1
|
Malolina EA, Galiakberova AA, Mun VV, Sabirov MS, Dashinimaev EB, Kulibin AY. A comparative analysis of genes differentially expressed between rete testis cells and Sertoli cells of the mouse testis. Sci Rep 2023; 13:20896. [PMID: 38017073 PMCID: PMC10684643 DOI: 10.1038/s41598-023-48149-7] [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: 09/06/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023] Open
Abstract
The rete testis (RT) is a region of the mammalian testis that plays an important role in testicular physiology. The RT epithelium consists of cells sharing some well-known gene markers with supporting Sertoli cells (SCs). However, little is known about the differences in gene expression between these two cell populations. Here, we used fluorescence-activated cell sorting (FACS) to obtain pure cultures of neonatal RT cells and SCs and identified differentially expressed genes (DEGs) between these cell types using RNA sequencing (RNA-seq). We then compared our data with the RNA-seq data of other studies that examined RT cells and SCs of mice of different ages and generated a list of DEGs permanently upregulated in RT cells throughout testis development and in culture, which included 86 genes, and a list of 79 DEGs permanently upregulated in SCs. The analysis of studies on DMRT1 function revealed that nearly half of the permanent DEGs could be regulated by this SC upregulated transcription factor. We suggest that useful cell lineage markers and candidate genes for the specification of both RT cells and SCs may be present among these permanent DEGs.
Collapse
Affiliation(s)
- Ekaterina A Malolina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia.
| | - Adelya A Galiakberova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | - Valery V Mun
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia
| | - Marat S Sabirov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia
| | - Erdem B Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
- Moscow Institute of Physics and Technology (State University), Institutskiy Per., 141701, Dolgoprudny, Russia
| | - Andrey Yu Kulibin
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334, Moscow, Russia
| |
Collapse
|
2
|
Wang H, Liu Z, Larsen M, Hastings R, Gunewardena S, Kumar TR. Identification of follicle-stimulating hormone-responsive genes in Sertoli cells during early postnatal mouse testis development. Andrology 2023; 11:860-871. [PMID: 37208854 DOI: 10.1111/andr.13459] [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: 12/16/2022] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND In the mouse testis, Sertoli cells rapidly divide during a narrow window of time pre-pubertally and differentiate thereafter. The number of Sertoli cells determines the testis size and germ cell-carrying capacity. Follicle-stimulating hormone (FSH) binds its cognate FSH-receptors expressed on Sertoli cells and acts as a mitogen to regulate their proliferation. Fshb-/- mutant adult male mice have reduced Sertoli cell number and testis size and reduced sperm number and motility. However, FSH-responsive genes in early postnatal mouse Sertoli cells are unknown. OBJECTIVES To identify FSH-responsive genes in early postnatal mouse Sertoli cells. MATERIALS AND METHODS A fluorescence-activated cell sorting method was developed to rapidly purify Sertoli cells from control and Fshb-/- mice carrying a Sox9 GfpKI allele. These pure Sertoli cells were used for large-scale gene expression analyses. RESULTS We show that mouse Sertoli cells rarely divide beyond postnatal day 7. Our in vivo BrdU labeling studies indicate loss of FSH results in a 30% reduction in Sertoli cell proliferation in mice at 5 days of age. Flowsorted GFP+ Sertoli cells with maximal Fshr expression were 97%-98% pure and mostly devoid of Leydig and germ cells as assessed by Taqman qPCR quantification of gene expression and immunolabeling of the corresponding cell-specific markers. Large-scale gene expression analysis identified several differentially regulated genes in flow-sorted GFP+ Sertoli cells obtained from testis of control and Fshb-/- mice at 5 days of age. The top 25 networks identified by pathway analysis include those related to the cell cycle, cell survival and most importantly, carbohydrate and lipid metabolism and molecular transport. DISCUSSION Several of the FSH-responsive genes identified in this study could serve as useful markers for Sertoli cell proliferation in normal physiology, toxicant-induced Sertoli cell/testis injury, and other pathological conditions. CONCLUSION Our studies reveal that FSH-regulates macromolecular metabolism and molecular transport networks of genes in early postnatal Sertoli cells most likely in preparation for establishment of functional associations with germ cells to successfully coordinate spermatogenesis.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Zhenghui Liu
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mark Larsen
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard Hastings
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sumedha Gunewardena
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - T Rajendra Kumar
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
3
|
Au TYK, Yip RKH, Wynn SL, Tan TY, Fu A, Geng YH, Szeto IYY, Niu B, Yip KY, Cheung MCH, Lovell-Badge R, Cheah KSE. Hypomorphic and dominant-negative impact of truncated SOX9 dysregulates Hedgehog-Wnt signaling, causing campomelia. Proc Natl Acad Sci U S A 2023; 120:e2208623119. [PMID: 36584300 PMCID: PMC9910594 DOI: 10.1073/pnas.2208623119] [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: 06/10/2022] [Accepted: 11/02/2022] [Indexed: 01/01/2023] Open
Abstract
Haploinsufficiency for SOX9, the master chondrogenesis transcription factor, can underlie campomelic dysplasia (CD), an autosomal dominant skeletal malformation syndrome, because heterozygous Sox9 null mice recapitulate the bent limb (campomelia) and some other phenotypes associated with CD. However, in vitro cell assays suggest haploinsufficiency may not apply for certain mutations, notably those that truncate the protein, but in these cases in vivo evidence is lacking and underlying mechanisms are unknown. Here, using conditional mouse mutants, we compared the impact of a heterozygous Sox9 null mutation (Sox9+/-) with the Sox9+/Y440X CD mutation that truncates the C-terminal transactivation domain but spares the DNA-binding domain. While some Sox9+/Y440X mice survived, all Sox9+/- mice died perinatally. However, the skeletal defects were more severe and IHH signaling in developing limb cartilage was significantly enhanced in Sox9+/Y440X compared with Sox9+/-. Activating Sox9Y440X specifically in the chondrocyte-osteoblast lineage caused milder campomelia, and revealed cell- and noncell autonomous mechanisms acting on chondrocyte differentiation and osteogenesis in the perichondrium. Transcriptome analyses of developing Sox9+/Y440X limbs revealed dysregulated expression of genes for the extracellular matrix, as well as changes consistent with aberrant WNT and HH signaling. SOX9Y440X failed to interact with β-catenin and was unable to suppress transactivation of Ihh in cell-based assays. We propose enhanced HH signaling in the adjacent perichondrium induces asymmetrically localized excessive perichondrial osteogenesis resulting in campomelia. Our study implicates combined haploinsufficiency/hypomorphic and dominant-negative actions of SOX9Y440X, cell-autonomous and noncell autonomous mechanisms, and dysregulated WNT and HH signaling, as the cause of human campomelia.
Collapse
Affiliation(s)
- Tiffany Y. K. Au
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Raymond K. H. Yip
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Sarah L. Wynn
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Tiong Y. Tan
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Alex Fu
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, New Territories, Shatin, Hong Kong SAR, China
| | - Yu Hong Geng
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Irene Y. Y. Szeto
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Ben Niu
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Kevin Y. Yip
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, New Territories, Shatin, Hong Kong SAR, China
| | - Martin C. H. Cheung
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | | | - Kathryn S. E. Cheah
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| |
Collapse
|
4
|
Chan CJ, Hirashima T. Tissue hydraulics in reproduction. Semin Cell Dev Biol 2022; 131:124-133. [PMID: 35606275 DOI: 10.1016/j.semcdb.2022.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
Abstract
The development of functional eggs and sperm are critical processes in mammalian development as they ensure successful reproduction and species propagation. While past studies have identified important genes that regulate these processes, the roles of luminal flow and fluid stress in reproductive biology remain less well understood. Here, we discuss recent evidence that support the diverse functions of luminal fluid in oogenesis, spermatogenesis and embryogenesis. We also review emerging techniques that allow for precise quantification and perturbation of tissue hydraulics in female and male reproductive systems, and propose new questions and approaches in this field. We hope this review will provide a useful resource to inspire future research in tissue hydraulics in reproductive biology and diseases.
Collapse
Affiliation(s)
- Chii Jou Chan
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore.
| | - Tsuyoshi Hirashima
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; The Hakubi Center/Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Japan Science and Technology Agency, PRESTO, Kawaguchi, Japan.
| |
Collapse
|
5
|
Petrusová J, Manning J, Kubovčiak J, Kolář M, Filipp D. Two complementary approaches for efficient isolation of Sertoli cells for transcriptomic analysis. Front Cell Dev Biol 2022; 10:972017. [PMID: 36158203 PMCID: PMC9495933 DOI: 10.3389/fcell.2022.972017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Sertoli cells (SCs) are the only somatic cells that reside in seminiferous tubules of testis. They directly interact with and support the development of germ cells, thus have an indispensable role in the process of spermatogenesis. SCs first appear in a proliferative state and then, with the initiation of the first wave of spermatogenesis, progress to a mature “nurturing” state which supports lifelong continuous sperm production. During this development, the SC transcriptome must adapt rapidly as obstacles in SC maturation often result in deficiencies in male fertility. Due to its importance in spermatogenesis, a reliable, rapid, and precise method for the isolation of high purity, viable and unadulterated SC has been largely missing. We have developed an improved method for the preparation of a testicular single cell suspension comprised of two alternative protocols to separate SCs from the rest of the testicular cells by FACS. The first sorting scheme is based on their co-expression of surface specific markers, FSHr and Occludin-1, while the second focuses on the co-staining of SCs with FSHr-specific antibody and Hoechst 33342, which discriminates DNA content of testicular cells. The entire procedure can be completed in less than 3 h which permits the analysis of the development-related transcriptional profile of these cells. Notably, our comparative study showed that this method resulted in a SC transcriptome that is largely comparable to SCs which were briskly isolated due to their cell-specific expression of fluorescent protein. Interestingly, we also show that SCs sorted as FSHr+Occludin+ cells contained a tangible portion of transcripts from all types of testicular germ cells. Sorting of SCs according to their 2C DNA content significantly reduced the presence of these transcripts, thus seems to be the most suitable approach for accurate determination of the SC transcriptome. We believe that these novel approaches for the isolation of SCs will assist researchers in the elucidation of their function as well as their role in spermatogenesis and disorders related to male infertility.
Collapse
Affiliation(s)
- Jana Petrusová
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jasper Manning
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jan Kubovčiak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Dominik Filipp
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
- *Correspondence: Dominik Filipp,
| |
Collapse
|
6
|
Live imaging approach of dynamic multicellular responses in ERK signaling during vertebrate tissue development. Biochem J 2022; 479:129-143. [PMID: 35050327 PMCID: PMC8883488 DOI: 10.1042/bcj20210557] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022]
Abstract
The chemical and mechanical responses of cells via the exchange of information during growth and development result in the formation of biological tissues. Information processing within the cells through the signaling pathways and networks inherent to the constituent cells has been well-studied. However, the cell signaling mechanisms responsible for generating dynamic multicellular responses in developing tissues remain unclear. Here, I review the dynamic multicellular response systems during the development and growth of vertebrate tissues based on the extracellular signal-regulated kinase (ERK) pathway. First, an overview of the function of the ERK signaling network in cells is provided, followed by descriptions of biosensors essential for live imaging of the quantification of ERK activity in tissues. Then adducing four examples, I highlight the contribution of live imaging techniques for studying the involvement of spatio-temporal patterns of ERK activity change in tissue development and growth. In addition, theoretical implications of ERK signaling are also discussed from the viewpoint of dynamic systems. This review might help in understanding ERK-mediated dynamic multicellular responses and tissue morphogenesis.
Collapse
|
7
|
Kulibin AY, Malolina EA. The Rete Testis: Development and Role in Testis Function. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421060072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Abstract
The rete testis connects seminiferous tubules in which germ cells develop to the efferent ducts and the epididymis, where gametes mature and gain mobility. Several recent studies have thoroughly explored the morphogenesis of this structure in mice during embryonic and postnatal periods. A part of the rete testis has been shown to derive from the precursors of gonad somatic cells before sex determination. The other part forms from embryonal Sertoli cells of testis cords adjacent to the mesonephros. The transformation of Sertoli cells into rete testis cells is apparently not limited to the embryonic stage of development and continues during postnatal testis development. Recently, it was found that the rete testis participates in the formation and maintenance of specialized Sertoli cells in terminal segments of seminiferous tubules, transitional zones. Current views suggest that the transitional zones of the seminiferous tubules may represent a niche for spermatogonial stem cells, the site of the prolonged proliferation of Sertoli cells in the pubertal and postpubertal periods of testis development, and also could be a generator of spermatogenic waves. To sum up, the rete testis transports gametes from the testis to the epididymis, maintains pressure within seminiferous tubules, regulates the composition of the testicular fluid, and impacts the spermatogenic process itself.
Collapse
|
8
|
Nakata H, Iseki S, Mizokami A. Three-dimensional reconstruction of testis cords/seminiferous tubules. Reprod Med Biol 2021; 20:402-409. [PMID: 34646067 PMCID: PMC8499590 DOI: 10.1002/rmb2.12413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Due to the development of novel equipment for the acquisition of two-dimensional serial images and software capable of displaying three-dimensional (3D) images from serial images, the accurate 3D reconstruction of organs and tissues has become possible. METHODS Based on published studies, this review summarizes techniques for the 3D reconstruction of the testis cords/seminiferous tubules, with special reference to our method using serial paraffin sections and 3D visualization software. MAIN FINDINGS The testes of mice, rats, and hamsters of various ages were 3D reconstructed and species and age differences in the structures of the testis cords/seminiferous tubules were analyzed. Our method is advantageous because conventional paraffin-embedded normal and pathological specimens may be utilized for the 3D analysis without the need for complicated and expensive equipment. CONCLUSION By further decreasing the time and labor required for the procedure and adding information on molecular localization, the technique for 3D reconstruction will contribute to the elucidation of not only the structures, but also the functions of various organs, including the testis.
Collapse
Affiliation(s)
- Hiroki Nakata
- Department of Histology and Cell Biology Graduate School of Medical Sciences Kanazawa University Kanazawa Japan
| | - Shoichi Iseki
- Department of Clinical Engineering Faculty of Health Sciences Komatsu University Komatsu Japan
| | - Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology Kanazawa University Graduate School of Medical Science Kanazawa Japan
| |
Collapse
|
9
|
Omotehara T, Nakata H, Itoh M. Three-dimensional analysis of mesonephric tubules remodeling into efferent tubules in the male mouse embryo. Dev Dyn 2021; 251:513-524. [PMID: 34378268 DOI: 10.1002/dvdy.410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Spermatozoa are transported to the epididymal duct through efferent tubules. Although the origin of the efferent tubules is thought to be mesonephric tubules (MTs), their detailed developmental process, for example, where the rete testis and efferent tubules are connected, is unclear. We investigated the structural changes of the MTs in the male mouse embryo using a three-dimensional reconstruction method. RESULTS Three to six MTs were connected to the Wolffian duct, and some of them branched, resulting in five to nine tips. Rete cells contacted the three to six tips. The MTs showed a folded shape when the gonadal fate was determined. After the testis development started, they became short and straight but emerged as long and twisting by birth. Before birth, the efferent duct was composed of MTs and a cranial portion of the folded Wolffian duct. CONCLUSIONS The contact between the rete testis and efferent tubules is possibly established at the tip of each MT. The MTs regress after gonadal fate is determined but is remodeled to the twisting efferent tubules by birth. The efferent tubules are composed of the MTs but also a cranial portion of the folded Wolffian duct in the mouse.
Collapse
Affiliation(s)
| | - Hiroki Nakata
- Department of Histology and Cell Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Masahiro Itoh
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
10
|
Nakata H, Omotehara T, Itoh M, Iseki S, Mizokami A. Three-dimensional structure of testis cords in mice and rats. Andrology 2021; 9:1911-1922. [PMID: 34128333 DOI: 10.1111/andr.13069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/01/2021] [Accepted: 06/11/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Testis cord elongation and coiling, which occur in the final stage of testis formation, have been attributed to Sertoli cell proliferation; however, the underlying mechanisms remain unclear. OBJECTIVE The aim of the present study was to clarify the precise three-dimensional structure of testis cords in the final stage of testis formation in mice and rats. MATERIALS AND METHODS We reconstructed whole testis cords in the final stage of testis formation in mice (on embryonic days 15.5 and 18.5) and rats (on embryonic days 16.5 and 19.5) using serial paraffin sections and high-performance three-dimensional reconstruction software. RESULTS Detailed morphometric parameters were calculated for three-dimensionally reconstructed testis cords in six mouse and rat testes each. The mean numbers of testis cords in mice and rats were 12.7 and 27.8, respectively. The mean number of branching points per testis cord was 1.52 in mice, whereas it was only 0.30 in rats. In contrast, the mean ratio of the inner cords, that is, cords not in contact with the tunica albuginea, was 23.0% in rats, whereas it was only 6.5% in mice. In both species, the cords on the cranial side coiled more strongly than those on the caudal side, consistent with the greater expansion of the testis volume on the caudal side. All cords formed right-handed helices from the rete testis side. DISCUSSION AND CONCLUSIONS The present results suggest that testis cords undergo anastomosis at a higher frequency in mice than in rats and that the coiling of testis cords proceeds from the cranial to caudal side of the testis in both species.
Collapse
Affiliation(s)
- Hiroki Nakata
- Department of Histology and Cell Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | | | - Masahiro Itoh
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Shoichi Iseki
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Komatsu, Japan
| | - Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
11
|
Higuch K, Matsumura T, Akiyama H, Kanai Y, Ogawa T, Sato T. Sertoli cell replacement in explanted mouse testis tissue supporting host spermatogenesis. Biol Reprod 2021; 105:934-943. [PMID: 34057178 DOI: 10.1093/biolre/ioab104] [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: 12/03/2020] [Revised: 03/25/2021] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
Spermatogenesis takes place in the seminiferous tubules, starting from the spermatogonial stem cell and maturing into sperm through multiple stages of cell differentiation. Sertoli cells, the main somatic cell constituting the seminiferous tubule, are in close contact with every germ cell and play pivotal roles in the progression of spermatogenesis. In this study, we developed an in vitro Sertoli cell replacement method by combining an organ culture technique and a toxin receptor-mediated cell knockout (Treck) system. We used Amh- diphtheria toxin receptor (DTR) transgenic mice, whose Sertoli cells specifically express human DTR, which renders them sensitive to diphtheria toxin (DT). An immature Amh-DTR testis was transplanted with donor testis cells followed by culturing in a medium containing DT. This procedure successfully replaced the original Sertoli cells with the transplanted Sertoli cells, and spermatogenesis originating from resident germ cells was confirmed. In addition, Sertoli cells in the mouse testis tissues were replaced by transplanted rat Sertoli cells within culture conditions, without requiring immunosuppressive treatments. This method works as a functional assay system, making it possible to evaluate any cells that might function as Sertoli cells. It would also be possible to investigate interactions between Sertoli and germ cells more closely, providing a new platform for the study of spermatogenesis and its impairments.
Collapse
Affiliation(s)
- Kazusa Higuch
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Yokohama City University Association of Medical Science, Yokohama 236-0004, Japan
| | - Takafumi Matsumura
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Yokohama City University Association of Medical Science, Yokohama 236-0004, Japan
| | - Haruhiko Akiyama
- Department of Orthopedics, Gifu University School of Medicine, Gifu, Japan
| | - Yoshiakira Kanai
- Department of Veterinary Anatomy, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takehiko Ogawa
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Yokohama City University Association of Medical Science, Yokohama 236-0004, Japan.,Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Takuya Sato
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Yokohama City University Association of Medical Science, Yokohama 236-0004, Japan
| |
Collapse
|
12
|
Frost ER, Taylor G, Baker MA, Lovell-Badge R, Sutherland JM. Establishing and maintaining fertility: the importance of cell cycle arrest. Genes Dev 2021; 35:619-634. [PMID: 33888561 PMCID: PMC8091977 DOI: 10.1101/gad.348151.120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this review, Frost et al. summarize the current knowledge on the Cip/Kip family of cyclin-dependent kinase inhibitors in mouse gonad development and highlight new roles for cell cycle inhibitors in controlling and maintaining female fertility. Development of the ovary or testis is required to establish reproductive competence. Gonad development relies on key cell fate decisions that occur early in embryonic development and are actively maintained. During gonad development, both germ cells and somatic cells proliferate extensively, a process facilitated by cell cycle regulation. This review focuses on the Cip/Kip family of cyclin-dependent kinase inhibitors (CKIs) in mouse gonad development. We particularly highlight recent single-cell RNA sequencing studies that show the heterogeneity of cyclin-dependent kinase inhibitors. This diversity highlights new roles for cell cycle inhibitors in controlling and maintaining female fertility.
Collapse
Affiliation(s)
- Emily R Frost
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia.,Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Güneş Taylor
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Mark A Baker
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia
| | - Robin Lovell-Badge
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Jessie M Sutherland
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia
| |
Collapse
|
13
|
McClusky LM. Simple, once-off mapping of various, recurrent immunostaining patterns of proliferating cell nuclear antigen in spermatogonia at the immature pole of the testis of adult wild-caught blue shark, Prionace glauca: Correlations with changes in testicular status. Mol Reprod Dev 2020; 87:1111-1123. [PMID: 33104292 DOI: 10.1002/mrd.23429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 09/11/2020] [Accepted: 10/05/2020] [Indexed: 12/28/2022]
Abstract
This study was a single time-point mapping of various immunostaining patterns revealed with the proliferating cell nuclear antigen (PCNA) PC10 antibody in spermatogonia at the immature pole of the testis of the Blue shark (Prionace glauca). Scattered in the stroma of the germinal ridge that demarcates the immature pole's outer boundary were nests of variously immunoreactive A-spermatogonia, each flanked by a fusiform cell. Spermatocysts were assembled from niche-derived stromal cells, displaced A-progenitors, and their progeny, which showed one of two main immunostaining patterns (i.e., an uneven light brown/globular and homogeneous dark [hod] brown appearance). The testes of wild-caught Prionace showed two conditions, namely, extensive multinucleate cell death (MNC) near the mitosis-meiosis transition or an early recovery phase from the latter showing vacuolated areas. Both the proportion of cysts with immature Bhod -spermatogonia and the frequency of mitotic figures in such cysts in the early recovery testis condition were significantly higher than the comparable parameters in MNC testis condition. Moreover, the post-MNC recovery phase revealed a decrease in the proportion of immature cysts with uneven light brown/globular-like spermatogonia. The protracted spread of a cell cycle signal in an anatomically discrete, syncytially connected spermatogonial clone manifests as different PCNA immunoreactivities.
Collapse
Affiliation(s)
- Leon M McClusky
- Department of Health and Care, UiT The Arctic University of Norway, Narvik, Norway
| |
Collapse
|
14
|
Yoshida T, Matsuda M, Hirashima T. Incoherent Feedforward Regulation via Sox9 and ERK Underpins Mouse Tracheal Cartilage Development. Front Cell Dev Biol 2020; 8:585640. [PMID: 33195234 PMCID: PMC7642454 DOI: 10.3389/fcell.2020.585640] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/15/2020] [Indexed: 11/13/2022] Open
Abstract
Tracheal cartilage provides architectural integrity to the respiratory airway, and defects in this structure during embryonic development cause severe congenital anomalies. Previous genetic studies have revealed genes that are critical for the development of tracheal cartilage. However, it is still unclear how crosstalk between these proteins regulates tracheal cartilage formation. Here we show a core regulatory network underlying murine tracheal chondrogenesis from embryonic day (E) 12.5 to E15.5, by combining volumetric imaging of fluorescence reporters, inhibitor assays, and mathematical modeling. We focused on SRY-box transcription factor 9 (Sox9) and extracellular signal-regulated kinase (ERK) in the tracheal mesenchyme, and observed a synchronous, inverted U-shaped temporal change in both Sox9 expression and ERK activity with a peak at E14.5, whereas the expression level of downstream cartilage matrix genes, such as collagen II alpha 1 (Col2a1) and aggrecan (Agc1), monotonically increased. Inhibitor assays revealed that the ERK signaling pathway functions as an inhibitory regulator of tracheal cartilage differentiation during this period. These results suggest that expression of the cartilage matrix genes is controlled by an incoherent feedforward loop via Sox9 and ERK, which is supported by a mathematical model. Furthermore, the modeling analysis suggests that a Sox9-ERK incoherent feedforward regulation augments the robustness against the variation of upstream factors. The present study provides a better understanding of the regulatory network underlying the tracheal development and will be helpful for efficient induction of tracheal organoids.
Collapse
Affiliation(s)
- Takuya Yoshida
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Michiyuki Matsuda
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.,Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Hirashima
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Japan Science and Technology Agency, PRESTO, Tokyo, Japan
| |
Collapse
|
15
|
Kulibin AY, Malolina EA. Formation of the rete testis during mouse embryonic development. Dev Dyn 2020; 249:1486-1499. [DOI: 10.1002/dvdy.242] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/18/2020] [Accepted: 08/22/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Andrey Yu. Kulibin
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences Moscow Russia
| | - Ekaterina A. Malolina
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences Moscow Russia
| |
Collapse
|
16
|
Ortega EA, Salvador Q, Fernandez M, Ward MA. Alterations of sex determination pathways in the genital ridges of males with limited Y chromosome genes†. Biol Reprod 2020; 100:810-823. [PMID: 30285093 DOI: 10.1093/biolre/ioy218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/09/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022] Open
Abstract
We previously demonstrated that in the mouse only two Y chromosome genes are required for a male to produce an offspring with the help of assisted reproduction technologies (ART): testis determinant Sry and spermatogonial proliferation factor Eif2s3y. Subsequently, we have shown that the function of these genes can be replaced by transgenic overexpression of their homologs, autosomally encoded Sox9 and X-chromosome encoded Eif2s3x. Males with Y chromosome contribution limited to two (XEif2s3yOSry), one (XEif2s3yOSox9 and XOSry,Eif2s3x), and no genes (XOSox9,Eif2s3x) produced haploid germ cells and sired offspring after ART. However, despite successful assisted reproductive outcome, they had smaller testes and displayed abnormal development of the seminiferous epithelium and testicular interstitium. Here we explored whether these testicular defects originated from altered pro-testis and pro-ovary factor signaling in genital ridges at the time of sex determination. Timed pregnancies were generated to obtain transgenic XEif2s3yOSry, XEif2s3yOSox9, XOSry,Eif2s3x, XOSox9,Eif2s3x, and wild-type XX and XY fetuses at 12.5 days post coitum. Dissected genital ridges were assessed for their morphology and anatomy, and expression of pro-testis and pro-ovary transcripts. All transgenic males displayed incomplete masculinization of gonadal shape, impaired development of testicular cords and gonadal vasculature, and decreased expression of factors promoting male pathway. Fetal gonad masculinization was more effective when sex determination was driven by the Sry transgene, in the presence of Y chromosome genes, and to a lesser extent a double dosage of X genes. The study adds to the understanding of the role of Y chromosome genes and their homologs during sex determination.
Collapse
Affiliation(s)
- Eglė A Ortega
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Quinci Salvador
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Mayumi Fernandez
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Monika A Ward
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| |
Collapse
|
17
|
Omotehara T, Wu X, Kuramasu M, Itoh M. Connection between seminiferous tubules and epididymal duct is originally induced before sex differentiation in a sex‐independent manner. Dev Dyn 2020; 249:754-764. [DOI: 10.1002/dvdy.155] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/03/2020] [Accepted: 01/07/2020] [Indexed: 12/31/2022] Open
Affiliation(s)
- Takuya Omotehara
- Department of AnatomyTokyo Medical University Shinjuku‐ku, Tokyo Japan
| | - Xi Wu
- Department of AnatomyTokyo Medical University Shinjuku‐ku, Tokyo Japan
| | - Miyuki Kuramasu
- Department of AnatomyTokyo Medical University Shinjuku‐ku, Tokyo Japan
| | - Masahiro Itoh
- Department of AnatomyTokyo Medical University Shinjuku‐ku, Tokyo Japan
| |
Collapse
|
18
|
Mäkelä JA, Koskenniemi JJ, Virtanen HE, Toppari J. Testis Development. Endocr Rev 2019; 40:857-905. [PMID: 30590466 DOI: 10.1210/er.2018-00140] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/17/2018] [Indexed: 12/28/2022]
Abstract
Production of sperm and androgens is the main function of the testis. This depends on normal development of both testicular somatic cells and germ cells. A genetic program initiated from the Y chromosome gene sex-determining region Y (SRY) directs somatic cell specification to Sertoli cells that orchestrate further development. They first guide fetal germ cell differentiation toward spermatogenic destiny and then take care of the full service to spermatogenic cells during spermatogenesis. The number of Sertoli cells sets the limits of sperm production. Leydig cells secrete androgens that determine masculine development. Testis development does not depend on germ cells; that is, testicular somatic cells also develop in the absence of germ cells, and the testis can produce testosterone normally to induce full masculinization in these men. In contrast, spermatogenic cell development is totally dependent on somatic cells. We herein review germ cell differentiation from primordial germ cells to spermatogonia and development of the supporting somatic cells. Testicular descent to scrota is necessary for normal spermatogenesis, and cryptorchidism is the most common male birth defect. This is a mild form of a disorder of sex differentiation. Multiple genetic reasons for more severe forms of disorders of sex differentiation have been revealed during the last decades, and these are described along with the description of molecular regulation of testis development.
Collapse
Affiliation(s)
- Juho-Antti Mäkelä
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jaakko J Koskenniemi
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Helena E Virtanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| |
Collapse
|
19
|
Nicol B, Grimm SA, Gruzdev A, Scott GJ, Ray MK, Yao HHC. Genome-wide identification of FOXL2 binding and characterization of FOXL2 feminizing action in the fetal gonads. Hum Mol Genet 2019; 27:4273-4287. [PMID: 30212841 DOI: 10.1093/hmg/ddy312] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/30/2018] [Indexed: 12/16/2022] Open
Abstract
The identity of the gonads is determined by which fate, ovarian granulosa cell or testicular Sertoli cell, the bipotential somatic cell precursors choose to follow. In most vertebrates, the conserved transcription factor FOXL2 contributes to the fate of granulosa cells. To understand FOXL2 functions during gonad differentiation, we performed genome-wide analysis of FOXL2 chromatin occupancy in fetal ovaries and established a genetic mouse model that forces Foxl2 expression in the fetal testis. When FOXL2 was ectopically expressed in the somatic cell precursors in the fetal testis, FOXL2 was sufficient to repress Sertoli cell differentiation, ultimately resulting in partial testis-to-ovary sex-reversal. Combining genome-wide analysis of FOXL2 binding in the fetal ovary with transcriptomic analyses of our Foxl2 gain-of-function and previously published Foxl2 loss-of-function models, we identified potential pathways responsible for the feminizing action of FOXL2. Finally, comparison of FOXL2 genome-wide occupancy in the fetal ovary with testis-determining factor SOX9 genome-wide occupancy in the fetal testis revealed extensive overlaps, implying that antagonistic signals between FOXL2 and SOX9 occur at the chromatin level.
Collapse
Affiliation(s)
- Barbara Nicol
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Sara A Grimm
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Artiom Gruzdev
- Knockout Mouse Core Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Greg J Scott
- Knockout Mouse Core Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Manas K Ray
- Knockout Mouse Core Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Humphrey H-C Yao
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
20
|
Piprek RP, Kloc M, Kubiak JZ. Matrix metalloproteinase-dependent regulation of extracellular matrix shapes the structure of sexually differentiating mouse gonads. Differentiation 2019; 106:23-34. [PMID: 30852470 DOI: 10.1016/j.diff.2019.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/24/2018] [Accepted: 01/18/2019] [Indexed: 11/19/2022]
Abstract
The extracellular matrix (ECM) proteins play an important role in the establishment of the sex-dependent structure of developing gonads. The matrix metalloproteinases (MMPs) are the major players in the regulation of ECM. Our hypothesis was that the MMPs-dependent regulation of EMC is crucial for the establishment of the correct, either testis or ovary, structure of developing gonad. We cultured developing mouse gonads in vitro in the presence of the MMPs inhibitors (α-2-macroglobulin, leupeptin, phosphoramidon) or the MMPs activator, APMA (4-aminophenylmercuric acetate). These inhibitors and activator inhibit/activate, to a different degree, matrix metalloproteinases, but the exact mechanism of inhibition/activation remains unknown. We found that the MMP inhibitors increased accumulation of ECM in the developing gonads. The α-2-macroglobulin had the weakest, and the phosphoramidon the strongest effect on the ECM and the structure of the gonads. The α-2-macroglobulin caused a slight increase of ECM and did not disrupt the gonad structure. Leupeptin led to the strong accumulation of ECM, resulted in the formation of the structures resembling testis cords in both testes and ovaries, and caused increase of apoptosis and complete loss of germ cells. Phosphoramidon caused the strongest accumulation of ECM, which separated individual cells and completely prevented intercellular adhesion both in the testes and in the ovaries. As a result of aberrant morphology, the sex of the phosphoramidon-treated gonads was morphologically unrecognizable. The APMA - the activator of MMP caused ECM loss, which led to the loss of cell adhesion, cell dispersion and an aberrant morphology of the gonads. These results indicate that the ECM accumulation is MMPs-dependent and that the correct amount and distribution of ECM during gonad development plays a key role in the formation of the gonad structure.
Collapse
Affiliation(s)
- Rafal P Piprek
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland.
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA; Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA; University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jacek Z Kubiak
- Univ Rennes, CNRS, Institute of Genetics and Development of Rennes, UMR 6290, Cell Cycle Group, Faculty of Medicine, F-35000, Rennes, France; Laboratory of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), Warsaw, Poland
| |
Collapse
|
21
|
Neirijnck Y, Kühne F, Mayère C, Pavlova E, Sararols P, Foti M, Atanassova N, Nef S. Tumor Suppressor PTEN Regulates Negatively Sertoli Cell Proliferation, Testis Size, and Sperm Production In Vivo. Endocrinology 2019; 160:387-398. [PMID: 30576429 DOI: 10.1210/en.2018-00892] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/17/2018] [Indexed: 11/19/2022]
Abstract
The IGFs are the major intratesticular factors regulating immature Sertoli cell proliferation and are, therefore, critical to establish the magnitude of sperm production. However, the intratesticular source of IGF production and the downstream signaling pathway mediating IGF-dependent Sertoli cell proliferation remain unclear. Single-cell RNA sequencing on mouse embryonic testis revealed a robust expression of Igf1 and Igf2 in interstitial steroidogenic progenitors, suggesting that IGFs exert paracrine actions on immature Sertoli cells. To elucidate the intracellular signaling mechanism that underlies the proliferative effects of IGFs on immature Sertoli cells, we have generated mice with Sertoli cell-specific deletion of the Pten gene, a negative regulator of the phosphatidylinositol-3 kinase (PI3K)/AKT pathway, alone or together with the insulin receptor (Insr) and the IGF1 receptor (Igf1r). Although ablation of Pten appears dispensable for Sertoli cell proliferation and spermatogenesis, inactivation of Pten in the absence of Insr and Igf1r rescued the Sertoli cell proliferation rate during late fetal development, testis size, and sperm production. Overall, these findings suggest that IGFs secreted by interstitial progenitor cells act in a paracrine fashion to promote the proliferation of immature Sertoli cells through the IGF/PTEN/PI3K pathway.
Collapse
Affiliation(s)
- Yasmine Neirijnck
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Françoise Kühne
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Chloé Mayère
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ekaterina Pavlova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Pauline Sararols
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Michelangelo Foti
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nina Atanassova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Serge Nef
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
22
|
Gregoire EP, Stevant I, Chassot AA, Martin L, Lachambre S, Mondin M, de Rooij DG, Nef S, Chaboissier MC. NRG1 signalling regulates the establishment of Sertoli cell stock in the mouse testis. Mol Cell Endocrinol 2018; 478:17-31. [PMID: 30040984 DOI: 10.1016/j.mce.2018.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/31/2018] [Accepted: 07/08/2018] [Indexed: 10/28/2022]
Abstract
Testis differentiation requires high levels of proliferation of progenitor cells that give rise to two cell lineages forming the testis, the Sertoli and the Leydig cells. Hence defective cell cycling leads to testicular dysgenesis that has profound effects on androgen production and fertility. The growth factor NRG1 has been implicated in adult Leydig cell proliferation, but a potential function in the fetal testis has not been analysed to date. Here we show that Nrg1 and its receptors ErbB2/3 are already expressed in early gonadal development. Using tissue-specific deletion, we further demonstrate that Nrg1 is required in a dose-dependent manner to induce proliferation of Sertoli progenitor cells and then differentiated Sertoli cells. As a result of reduced numbers of Sertoli cells, Nrg1 knockout mice display a delay in testis differentiation and defects in sex cord partitioning. Taken together Nrg1 signalling is essential for the establishment of the stock of Sertoli cells and thus required to prevent testicular hypoplasia.
Collapse
Affiliation(s)
| | - Isabelle Stevant
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Switzerland
| | | | - Luc Martin
- Université Côte d'Azur, CNRS, Inserm, iBV, France
| | | | | | - Dirk G de Rooij
- Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands
| | - Serge Nef
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Switzerland
| | | |
Collapse
|
23
|
Rotgers E, Jørgensen A, Yao HHC. At the Crossroads of Fate-Somatic Cell Lineage Specification in the Fetal Gonad. Endocr Rev 2018; 39:739-759. [PMID: 29771299 PMCID: PMC6173476 DOI: 10.1210/er.2018-00010] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/09/2018] [Indexed: 01/07/2023]
Abstract
The reproductive endocrine systems are vastly different between males and females. This sexual dimorphism of the endocrine milieu originates from sex-specific differentiation of the somatic cells in the gonads during fetal life. Most gonadal somatic cells arise from the adrenogonadal primordium. After separation of the adrenal and gonadal primordia, the gonadal somatic cells initiate sex-specific differentiation during gonadal sex determination with the specification of the supporting cell lineages: Sertoli cells in the testis vs granulosa cells in the ovary. The supporting cell lineages then facilitate the differentiation of the steroidogenic cell lineages, Leydig cells in the testis and theca cells in the ovary. Proper differentiation of these cell types defines the somatic cell environment that is essential for germ cell development, hormone production, and establishment of the reproductive tracts. Impairment of lineage specification and function of gonadal somatic cells can lead to disorders of sexual development (DSDs) in humans. Human DSDs and processes for gonadal development have been successfully modeled using genetically modified mouse models. In this review, we focus on the fate decision processes from the initial stage of formation of the adrenogonadal primordium in the embryo to the maintenance of the somatic cell identities in the gonads when they become fully differentiated in adulthood.
Collapse
Affiliation(s)
- Emmi Rotgers
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, North Carolina
| | - Anne Jørgensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,International Research and Research Training Center in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen, Denmark
| | - Humphrey Hung-Chang Yao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, North Carolina
| |
Collapse
|
24
|
Kojima K, Nakamura H, Komeya M, Yamanaka H, Makino Y, Okada Y, Akiyama H, Torikai N, Sato T, Fujii T, Kimura H, Ogawa T. Neonatal testis growth recreated in vitro by two-dimensional organ spreading. Biotechnol Bioeng 2018; 115:3030-3041. [PMID: 30144353 PMCID: PMC6283240 DOI: 10.1002/bit.26822] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 07/27/2018] [Accepted: 08/23/2018] [Indexed: 01/15/2023]
Abstract
Organ culture experiments can be hampered by central degeneration or necrosis due to the inadequate permeation of oxygen and nutrients, which deteriorates the function and growth of cultured tissues. In the current study, we aimed to overcome this limitation of organ culture through spreading the tissue two dimensionally on an agarose gel stand and molding into a disc shape by placing a ceiling of polydimethylsiloxane (PDMS) chip, which is highly oxygen permeable. By this, every part of the tissue can receive a sufficient supply of oxygen through PDMS as well as nutrients through the agarose gel below. This method not only prevented central necrosis of tissues, but also supported the tissue growth over time. In addition, such growth, as volume enlargement, could be easily measured. Under these conditions, we examined the effect of several factors on the growth of neonatal mouse testis, and found that follicle stimulating hormone (FSH) and insulin significantly promoted the growth. These results are in good agreement with previous in vivo reports. Notably, the growth achieved over 7 days in our in vitro system is almost comparable to, about 80% of, that observed in vivo. Thus, we successfully monitored the promotion of tissue growth beyond the limits of the conventional organ culture method. This extremely simple method could offer a unique platform to evaluate the growth as well as functional properties of organs, not only the testis but also others as well.
Collapse
Affiliation(s)
- Kazuaki Kojima
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Association of Medical Science, Yokohama City University, Yokohama, Japan
| | - Hiroko Nakamura
- Department of Mechanical Engineering, Tokai University, Hiratsuka, Japan
| | - Mitsuru Komeya
- Department of Urology, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Hiroyuki Yamanaka
- Department of Urology, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yoshinori Makino
- Laboratory of Pathology and Development, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Yuki Okada
- Laboratory of Pathology and Development, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo, Japan
| | - Haruhiko Akiyama
- Department of Orthopaedic Surgery, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Nobuhito Torikai
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Association of Medical Science, Yokohama City University, Yokohama, Japan
| | - Takuya Sato
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Association of Medical Science, Yokohama City University, Yokohama, Japan
| | - Teruo Fujii
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Hiroshi Kimura
- Department of Mechanical Engineering, Tokai University, Hiratsuka, Japan
| | - Takehiko Ogawa
- Laboratory of Biopharmaceutical and Regenerative Sciences, Institute of Molecular Medicine and Life Science, Association of Medical Science, Yokohama City University, Yokohama, Japan.,Department of Urology, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| |
Collapse
|
25
|
Morphology of mouse seminiferous tubules. Anat Sci Int 2018; 94:1-10. [DOI: 10.1007/s12565-018-0455-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/11/2018] [Indexed: 02/01/2023]
|
26
|
Inoue M, Baba T, Morohashi KI. Recent progress in understanding the mechanisms of Leydig cell differentiation. Mol Cell Endocrinol 2018; 468:39-46. [PMID: 29309805 DOI: 10.1016/j.mce.2017.12.013] [Citation(s) in RCA: 12] [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: 10/04/2017] [Revised: 12/25/2017] [Accepted: 12/26/2017] [Indexed: 01/26/2023]
Abstract
Leydig cells in fetal and adult testes play pivotal roles in eliciting male characteristics by producing androgen. Although numerous studies of Leydig cells have been performed, the mechanisms for differentiation of the two cell types (fetal Leydig and adult Leydig cells), their developmental and functional relationship, and their differential characteristics remain largely unclear. Based on recent technical progress in genome-wide analysis and in vitro investigation, novel and fascinating observations concerning the issues above have been obtained. Focusing on fetal and adult Leydig cells, this review summarizes the recent progress that has advanced our understanding of the cells.
Collapse
Affiliation(s)
- Miki Inoue
- Division of Molecular Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takashi Baba
- Division of Molecular Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan; Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ken-Ichirou Morohashi
- Division of Molecular Life Sciences, Graduate School of Systems Life Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan; Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
27
|
Mincheva M, Sandhowe-Klaverkamp R, Wistuba J, Redmann K, Stukenborg JB, Kliesch S, Schlatt S. Reassembly of adult human testicular cells: can testis cord-like structures be created in vitro? Mol Hum Reprod 2017; 24:55-63. [DOI: 10.1093/molehr/gax063] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/11/2017] [Indexed: 02/06/2023] Open
Affiliation(s)
- M Mincheva
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - R Sandhowe-Klaverkamp
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - J Wistuba
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - K Redmann
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - J -B Stukenborg
- Department of Women’s and Children’s Health, NORDFERTIL research lab Stockholm, Pediatric Endocrinology Unit, Q2:08, Karolinska Institutet and University Hospital, SE-17176 Stockholm, Sweden
| | - S Kliesch
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - S Schlatt
- Centre of Reproductive Medicine and Andrology, University Hospital of Münster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| |
Collapse
|
28
|
Roles of CD34+ cells and ALK5 signaling in the reconstruction of seminiferous tubule-like structures in 3-D re-aggregate culture of dissociated cells from neonatal mouse testes. PLoS One 2017; 12:e0188705. [PMID: 29190781 PMCID: PMC5708723 DOI: 10.1371/journal.pone.0188705] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 11/10/2017] [Indexed: 11/19/2022] Open
Abstract
Tissue reconstruction in vitro can provide, if successful, a refined and simple system to analyze the underlying mechanisms that drive the morphogenesis and maintain the ordered structure. We have recently succeeded in reconstruction of seminiferous cord-like and tubule-like structures using 3-D re-aggregate culture of dissociated testicular cells. In testis formation, endothelial cells that migrated from mesonephroi to embryonic gonads have been shown to be critical for development of testis cords, but how endothelial cells contribute to testis cord formation remains unknown. To decipher the roles of endothelial and peritubular cells in the reconstruction of cord-like and tubule-like structures, we investigated the behavior of CD34+ endothelial and p75+ cells, and peritubular myoid cells (PTMCs) in 3-D re-aggregate cultures of testicular cells. The results showed that these 3 types of cells had the capacity of re-aggregation on their own and with each other, and of segregation into 3 layers in a re-aggregate, which were very similar to interstitial and peritubular tissues in vivo. Observation of behaviors of fluorescent Sertoli cells and other non-fluorescent types of cells using testes from Sox9-EGFP transgenic mice showed dynamic cell movement and segregation in re-aggregate cultures. Cultures of testicular cells deprived of interstitial and peritubular cells resulted in dysmorphic structures, but re-addition of them restored tubule-like structures. Purified CD34+ cells in culture differentiated into p75+ cells and PTMCs. These results indicate that CD34+ cells differentiate into p75+ cells, which then differentiate into PTMCs. TGFβ signaling inhibitors, SB431542 and ALK5i, disturbed the reconstruction of cord-like and tubule-like structures, and the latter compromised re-construction of interstitial-like and peritubular-like structures, as well as the proliferation of CD34+, p75+, PTMCs, and Sertoli cells, and their movement and differentiation. These results indicate that CD34+ cells and signaling through ALK5 play pivotal roles in the morphogenesis of interstitial-like, peritubular-like and cord-like structures.
Collapse
|
29
|
Piprek RP, Kloc M, Tassan JP, Kubiak JZ. Development of Xenopus laevis bipotential gonads into testis or ovary is driven by sex-specific cell-cell interactions, proliferation rate, cell migration and deposition of extracellular matrix. Dev Biol 2017; 432:298-310. [PMID: 29080791 DOI: 10.1016/j.ydbio.2017.10.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/21/2017] [Accepted: 10/24/2017] [Indexed: 01/10/2023]
Abstract
Information on the mechanisms orchestrating sexual differentiation of the bipotential gonads into testes or ovaries in amphibians is limited. The aim of this study was to investigate the development of Xenopus laevis gonad, to identify the earliest signs of sexual differentiation, and to describe mechanisms driving these processes. We used light and electron microscopy, immunofluorescence and cell tracing. In order to identify the earliest signs of sexual differentiation the sex of each tadpole was determined using genotyping with the sex markers. Our analysis revealed a series of events participating in the gonadal development, including cell proliferation, migration, cell adhesion, stroma penetration, and basal lamina formation. We found that during the period of sexual differentiation the sites of intensive cell proliferation and migration differ between male and female gonads. In the differentiating ovaries the germ cells remain associated with the gonadal surface epithelium (cortex) and a sterile medulla forms in the ovarian center, whereas in the differentiating testes the germ cells detach from the surface epithelium, disperse, and the cortex and medulla fuse. Cell junctions that are more abundant in the ovarian cortex possibly can favor the persistence of germ cells in the cortex. Also the stroma penetrates the female and male gonads differently. These finding indicate that the crosstalk between the stroma and the coelomic epithelium-derived cells is crucial for development of male and female gonad.
Collapse
Affiliation(s)
- Rafal P Piprek
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland.
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA; Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA; University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jean-Pierre Tassan
- CNRS, UMR 6290, Institute of Genetics and Development of Rennes, Cell Cycle Group, 35043 Rennes, France; Université Rennes 1, Faculty of Medicine, 35043 Rennes, France
| | - Jacek Z Kubiak
- CNRS, UMR 6290, Institute of Genetics and Development of Rennes, Cell Cycle Group, 35043 Rennes, France; Université Rennes 1, Faculty of Medicine, 35043 Rennes, France; Laboratory of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), Warsaw, Poland
| |
Collapse
|
30
|
Nakata H, Sonomura T, Iseki S. Three-dimensional analysis of seminiferous tubules and spermatogenic waves in mice. Reproduction 2017; 154:569-579. [DOI: 10.1530/rep-17-0391] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 07/25/2017] [Accepted: 07/31/2017] [Indexed: 01/20/2023]
Abstract
The aim of the present study was to reconstruct seminiferous tubules and analyze spermatogenic waves in seminiferous epithelia in developing and adult mice using serial paraffin sections and high-performance three-dimensional (3D) reconstruction software. By labeling the basement membrane of seminiferous tubules with fluorescent immunohistochemistry or periodic acid-Schiff-hematoxylin staining, all seminiferous tubules were reconstructed in 9 testes from 9 different mice, 3 each at 0, 21 and 90 days (adult) postpartum. The 3D structure of seminiferous tubules, including the number and length of tubules as well as the number of connections with the rete testis, branching points and blind ends, was assessed accurately. Although tubules showed marked variations among individual mice, their overall structure was regular and retained from newborn to adult mice. Some seminiferous tubules contained inner portions running distant from the testis surface. In a representative testis at 21 days, the sites at which spermatids initially occurred were examined by labeling acrosomes and were found to be preferentially distributed in the upper and medial portions of the testis close to the rete testis. In a representative adult testis, 76 complete waves with an average length of 16.9 mm were found and their directions were analyzed. The methods used in the present study will be useful for investigating the structure and function of seminiferous tubules in mice and humans under normal and pathological conditions, such as infertility.
Collapse
|
31
|
Piprek RP, Kolasa M, Podkowa D, Kloc M, Kubiak JZ. Cell adhesion molecules expression pattern indicates that somatic cells arbitrate gonadal sex of differentiating bipotential fetal mouse gonad. Mech Dev 2017; 147:17-27. [PMID: 28760667 DOI: 10.1016/j.mod.2017.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Indexed: 01/22/2023]
Abstract
Unlike other organ anlagens, the primordial gonad is sexually bipotential in all animals. In mouse, the bipotential gonad differentiates into testis or ovary depending on the genetic sex (XY or XX) of the fetus. During gonad development cells segregate, depending on genetic sex, into distinct compartments: testis cords and interstitium form in XY gonad, and germ cell cysts and stroma in XX gonad. However, our knowledge of mechanisms governing gonadal sex differentiation remains very vague. Because it is known that adhesion molecules (CAMs) play a key role in organogenesis, we suspected that diversified expression of CAMs should also play a crucial role in gonad development. Using microarray analysis we identified 129 CAMs and factors regulating cell adhesion during sexual differentiation of mouse gonad. To identify genes expressed differentially in three cell lines in XY and XX gonads: i) supporting (Sertoli or follicular cells), ii) interstitial or stromal cells, and iii) germ cells, we used transgenic mice expressing EGFP reporter gene and FACS cell sorting. Although a large number of CAMs expressed ubiquitously, expression of certain genes was cell line- and genetic sex-specific. The sets of CAMs differentially expressed in supporting versus interstitial/stromal cells may be responsible for segregation of these two cell lines during gonadal development. There was also a significant difference in CAMs expression pattern between XY supporting (Sertoli) and XX supporting (follicular) cells but not between XY and XX germ cells. This indicates that differential CAMs expression pattern in the somatic cells but not in the germ line arbitrates structural organization of gonadal anlagen into testis or ovary.
Collapse
Affiliation(s)
- Rafal P Piprek
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland.
| | - Michal Kolasa
- Institute of Systematics and Evolution of Animals, Polish Academy of Sciences, Krakow, Poland
| | - Dagmara Podkowa
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA; The Houston Methodist Hospital, Department of Surgery, Houston, TX, USA; The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jacek Z Kubiak
- CNRS, UMR 6290, Institute of Genetics and Development of Rennes, Cell Cycle Group, F-35043, France; Université Rennes 1, UEB, UMS Biosit, Faculty of Medicine, F-35043 Rennes, France; Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
| |
Collapse
|
32
|
Zhao L, Arsenault M, Ng ET, Longmuss E, Chau TCY, Hartwig S, Koopman P. SOX4 regulates gonad morphogenesis and promotes male germ cell differentiation in mice. Dev Biol 2017; 423:46-56. [PMID: 28118982 DOI: 10.1016/j.ydbio.2017.01.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/14/2016] [Accepted: 01/09/2017] [Indexed: 01/13/2023]
Abstract
The group C SOX transcription factors SOX4, -11 and -12 play important and mutually overlapping roles in development of a number of organs. Here, we examined the role of SoxC genes during gonadal development in mice. All three genes were expressed in developing gonads of both sexes, predominantly in somatic cells, with Sox4 being most strongly expressed. Sox4 deficiency resulted in elongation of both ovaries and testes, and an increased number of testis cords. While female germ cells entered meiosis normally, male germ cells showed reduced levels of differentiation markers Nanos2 and Dnmt3l and increased levels of pluripotency genes Cripto and Nanog, suggesting that SOX4 may normally act to restrict the pluripotency period of male germ cells and ensure their proper differentiation. Finally, our data reveal that SOX4 (and, to a lesser extent, SOX11 and -12) repressed transcription of the sex-determining gene Sox9 via an upstream testis-specific enhancer core (TESCO) element in fetal gonads, raising the possibility that SOXC proteins may function as transcriptional repressors in a context-dependent manner.
Collapse
Affiliation(s)
- Liang Zhao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Michel Arsenault
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island,550 University Avenue, Charlottetown, PE, Canada C1A 4P3
| | - Ee Ting Ng
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Enya Longmuss
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Tevin Chui-Ying Chau
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Sunny Hartwig
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island,550 University Avenue, Charlottetown, PE, Canada C1A 4P3
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
33
|
Progressive induction of hepatocyte progenitor cells in chronically injured liver. Sci Rep 2017; 7:39990. [PMID: 28051157 PMCID: PMC5209740 DOI: 10.1038/srep39990] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/30/2016] [Indexed: 12/19/2022] Open
Abstract
Differentiated epithelial cells show substantial lineage plasticity upon severe tissue injuries. In chronically injured mouse livers, part of hepatocytes become Sry-HMG box containing 9 (Sox9) (+) epithelial cell adhesion molecule (−) hepatocyte nuclear factor 4 α (+) biphenotypic hepatocytes. However, it is not clear whether all Sox9+ hepatocytes uniformly possess cellular properties as hepatocyte progenitors. Here, we examined the microarray data comparing Sox9+ hepatocytes with mature hepatocytes and identified CD24 as a novel marker for biphenotypic hepatocytes. Immunohistochemical analyses showed that part of Sox9+ hepatocytes near expanded ductular structures expressed CD24 in the liver injured by 3,5-diethoxycarbonyl-1,4-dihydro-collidine (DDC) diet and by bile duct ligation. Indeed, Sox9+ hepatocytes could be separated into CD24− and CD24+ cells by fluorescence activated cell sorting. The ratio of CD24+ cells against CD24− ones in Sox9+ hepatocytes gradually increased while DDC-injury progressed and colony-forming capability mostly attributed to CD24+ cells. Although hepatocyte markers were remarkably downregulated in of Sox9+ CD24+ hepatocytes, they re-differentiated into mature hepatocytes in vitro and in vivo. Our current results demonstrate that the emergence of biphenotypic hepatocytes is a sequential event including the transition from CD24− and CD24+ status, which may be a crucial step for hepatocytes to acquire progenitor properties.
Collapse
|
34
|
Inoue M, Shima Y, Miyabayashi K, Tokunaga K, Sato T, Baba T, Ohkawa Y, Akiyama H, Suyama M, Morohashi KI. Isolation and Characterization of Fetal Leydig Progenitor Cells of Male Mice. Endocrinology 2016; 157:1222-33. [PMID: 26697723 DOI: 10.1210/en.2015-1773] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fetal and adult Leydig cells develop in mammalian prenatal and postnatal testes, respectively. In mice, fetal Leydig cells (FLCs) emerge in the interstitial space of the testis at embryonic day 12.5 and thereafter increase in number, possibly through differentiation from progenitor cells. However, the progenitor cells have not yet been identified. Previously, we established transgenic mice in which FLCs are labeled strongly with enhanced green fluorescent protein (EGFP). Interestingly, fluorescence-activated cell sorting provided us with weakly EGFP-labeled cells as well as strongly EGFP-labeled FLCs. In vitro reconstruction of fetal testes demonstrated that weakly EGFP-labeled cells contain FLC progenitors. Transcriptome from the 2 cell populations revealed, as expected, marked differences in the expression of genes required for growth factor/receptor signaling and steroidogenesis. In addition, genes for energy metabolisms such as glycolytic pathways and the citrate cycle were activated in strongly EGFP-labeled cells, suggesting that metabolism is activated during FLC differentiation.
Collapse
Affiliation(s)
- Miki Inoue
- Division of Molecular Life Science (M.I., Y.S., T.B., K.-i.M.), Graduate School of Systems Life Science; Department of Molecular Biology (Y.S., K.M., K.T., T.B., K.-i.M.), Graduate School of Medical Sciences; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation; and Department of Advanced Medical Initiatives (Y.O.), Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Faculty of Medicine, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; and Department of Orthopaedics (H.A.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Yuichi Shima
- Division of Molecular Life Science (M.I., Y.S., T.B., K.-i.M.), Graduate School of Systems Life Science; Department of Molecular Biology (Y.S., K.M., K.T., T.B., K.-i.M.), Graduate School of Medical Sciences; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation; and Department of Advanced Medical Initiatives (Y.O.), Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Faculty of Medicine, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; and Department of Orthopaedics (H.A.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Kanako Miyabayashi
- Division of Molecular Life Science (M.I., Y.S., T.B., K.-i.M.), Graduate School of Systems Life Science; Department of Molecular Biology (Y.S., K.M., K.T., T.B., K.-i.M.), Graduate School of Medical Sciences; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation; and Department of Advanced Medical Initiatives (Y.O.), Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Faculty of Medicine, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; and Department of Orthopaedics (H.A.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Kaori Tokunaga
- Division of Molecular Life Science (M.I., Y.S., T.B., K.-i.M.), Graduate School of Systems Life Science; Department of Molecular Biology (Y.S., K.M., K.T., T.B., K.-i.M.), Graduate School of Medical Sciences; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation; and Department of Advanced Medical Initiatives (Y.O.), Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Faculty of Medicine, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; and Department of Orthopaedics (H.A.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Tetsuya Sato
- Division of Molecular Life Science (M.I., Y.S., T.B., K.-i.M.), Graduate School of Systems Life Science; Department of Molecular Biology (Y.S., K.M., K.T., T.B., K.-i.M.), Graduate School of Medical Sciences; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation; and Department of Advanced Medical Initiatives (Y.O.), Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Faculty of Medicine, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; and Department of Orthopaedics (H.A.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Takashi Baba
- Division of Molecular Life Science (M.I., Y.S., T.B., K.-i.M.), Graduate School of Systems Life Science; Department of Molecular Biology (Y.S., K.M., K.T., T.B., K.-i.M.), Graduate School of Medical Sciences; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation; and Department of Advanced Medical Initiatives (Y.O.), Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Faculty of Medicine, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; and Department of Orthopaedics (H.A.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Yasuyuki Ohkawa
- Division of Molecular Life Science (M.I., Y.S., T.B., K.-i.M.), Graduate School of Systems Life Science; Department of Molecular Biology (Y.S., K.M., K.T., T.B., K.-i.M.), Graduate School of Medical Sciences; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation; and Department of Advanced Medical Initiatives (Y.O.), Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Faculty of Medicine, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; and Department of Orthopaedics (H.A.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Haruhiko Akiyama
- Division of Molecular Life Science (M.I., Y.S., T.B., K.-i.M.), Graduate School of Systems Life Science; Department of Molecular Biology (Y.S., K.M., K.T., T.B., K.-i.M.), Graduate School of Medical Sciences; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation; and Department of Advanced Medical Initiatives (Y.O.), Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Faculty of Medicine, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; and Department of Orthopaedics (H.A.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Mikita Suyama
- Division of Molecular Life Science (M.I., Y.S., T.B., K.-i.M.), Graduate School of Systems Life Science; Department of Molecular Biology (Y.S., K.M., K.T., T.B., K.-i.M.), Graduate School of Medical Sciences; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation; and Department of Advanced Medical Initiatives (Y.O.), Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Faculty of Medicine, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; and Department of Orthopaedics (H.A.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Ken-ichirou Morohashi
- Division of Molecular Life Science (M.I., Y.S., T.B., K.-i.M.), Graduate School of Systems Life Science; Department of Molecular Biology (Y.S., K.M., K.T., T.B., K.-i.M.), Graduate School of Medical Sciences; Division of Bioinformatics (T.S., M.S.), Medical Institute of Bioregulation; and Department of Advanced Medical Initiatives (Y.O.), Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, Faculty of Medicine, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan; and Department of Orthopaedics (H.A.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| |
Collapse
|
35
|
Chen SR, Liu YX. Testis Cord Maintenance in Mouse Embryos: Genes and Signaling1. Biol Reprod 2016; 94:42. [DOI: 10.1095/biolreprod.115.137117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/12/2016] [Indexed: 12/12/2022] Open
|
36
|
Abstract
Current knowledge on gonadal development and sex determination is the product of many decades of research involving a variety of scientific methods from different biological disciplines such as histology, genetics, biochemistry, and molecular biology. The earliest embryological investigations, followed by the invention of microscopy and staining methods, were based on histological examinations. The most robust development of histological staining techniques occurred in the second half of the nineteenth century and resulted in structural descriptions of gonadogenesis. These first studies on gonadal development were conducted on domesticated animals; however, currently the mouse is the most extensively studied species. The next key point in the study of gonadogenesis was the advancement of methods allowing for the in vitro culture of fetal gonads. For instance, this led to the description of the origin of cell lines forming the gonads. Protein detection using antibodies and immunolabeling methods and the use of reporter genes were also invaluable for developmental studies, enabling the visualization of the formation of gonadal structure. Recently, genetic and molecular biology techniques, especially gene expression analysis, have revolutionized studies on gonadogenesis and have provided insight into the molecular mechanisms that govern this process. The successive invention of new methods is reflected in the progress of research on gonadal development.
Collapse
Affiliation(s)
- Rafal P Piprek
- Department of Comparative Anatomy, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland.
| |
Collapse
|
37
|
Romereim SM, Cupp AS. Mesonephric Cell Migration into the Gonads and Vascularization Are Processes Crucial for Testis Development. Results Probl Cell Differ 2016; 58:67-100. [PMID: 27300176 DOI: 10.1007/978-3-319-31973-5_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Testis morphogenesis requires the integration and reorganization of multiple cell types from several sources, one of the more notable being the mesonephric-derived cell population. One of the earliest sex-specific morphogenetic events in the gonad is a wave of endothelial cell migration from the mesonephros that is crucial for (1) partitioning the gonad into domains for testis cords, (2) providing the vasculature of the testis, and (3) signaling to cells both within the gonad and beyond it to coordinately regulate testis development. In addition to endothelial cell migration, there is evidence that precursors of peritubular myoid cells migrate from the mesonephros, an event which is also important for testis cord architecture. Investigation of the mesonephric cell migration event has utilized histology, lineage tracing with mouse genetic markers, and many studies of the signaling molecules/pathways involved. Some of the more well-studied signaling molecules involved include vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and neurotrophins. In this chapter, the morphogenetic events, relevant signaling pathways, mechanisms underlying the migration, and the role of the migratory cells within the testis will be discussed. Overall, the migration of mesonephric cells into the early testis is indispensable for its development and future functionality.
Collapse
|
38
|
Potter SJ, DeFalco T. Using Ex Vivo Upright Droplet Cultures of Whole Fetal Organs to Study Developmental Processes during Mouse Organogenesis. J Vis Exp 2015:e53262. [PMID: 26556004 DOI: 10.3791/53262] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Investigating organogenesis in utero is a technically challenging process in placental mammals due to inaccessibility of reagents to embryos that develop within the uterus. A newly developed ex vivo upright droplet culture method provides an attractive alternative to studies performed in utero. The ex vivo droplet culture provides the ability to examine and manipulate cellular interactions and diverse signaling pathways through use of various blocking and activating compounds; additionally, the effects of various pharmacological reagents on the development of specific organs can be studied without unwanted side effects of systemic drug delivery in utero. As compared to other in vitro systems, the droplet culture not only allows for the ability to study three-dimensional morphogenesis and cell-cell interactions, which cannot be reproduced in mammalian cell lines, but also requires significantly less reagents than other ex vivo and in vitro protocols. This paper demonstrates proper mouse fetal organ dissection and upright droplet culture techniques, followed by whole organ immunofluorescence to demonstrate the effectiveness of the method. The ex vivo droplet culture method allows the formation of organ architecture comparable to what is observed in vivo and can be utilized to study otherwise difficult-to-study processes due to embryonic lethality in in vivo models. As a model application system, a small-molecule inhibitor will be utilized to probe the role of vascularization in testicular morphogenesis. This ex vivo droplet culture method is expandable to other fetal organ systems, such as lung and potentially others, although each organ must be extensively studied to determine any organ-specific modifications to the protocol. This organ culture system provides flexibility in experimentation with fetal organs, and results obtained using this technique will help researchers gain insights into fetal development.
Collapse
Affiliation(s)
- Sarah J Potter
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center;
| |
Collapse
|
39
|
Nakata H, Wakayama T, Sonomura T, Honma S, Hatta T, Iseki S. Three-dimensional structure of seminiferous tubules in the adult mouse. J Anat 2015; 227:686-94. [PMID: 26391090 DOI: 10.1111/joa.12375] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2015] [Indexed: 11/28/2022] Open
Abstract
Seminiferous tubules develop from sex cords, which are embryonic structures with simple C-shaped arches. Histologically, the epithelium of adult mouse seminiferous tubules has been divided into 12 stages based on the associations of spermatogenic cells in four cycles of spermatogenesis. However, the gross characteristics of the seminiferous tubules themselves, including their number, length, run, and mutual relationships remain largely unknown. In the present study, we analyzed all seminiferous tubules in a single adult mouse testis with high resolution using serial paraffin sections and high-perfomance three-dimensional reconstruction software. There were 11 seminiferous tubules with an average length of 140 mm. Each tubule ran along circular paths within the testis while making convolutions with cranial and caudal hairpin turns. The cranial turns of all tubules were in contact with the tunica albuginea, whereas the caudal turns were not, resulting in funnel-shaped networks of these tubules with tapered caudal portions. The caudally located networks surrounded the preceding cranially located networks from the bottom and outside, similar to stacked paper cups. Five out of the 11 seminiferous tubules were continuous from one end to the other both connected with the rete testis (10 connection points). Nine branching points, one blind end, and 18 more connection points with the rete testis were detected in the remaining six seminiferous tubules, making the paths of these tubules complicated to various degrees. The present study revealed that the 3D structures of seminiferous tubules were highly regular as a whole in the adult mouse testis.
Collapse
Affiliation(s)
- Hiroki Nakata
- Department of Histology and Embryology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Tomohiko Wakayama
- Department of Histology and Embryology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Takahiro Sonomura
- Department of Anatomy II, School of Medicine, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Satoru Honma
- Department of Anatomy II, School of Medicine, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Toshihisa Hatta
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Shoichi Iseki
- Department of Histology and Embryology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
40
|
Zimmermann C, Stévant I, Borel C, Conne B, Pitetti JL, Calvel P, Kaessmann H, Jégou B, Chalmel F, Nef S. Research resource: the dynamic transcriptional profile of sertoli cells during the progression of spermatogenesis. Mol Endocrinol 2015; 29:627-42. [PMID: 25710594 DOI: 10.1210/me.2014-1356] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Sertoli cells (SCs), the only somatic cells within seminiferous tubules, associate intimately with developing germ cells. They not only provide physical and nutritional support but also secrete factors essential to the complex developmental processes of germ cell proliferation and differentiation. The SC transcriptome must therefore adapt rapidly during the different stages of spermatogenesis. We report comprehensive genome-wide expression profiles of pure populations of SCs isolated at 5 distinct stages of the first wave of mouse spermatogenesis, using RNA sequencing technology. We were able to reconstruct about 13 901 high-confidence, nonredundant coding and noncoding transcripts, characterized by complex alternative splicing patterns with more than 45% comprising novel isoforms of known genes. Interestingly, roughly one-fifth (2939) of these genes exhibited a dynamic expression profile reflecting the evolving role of SCs during the progression of spermatogenesis, with stage-specific expression of genes involved in biological processes such as cell cycle regulation, metabolism and energy production, retinoic acid synthesis, and blood-testis barrier biogenesis. Finally, regulatory network analysis identified the transcription factors endothelial PAS domain-containing protein 1 (EPAS1/Hif2α), aryl hydrocarbon receptor nuclear translocator (ARNT/Hif1β), and signal transducer and activator of transcription 1 (STAT1) as potential master regulators driving the SC transcriptional program. Our results highlight the plastic transcriptional landscape of SCs during the progression of spermatogenesis and provide valuable resources to better understand SC function and spermatogenesis and its related disorders, such as male infertility.
Collapse
Affiliation(s)
- Céline Zimmermann
- Department of Genetic Medicine and Development (C.Z., I.S., C.B., B.C., J.-L.P., P.C., S.N.), University of Geneva Medical School, 1211 Geneva 4, Switzerland; Center for Integrative Genomics (H.K.), University of Lausanne, Génopode, CH-1015 Lausanne, Switzerland; and Inserm U1085-IRSET (B.J., F.C.), Université de Rennes 1, Campus de Beaulieu, F-35042 Rennes, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Huang CC, Orvis GD, Kwan KM, Behringer RR. Lhx1 is required in Müllerian duct epithelium for uterine development. Dev Biol 2014; 389:124-36. [PMID: 24560999 DOI: 10.1016/j.ydbio.2014.01.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 02/08/2023]
Abstract
The female reproductive tract organs of mammals, including the oviducts, uterus, cervix and upper vagina, are derived from the Müllerian ducts, a pair of epithelial tubes that form within the mesonephroi. The Müllerian ducts form in a rostral to caudal manner, guided by and dependent on the Wolffian ducts that have already formed. Experimental embryological studies indicate that caudal elongation of the Müllerian duct towards the urogenital sinus occurs in part by proliferation at the ductal tip. The molecular mechanisms that regulate the elongation of the Müllerian duct are currently unclear. Lhx1 encodes a LIM-homeodomain transcription factor that is essential for male and female reproductive tract development. Lhx1 is expressed in both the Wolffian and Müllerian ducts. Wolffian duct-specific knockout of Lhx1 results in degeneration of the Wolffian duct and consequently the non-cell-autonomous loss of the Müllerian duct. To determine the role of Lhx1 specifically in the Müllerian duct epithelium, we performed a Müllerian duct-specific knockout study using Wnt7a-Cre mice. Loss of Lhx1 in the Müllerian duct epithelium led to a block in Müllerian duct elongation and uterine hypoplasia characterized by loss of the entire endometrium (luminal and glandular epithelium and stroma) and inner circular but not the outer longitudinal muscle layer. Time-lapse imaging and molecular analyses indicate that Lhx1 acts cell autonomously to maintain ductal progenitor cells for Müllerian duct elongation. These studies identify LHX1 as the first transcription factor that is essential in the Müllerian duct epithelial progenitor cells for female reproductive tract development. Furthermore, these genetic studies demonstrate the requirement of epithelial-mesenchymal interactions for uterine tissue compartment differentiation.
Collapse
Affiliation(s)
- Cheng-Chiu Huang
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Grant D Orvis
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Kin Ming Kwan
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, PR China
| | - Richard R Behringer
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
42
|
Tanimizu N, Nishikawa Y, Ichinohe N, Akiyama H, Mitaka T. Sry HMG box protein 9-positive (Sox9+) epithelial cell adhesion molecule-negative (EpCAM-) biphenotypic cells derived from hepatocytes are involved in mouse liver regeneration. J Biol Chem 2014; 289:7589-98. [PMID: 24482234 DOI: 10.1074/jbc.m113.517243] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
It has been shown that mature hepatocytes compensate tissue damages not only by proliferation and/or hypertrophy but also by conversion into cholangiocyte-like cells. We found that Sry HMG box protein 9-positive (Sox9(+)) epithelial cell adhesion molecule-negative (EpCAM(-)) hepatocyte nuclear factor 4α-positive (HNF4α(+)) biphenotypic cells showing hepatocytic morphology appeared near EpCAM(+) ductular structures in the livers of mice fed 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-containing diet. When Mx1-Cre:ROSA mice, which were injected with poly(I:C) to label mature hepatocytes, were fed with the DDC diet, we found LacZ(+)Sox9(+) cells near ductular structures. Although Sox9(+)EpCAM(-) cells adjacent to expanding ducts likely further converted into ductular cells, the incidence was rare. To know the cellular characteristics of Sox9(+)EpCAM(-) cells, we isolated them as GFP(+)EpCAM(-) cells from DDC-injured livers of Sox9-EGFP mice. Sox9(+)EpCAM(-) cells proliferated and could differentiate to functional hepatocytes in vitro. In addition, Sox9(+)EpCAM(-) cells formed cysts with a small central lumen in collagen gels containing Matrigel® without expressing EpCAM. These results suggest that Sox9(+)EpCAM(-) cells maintaining biphenotypic status can establish cholangiocyte-type polarity. Interestingly, we found that some of the Sox9(+) cells surrounded luminal spaces in DDC-injured liver while they expressed HNF4α. Taken together, we consider that in addition to converting to cholangiocyte-like cells, Sox9(+)EpCAM(-) cells provide luminal space near expanded ductular structures to prevent deterioration of the injuries and potentially supply new hepatocytes to repair damaged tissues.
Collapse
Affiliation(s)
- Naoki Tanimizu
- From the Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556
| | | | | | | | | |
Collapse
|
43
|
Svingen T, Koopman P. Building the mammalian testis: origins, differentiation, and assembly of the component cell populations. Genes Dev 2013; 27:2409-26. [PMID: 24240231 PMCID: PMC3841730 DOI: 10.1101/gad.228080.113] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Development of testes in the mammalian embryo requires the formation and assembly of several cell types that allow these organs to achieve their roles in male reproduction and endocrine regulation. Testis development is unusual in that several cell types such as Sertoli, Leydig, and spermatogonial cells arise from bipotential precursors present in the precursor tissue, the genital ridge. These cell types do not differentiate independently but depend on signals from Sertoli cells that differentiate under the influence of transcription factors SRY and SOX9. While these steps are becoming better understood, the origins and roles of many testicular cell types and structures-including peritubular myoid cells, the tunica albuginea, the arterial and venous blood vasculature, lymphatic vessels, macrophages, and nerve cells-have remained unclear. This review synthesizes current knowledge of how the architecture of the testis unfolds and highlights the questions that remain to be explored, thus providing a roadmap for future studies that may help illuminate the causes of XY disorders of sex development, infertility, and testicular cancers.
Collapse
Affiliation(s)
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|
44
|
A conditional mouse line for lineage tracing of Sox9 loss-of-function cells using enhanced green fluorescent protein. Biotechnol Lett 2013; 35:1991-6. [PMID: 23907671 DOI: 10.1007/s10529-013-1303-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 07/10/2013] [Indexed: 10/26/2022]
Abstract
Traditionally, conditional knockout studies in mouse have utilized the Cre or Flpe technology to activate the expression of reporter genes such as lacZ or PLAP. Employing these reporter genes, however, requires tissue fixation. To make way for downstream in vivo or in vitro applications, we have inserted enhanced green fluorescent protein (EGFP) into the endogenous Sox9 locus and generated a novel conditional Sox9 null allele, by flanking the entire Sox9 coding region with loxP sites and inserting an EGFP reporter gene into the 3'-UTR allowing for EGFP to be expressed upon Sox9 loss of function yet under the control of the endogenous Sox9 promoter. Mating this new allele to any Cre-expressing line, the fate of Sox9 null cells can be traced in the cell type of interest in vivo or in vitro after fluorescence-activated cell sorting.
Collapse
|
45
|
Pitetti JL, Calvel P, Zimmermann C, Conne B, Papaioannou MD, Aubry F, Cederroth CR, Urner F, Fumel B, Crausaz M, Docquier M, Herrera PL, Pralong F, Germond M, Guillou F, Jégou B, Nef S. An essential role for insulin and IGF1 receptors in regulating sertoli cell proliferation, testis size, and FSH action in mice. Mol Endocrinol 2013; 27:814-27. [PMID: 23518924 DOI: 10.1210/me.2012-1258] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Testis size and sperm production are directly correlated to the total number of adult Sertoli cells (SCs). Although the establishment of an adequate number of SCs is crucial for future male fertility, the identification and characterization of the factors regulating SC survival, proliferation, and maturation remain incomplete. To investigate whether the IGF system is required for germ cell (GC) and SC development and function, we inactivated the insulin receptor (Insr), the IGF1 receptor (Igf1r), or both receptors specifically in the GC lineage or in SCs. Whereas ablation of insulin/IGF signaling appears dispensable for GCs and spermatogenesis, adult testes of mice lacking both Insr and Igf1r in SCs (SC-Insr;Igf1r) displayed a 75% reduction in testis size and daily sperm production as a result of a reduced proliferation rate of immature SCs during the late fetal and early neonatal testicular period. In addition, in vivo analyses revealed that FSH requires the insulin/IGF signaling pathway to mediate its proliferative effects on immature SCs. Collectively, these results emphasize the essential role played by growth factors of the insulin family in regulating the final number of SCs, testis size, and daily sperm output. They also indicate that the insulin/IGF signaling pathway is required for FSH-mediated SC proliferation.
Collapse
Affiliation(s)
- Jean-Luc Pitetti
- Department of Genetic Medicine and Development, National Center of Competence in Research, Frontiers in Genetics, University of Geneva, 1211 Geneva 4, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Shima Y, Miyabayashi K, Haraguchi S, Arakawa T, Otake H, Baba T, Matsuzaki S, Shishido Y, Akiyama H, Tachibana T, Tsutsui K, Morohashi KI. Contribution of Leydig and Sertoli cells to testosterone production in mouse fetal testes. Mol Endocrinol 2012; 27:63-73. [PMID: 23125070 DOI: 10.1210/me.2012-1256] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Testosterone is a final product of androgenic hormone biosynthesis, and Leydig cells are known to be the primary source of androgens. In the mammalian testis, two distinct populations of Leydig cells, the fetal and the adult Leydig cells, develop sequentially, and these two cell types differ both morphologically and functionally. It is well known that the adult Leydig cells maintain male reproductive function by producing testosterone. However, it has been controversial whether fetal Leydig cells can produce testosterone, and the synthetic pathway of testosterone in the fetal testis is not fully understood. In the present study, we generated transgenic mice in which enhanced green fluorescence protein was expressed under the control of a fetal Leydig cell-specific enhancer of the Ad4BP/SF-1 (Nr5a1) gene. The transgene construct was prepared by mutating the LIM homeodomain transcription factor (LHX9)-binding sequence in the promoter, which abolished promoter activity in the undifferentiated testicular cells. These transgenic mice were used to collect highly pure fetal Leydig cells. Gene expression and steroidogenic enzyme activities in the fetal Leydig cells as well as in the fetal Sertoli cells and adult Leydig cells were analyzed. Our results revealed that the fetal Leydig cells synthesize only androstenedione because they lack expression of Hsd17b3, and fetal Sertoli cells convert androstenedione to testosterone, whereas adult Leydig cells synthesize testosterone by themselves. The current study demonstrated that both Leydig and Sertoli cells are required for testosterone synthesis in the mouse fetal testis.
Collapse
Affiliation(s)
- Yuichi Shima
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Tanwar PS, Commandeur AE, Zhang L, Taketo MM, Teixeira JM. The Müllerian inhibiting substance type 2 receptor suppresses tumorigenesis in testes with sustained β-catenin signaling. Carcinogenesis 2012; 33:2351-61. [PMID: 22962306 DOI: 10.1093/carcin/bgs281] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Dysregulated WNT/β-catenin signaling in murine testes results in a phenotype with complete germ cell loss that resembles human Sertoli cell-only syndrome. In other systems, including the ovary, dysregulated WNT/β-catenin induces tumorigenesis but no tumors are observed in the mutant testes without deletion of a tumor suppressor, such as phosphatase and tensin homolog (PTEN). Müllerian inhibiting substance (MIS, also known as AMH), a member of the transforming growth factor-β family of growth factors responsible for Müllerian duct regression in fetal males, has been shown to inhibit tumor growth in vitro and in vivo but its role as an endogenous tumor suppressor has never been reported. We have deleted the MIS type 2 receptor (MISR2), and thus MIS signaling, in mice with dysregulated WNT/β-catenin and show that these mice develop testicular stromal tumors with 100% penetrance within a few months postnatal. The tumors are highly proliferative and have characteristics of either Sertoli cell tumors or progenitor Leydig cell tumors based on their marker profiles and histology. Phosphorylated Sma and mothers against decapentaplegic-related homolog 1/5/8 is absent in the tumors and β-catenin target genes are induced. The tumor suppressor TP53 is also highly expressed in the tumors, as is phosphorylated γH2AX, which is indicative of DNA damage. The phenotype of these tumors closely resembles those observed when PTEN is also deleted in mice with dysregulated WNT/β-catenin. Tumorigenesis in these mice provides conclusive evidence that physiological MIS signaling is a tumor suppressor mechanism and suggests that targeted treatment of MISR2-expressing cancers with therapeutic MIS should have a beneficial effect on tumor progression.
Collapse
Affiliation(s)
- Pradeep S Tanwar
- Vincent Center for Reproductive Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
48
|
Ungewitter EK, Yao HHC. How to make a gonad: cellular mechanisms governing formation of the testes and ovaries. Sex Dev 2012; 7:7-20. [PMID: 22614391 PMCID: PMC3474884 DOI: 10.1159/000338612] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Sex determination of the gonad is an extraordinary process by which a single organ anlage is directed to form one of two different structures, a testis or an ovary. Morphogenesis of these two organs utilizes many common cellular events; differences in the timing and execution of these events must combine to generate sexually dimorphic structures. In this chapter, we review recent research on the cellular processes of gonad morphogenesis, focusing on data from mouse models. We highlight the shared cellular mechanisms in testis and ovary morphogenesis and examine the differences that enable formation of the two organs responsible for the perpetuation of all sexually reproducing species.
Collapse
Affiliation(s)
- E K Ungewitter
- Reproductive Developmental Biology Group, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
49
|
Cool J, DeFalco T, Capel B. Testis formation in the fetal mouse: dynamic and complex de novo
tubulogenesis. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 1:847-59. [DOI: 10.1002/wdev.62] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
50
|
Ohta K, Yamamoto M, Lin Y, Hogg N, Akiyama H, Behringer RR, Yamazaki Y. Male differentiation of germ cells induced by embryonic age-specific Sertoli cells in mice. Biol Reprod 2012; 86:112. [PMID: 22262692 PMCID: PMC3338658 DOI: 10.1095/biolreprod.111.095943] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 09/13/2011] [Accepted: 01/05/2012] [Indexed: 11/01/2022] Open
Abstract
Retinoic acid (RA) is a meiosis-inducing factor. Primordial germ cells (PGCs) in the developing ovary are exposed to RA, resulting in entry into meiosis. In contrast, PGCs in the developing testis enter mitotic arrest to differentiate into prospermatogonia. Sertoli cells express CYP26B1, an RA-metabolizing enzyme, providing a simple explanation for why XY PGCs do not initiate meios/is. However, regulation of entry into mitotic arrest is likely more complex. To investigate the mechanisms that regulate male germ cell differentiation, we cultured XX and XY germ cells at 11.5 and 12.5 days postcoitus (dpc) with an RA receptor inhibitor. Expression of Stra8, a meiosis initiation gene, was suppressed in all groups. However, expression of Dnmt3l, a male-specific gene, during embryogenesis was elevated but only in 12.5-dpc XY germ cells. This suggests that inhibiting RA signaling is not sufficient for male germ cell differentiation but that the male gonadal environment also contributes to this pathway. To define the influence of Sertoli cells on male germ cell differentiation, Sertoli cells at 12.5, 15.5, and 18.5 dpc were aggregated with 11.5 dpc PGCs, respectively. After culture, PGCs aggregated with 12.5 dpc Sertoli cells increased Nanos2 and Dnmt3l expression. Furthermore, these PGCs established male-specific methylation imprints of the H19 differentially methylated domains. In contrast, PGCs aggregated with Sertoli cells at late embryonic ages did not commit to the male pathway. These findings suggest that male germ cell differentiation is induced both by inhibition of RA signaling and by molecule(s) production by embryonic age-specific Sertoli cells.
Collapse
Affiliation(s)
- Kohei Ohta
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Hawaii, USA.
| | | | | | | | | | | | | |
Collapse
|