USC1361, Bron, France; Université de Lyon, Lyon 1, UMR S 846, Lyon, France.
| |
Collapse
|
21
|
Georges A, L'Hôte D, Todeschini AL, Auguste A, Legois B, Zider A, Veitia RA. The transcription factor FOXL2 mobilizes estrogen signaling to maintain the identity of ovarian granulosa cells. eLife 2014; 3. [PMID: 25369636 PMCID: PMC4356143 DOI: 10.7554/elife.04207] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/07/2014] [Indexed: 12/19/2022] Open
Abstract
FOXL2 is a lineage determining transcription factor in the ovary, but its direct targets and modes of action are not fully characterized. In this study, we explore the targets of FOXL2 and five nuclear receptors in murine primary follicular cells. We found that FOXL2 is required for normal gene regulation by steroid receptors, and we show that estrogen receptor beta (ESR2) is the main vector of estradiol signaling in these cells. Moreover, we found that FOXL2 directly modulates Esr2 expression through a newly identified intronic element. Interestingly, we found that FOXL2 repressed the testis-determining gene Sox9 both independently of estrogen signaling and through the activation of ESR2 expression. Altogether, we show that FOXL2 mobilizes estrogen signaling to establish a coherent feed-forward loop repressing Sox9. This sheds a new light on the role of FOXL2 in ovarian maintenance and function. DOI:http://dx.doi.org/10.7554/eLife.04207.001 In female mammals, granulosa cells in the ovaries help egg cells to grow and develop by secreting nutrients and estrogens—the female sex hormones. A protein called FOXL2 helps granulosa cells to develop and functions by binding to the DNA of the cells to switch certain genes either on or off. In humans, mutations in the gene that codes for the FOXL2 protein are associated with granulosa cell tumors and with a loss of female fertility in early adulthood. In addition, if the amount of FOXL2 is artificially reduced in granulosa cells in female mice, the cells take on many of the characteristics of supporting cells found in the testes of males. To investigate in more detail how FOXL2 works, Georges et al. grew mouse granulosa cells in the laboratory to identify the DNA sequences where FOXL2 will bind, and to uncover how this binding affects gene expression. Georges et al. conclude that FOXL2 orchestrates a network involving many different proteins that allows estrogen to be produced and used by granulosa cells; and in doing so these cells maintain their identity as ovarian cells. FOXL2 was also shown to work closely with the receptor proteins that detect the sex hormones, and which help to control whether particular sex-specific genes are switched on or off. One particularly important role of FOXL2 in granulosa cells is that it represses a gene called Sox9. By repressing Sox9, the granulosa cells do not transform into their counterparts normally found in testes. Although FOXL2 was previously reported to directly regulate the Sox9 gene, Georges et al. find that it also acts through other molecules, and that there are alternative ways in which it can do so. Although Georges et al. have established some of the ways that FOXL2 functions, this protein can work via other pathways; these will require further investigation to be fully understood. DOI:http://dx.doi.org/10.7554/eLife.04207.002
Collapse
|
22
|
Identification and characterization of germ cell genes expressed in the F9 testicular teratoma stem cell line. PLoS One 2014; 9:e103837. [PMID: 25153150 PMCID: PMC4143169 DOI: 10.1371/journal.pone.0103837] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 07/02/2014] [Indexed: 02/06/2023] Open
Abstract
The F9 cell line, which was derived from a mouse testicular teratoma that originated from pluripotent germ cells, has been used as a model for differentiation. However, it is largely unknown whether F9 cells possess the characteristics of male germ cells. In the present study, we investigated spermatogenic stage- and cell type-specific gene expression in F9 cells. Analysis of previous microarray data showed that a large number of stage-regulated germ cell genes are expressed in F9 cells. Specifically, genes that are prominently expressed in spermatogonia and have transcriptional regulatory functions appear to be enriched in F9 cells. Our in silico and in vitro analyses identified several germ cell-specific or -predominant genes that are expressed in F9 cells. Among them, strong promoter activities were observed in the regions upstream of the spermatogonial genes, Dmrt1 (doublesex and mab-3 related transcription factor 1), Stra8 (stimulated by retinoic acid gene 8) and Tex13 (testis expressed gene 13), in F9 cells. A detailed analysis of the Tex13 promoter allowed us to identify an enhancer and a region that is implicated in germ cell-specificity. We also found that Tex13 expression is regulated by DNA methylation. Finally, analysis of GFP (green fluorescent protein) TEX13 localization revealed that the protein distributes heterogeneously in the cytoplasm and nucleus, suggesting that TEX13 shuttles between these two compartments. Taken together, our results demonstrate that F9 cells express numerous spermatogonial genes and could be used for transcriptional studies focusing on such genes. As an example of this, we use F9 cells to provide comprehensive expressional information about Tex13, and report that this gene appears to encode a germ cell-specific protein that functions in the nucleus during early spermatogenesis.
Collapse
|
23
|
Lan KC, Chen YT, Chang C, Chang YC, Lin HJ, Huang KE, Kang HY. Up-regulation of SOX9 in sertoli cells from testiculopathic patients accounts for increasing anti-mullerian hormone expression via impaired androgen receptor signaling. PLoS One 2013; 8:e76303. [PMID: 24098470 PMCID: PMC3788123 DOI: 10.1371/journal.pone.0076303] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 08/23/2013] [Indexed: 01/29/2023] Open
Abstract
Background Testosterone provokes Sertoli cell maturation and represses AMH production. In adult patients with Sertoli-cells-only syndrome (SCOS) and androgen insensitivity syndrome (AIS), high level of AMH expression is detected in Sertoli cells due to defect of androgen/AR signaling. Objective We postulated that up-regulation of SOX9 due to impairment of androgen/AR signaling in Sertoli cells might explain why high level of anti-Mullerian hormone (AMH) expression occur in these testiculopathic patients. Methods Biological research of testicular specimens from men with azoospermia or mouse. The serum hormone levels were studied in 23 men with obstructive azoospermia, 33 men with SCOS azoospermia and 21 volunteers with normal seminograms during a period of 4 years. Immunohistochemical staining and reverse-transcription PCR were used to examine the relationships among AR, SOX9 and AMH expression in adult human and mouse testes. The ability of AR to repress the expression of SOX9 and AMH was evaluated in vitro in TM4 Sertoli cells and C3H10T1/2 cells. Results SCOS specimens showed up-regulation of SOX9 and AMH proteins but down-regulation of AR proteins in Sertoli cells. The mRNA levels of AR were significantly lower and the SOX9, AMH mRNA levels higher in all SCOS patients compared to controls (P< 0.05). The testosterone levels in the SCOS patients were within the normal range, but most were below the median of the controls. Furthermore, our invitro cell line experiments demonstrated that androgen/AR signaling suppressed the gene and protein levels of AMH via repression of SOX9. Conclusions Our data show that the functional androgen/AR signaling to repress SOX9 and AMH expression is essential for Sertoli cell maturation. Impairment of androgen/AR signaling promotes SOX9-mediated AMH production, accounts for impairments of Sertoli cells in SCOS azoospermic patients.
Collapse
Affiliation(s)
- Kuo-Chung Lan
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Kaohsiung, Taiwan
- Hormone Research Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yen-Ta Chen
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chawnshang Chang
- George H. Whipple Lab for Cancer Research, Departments of Pathology, Urology and Radiation Oncology, and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Yung-Chiao Chang
- Hormone Research Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsin-Jung Lin
- Hormone Research Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ko-En Huang
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Hormone Research Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hong-Yo Kang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Kaohsiung, Taiwan
- Hormone Research Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
24
|
Hazra R, Jimenez M, Desai R, Handelsman DJ, Allan CM. Sertoli cell androgen receptor expression regulates temporal fetal and adult Leydig cell differentiation, function, and population size. Endocrinology 2013; 154:3410-22. [PMID: 23766127 DOI: 10.1210/en.2012-2273] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We recently created a mouse model displaying precocious Sertoli cell (SC) and spermatogenic development induced by SC-specific transgenic androgen receptor expression (TgSCAR). Here we reveal that TgSCAR regulates the development, function, and absolute number of Leydig cells (LCs). Total fetal and adult type LC numbers were reduced in postnatal and adult TgSCAR vs control testes, despite normal circulating LH levels. Normal LC to SC ratios found in TgSCAR testes indicate that SC androgen receptor (SCAR)-mediated activity confers a quorum-dependent relationship between total SC and LC numbers. TgSCAR enhanced LC differentiation, shown by elevated ratios of advanced to immature LC types, and reduced LC proliferation in postnatal TgSCAR vs control testes. Postnatal TgSCAR testes displayed up-regulated expression of coupled ligand-receptor transcripts (Amh-Amhr2, Dhh-Ptch1, Pdgfa-Pdgfra) for potential SCAR-stimulated paracrine pathways, which may coordinate LC differentiation. Neonatal TgSCAR testes displayed normal T and dihydrotestosterone levels despite differential changes to steroidogenic gene expression, with down-regulated Star, Cyp11a1, and Cyp17a1 expression contrasting with up-regulated Hsd3b1, Hsd17b3, and Srd5a1 expression. TgSCAR males also displayed elevated postnatal and normal adult serum testosterone levels, despite reduced LC numbers. Enhanced adult-type LC steroidogenic output was revealed by increased pubertal testicular T, dihydrotestosterone, 3α-diol and 3β-diol levels per LC and up-regulated steroidogenic gene (Nr5a1, Lhr, Cyp11a1, Cyp17a1, Hsd3b6, Srd5a1) expression in pubertal or adult TgSCAR vs control males, suggesting regulatory mechanisms maintain androgen levels independently of absolute LC numbers. Our unique gain-of-function TgSCAR model has revealed that SCAR activity controls temporal LC differentiation, steroidogenic function, and population size.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Biomarkers/metabolism
- Cell Count
- Cell Differentiation
- Hemizygote
- Isoenzymes/biosynthesis
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Leydig Cells/cytology
- Leydig Cells/metabolism
- Ligands
- Male
- Mice
- Mice, Transgenic
- Patched Receptors
- Patched-1 Receptor
- Receptor, Platelet-Derived Growth Factor alpha/biosynthesis
- Receptor, Platelet-Derived Growth Factor alpha/genetics
- Receptor, Platelet-Derived Growth Factor alpha/metabolism
- Receptors, Androgen/biosynthesis
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Peptide/biosynthesis
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
- Receptors, Transforming Growth Factor beta/biosynthesis
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Sertoli Cells/cytology
- Sertoli Cells/metabolism
- Sexual Development
- Testis/cytology
- Testis/growth & development
- Testis/metabolism
- Testosterone Congeners/blood
- Testosterone Congeners/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Rasmani Hazra
- ANZAC Research Institute, Concord Hospital, Sydney, New South Wales 2139, Australia
| | | | | | | | | |
Collapse
|
25
|
Tanwar PS, Kaneko-Tarui T, Zhang L, Teixeira JM. Altered LKB1/AMPK/TSC1/TSC2/mTOR signaling causes disruption of Sertoli cell polarity and spermatogenesis. Hum Mol Genet 2012; 21:4394-405. [PMID: 22791749 DOI: 10.1093/hmg/dds272] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Male patients with Peutz-Jeghers syndrome (PJS) have defective spermatogenesis and are at increased risk of developing Sertoli cell tumors. Mutations in the Liver Kinase B1 (LKB1/STK11) gene are associated with the pathogenesis of PJS and have been identified in non-PJS patients with sporadic testicular cancers. The mechanisms controlled by LKB1 signaling in Sertoli cell functions and testicular biology have not been described. We have conditionally deleted the Lkb1 gene (Lkb1(cko)) in somatic testicular cells to define the molecular mechanisms involved in the development of the testicular phenotype observed in PJS patients. Focal vacuolization in some of the seminiferous tubules was observed in 4-week-old mutant testes but germ cell development appeared to be normal. However, similar to PJS patients, we observed progressive germ cell loss and Sertoli cell only tubules in Lkb1(cko) testes from mice older than 10 weeks, accompanied by defects in Sertoli cell polarity and testicular junctional complexes and decreased activation of the MAP/microtubule affinity regulating and focal adhesion kinases. Suppression of AMP kinase and activation of mammalian target of rapamycin (mTOR) signaling were also observed in Lkb1(cko) testes. Loss of Tsc1 or Tsc2 copies the progressive Lkb1(cko) phenotype, suggesting that dysregulated activation of mTOR contributes to the pathogenesis of the Lkb1(cko) testicular phenotype. Pten(cko) mice had a normal testicular phenotype, which could be explained by the comparative lack of mTOR activation detected. These studies describe the importance of LKB1 signaling in testicular biology and the possible molecular mechanisms driving the pathogenesis of the testicular defects observed in PJS patients.
Collapse
Affiliation(s)
- Pradeep S Tanwar
- Vincent Center For Reproductive Biology/Thier 931, Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | |
Collapse
|