1
|
Chen H, Fang HQ, Liu JT, Chang SY, Cheng LB, Sun MX, Feng JR, Liu ZM, Zhang YH, Rosen CJ, Liu P. Atlas of Fshr expression from novel reporter mice. eLife 2025; 13:RP93413. [PMID: 39773308 PMCID: PMC11709436 DOI: 10.7554/elife.93413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
The FSH-FSHR pathway has been considered an essential regulator in reproductive development and fertility. But there has been emerging evidence of FSHR expression in extragonadal organs. This poses new questions and long-term debates regarding the physiological role of the FSH-FSHR, and underscores the need for reliable, in vivo analysis of FSHR expression in animal models. However, conventional methods have proven insufficient for examining FSHR expression due to several limitations. To address this challenge, we developed Fshr-ZsGreen reporter mice under the control of Fshr endogenous promoter using CRISPR-Cas9. With this novel genetic tool, we provide a reliable readout of Fshr expression at single-cell resolution level in vivo and in real time. Reporter animals were also subjected to additional analyses,to define the accurate expression profile of FSHR in gonadal and extragonadal organs/tissues. Our compelling results not only demonstrated Fshr expression in intragonadal tissues but also, strikingly, unveiled notably increased expression in Leydig cells, osteoblast lineage cells, endothelial cells in vascular structures, and epithelial cells in bronchi of the lung and renal tubes. The genetic decoding of the widespread pattern of Fshr expression highlights its physiological relevance beyond reproduction and fertility, and opens new avenues for therapeutic options for age-related disorders of the bones, lungs, kidneys, and hearts, among other tissues. Exploiting the power of the Fshr knockin reporter animals, this report provides the first comprehensive genetic record of the spatial distribution of FSHR expression, correcting a long-term misconception about Fshr expression and offering prospects for extensive exploration of FSH-FSHR biology.
Collapse
Affiliation(s)
- Hongqian Chen
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Hui-Qing Fang
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
- Department of Dentistry, The 980th Hospital of the PLA Joint Logistic Support ForceShijiazhuangChina
| | - Jin-Tao Liu
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Shi-Yu Chang
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Li-Ben Cheng
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Ming-Xin Sun
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Jian-Rui Feng
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Ze-Min Liu
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
- Shanxi Medical Universityersity, The Second Hospital, University Shanxi Medical UniversityTaiyuanChina
| | - Yong-Hong Zhang
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
- Shanxi Medical Universityersity, The Second Hospital, University Shanxi Medical UniversityTaiyuanChina
| | | | - Peng Liu
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
2
|
Jeanne F, Pilet S, Klett D, Combarnous Y, Bernay B, Dufour S, Favrel P, Sourdaine P. Characterization of gonadotropins and their receptors in a chondrichthyan, Scyliorhinus canicula, fills a gap in the understanding of their coevolution. Gen Comp Endocrinol 2024; 358:114614. [PMID: 39326529 DOI: 10.1016/j.ygcen.2024.114614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/18/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
In Gnathostomes, reproduction is mainly controlled by the hypothalamic-pituitary-gonadal (HPG) axis, with the involvement of the pituitary gonadotropic hormones (GTH), follicle-stimulating hormone (FSH) and luteinizing hormone (LH), which activate their cognate receptors, FSHR and LHR, expressed in gonads. Each GTH consists of a common α subunit and of a specific FSHβ or LHβ subunit. Chondrichthyes (holocephalans and elasmobranchs) is a sister group of bony vertebrates. This position is highly favorable for the understanding of the evolution of endocrine regulations of reproduction among gnathostomes. Surprisingly, the characterization of gonadotropins and their receptors is still limited in chondrichthyes. In the present study, GTH and GTHR sequences have been identified from several chondrichthyan genomes, and their primary structures were analyzed relative to human orthologs. 3D models of GTH/GTHR interaction were built, highlighting the importance of the receptor hinge region for ligand recognition. Functional hormone-receptor interactions have been studied in HEK cells using the small-spotted catshark (Scyliorhinus canicula) recombinant proteins and showed that LHR was specifically activated by LH whereas FSHR was activated by both FSH and LH. Expression profiles of GTHs and their receptors were explored by real-time PCR, in situ hybridization and immunohistochemistry during spermatogenesis, along the male genital tract and other tissues, as well as in some female tissues for comparison. Tissue-expression analyses showed that the highest levels were observed for fshr transcripts in testis and ovary and for lhr in specific extragonadal tissues. The two receptors were expressed at all stages of spermatogenesis by both germ cells and somatic cells, including undifferentiated spermatogonia, spermatocytes, spermatids, somatic precursors and Sertoli cells; differentiated Leydig cells being absent in the testis of S. canicula. Receptors were also expressed by the lymphomyeloid epigonal tissue and the testicular tubules. These results, suggest a wide range of gonadotropin-regulated functions in Elasmobranchs, as well as functional redundancy during spermatogenesis. These extended functions are discussed in an evolutionary context in which the specificity of gonadotropin signaling must have contributed to the evolution of gonadal cells' morphology and function.
Collapse
Affiliation(s)
- Fabian Jeanne
- Université de Caen Normandie, MNHN, SU, UA, CNRS, IRD, Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), UMR 8067, 14032 Caen cedex 5, France
| | - Stanislas Pilet
- Université de Caen Normandie, MNHN, SU, UA, CNRS, IRD, Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), UMR 8067, 14032 Caen cedex 5, France
| | - Danièle Klett
- INRAE, CNRS, UMR Physiologie de la Reproduction & des Comportements, 37380 Nouzilly, France
| | - Yves Combarnous
- INRAE, CNRS, UMR Physiologie de la Reproduction & des Comportements, 37380 Nouzilly, France
| | - Benoît Bernay
- Université de Caen Normandie - Plateforme PROTEOGEN, US EMerode, 14032 Caen cedex 5, France
| | - Sylvie Dufour
- Université de Caen Normandie, MNHN, SU, UA, CNRS, IRD, Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), UMR 8067, 14032 Caen cedex 5, France
| | - Pascal Favrel
- Université de Caen Normandie, MNHN, SU, UA, CNRS, IRD, Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), UMR 8067, 14032 Caen cedex 5, France
| | - Pascal Sourdaine
- Université de Caen Normandie, MNHN, SU, UA, CNRS, IRD, Laboratoire de Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), UMR 8067, 14032 Caen cedex 5, France.
| |
Collapse
|
3
|
Zanardini M, Zhang W, Habibi HR. Arginine Vasotocin Directly Regulates Spermatogenesis in Adult Zebrafish ( Danio rerio) Testes. Int J Mol Sci 2024; 25:6564. [PMID: 38928267 PMCID: PMC11204076 DOI: 10.3390/ijms25126564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The neuropeptide vasopressin is known for its regulation of osmotic balance in mammals. Arginine vasotocin (AVT) is a non-mammalian homolog of this neuropeptide that is present in fish. Limited information suggested that vasopressin and its homologs may also influence reproductive function. In the present study, we investigated the direct effect of AVT on spermatogenesis, using zebrafish as a model organism. Results demonstrate that AVT and its receptors (avpr1aa, avpr2aa, avpr1ab, avpr2ab, and avpr2l) are expressed in the zebrafish brain and testes. The direct action of AVT on spermatogenesis was investigated using an ex vivo culture of mature zebrafish testes for 7 days. Using histological, morphometric, and biochemical approaches, we observed direct actions of AVT on zebrafish testicular function. AVT treatment directly increased the number of spermatozoa in an androgen-dependent manner, while reducing mitotic cells and the proliferation activity of type B spermatogonia. The observed stimulatory action of AVT on spermiogenesis was blocked by flutamide, an androgen receptor antagonist. The present results support the novel hypothesis that AVT stimulates short-term androgen-dependent spermiogenesis. However, its prolonged presence may lead to diminished spermatogenesis by reducing the proliferation of spermatogonia B, resulting in a diminished turnover of spermatogonia, spermatids, and spermatozoa. The overall findings offer an insight into the physiological significance of vasopressin and its homologs in vertebrates as a contributing factor in the multifactorial regulation of male reproduction.
Collapse
Affiliation(s)
- Maya Zanardini
- Department of Biological Sciences, University of Calgary, Calgary, AB 2500, Canada;
| | - Weimin Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China;
| | - Hamid R. Habibi
- Department of Biological Sciences, University of Calgary, Calgary, AB 2500, Canada;
| |
Collapse
|
4
|
Shinde D, Bhat SK, Ganesh CB. The opioid peptide leucine enkephalin modulates hypothalamic-hypophysial axis in the cichlid fish Oreochromis mossambicus. Anim Reprod Sci 2024; 263:107451. [PMID: 38490066 DOI: 10.1016/j.anireprosci.2024.107451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/25/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
In vertebrates, opioid peptides are thought to be involved in the regulation of reproduction; however, the significance of enkephalins in testicular function remains unclear. We examined the influence of δ-opioid receptor agonist leucine enkephalin (L-ENK) on the hypophysial-testicular axis of the cichlid fish Oreochromis mossambicus. Treatment with a low dose of L-ENK (60 µg) caused a significant increase in the numbers of primary and secondary spermatocytes and early and late spermatids, concomitant with intense immunolabelling of testicular androgen receptors, but did not significantly alter serum luteinizing hormone (LH) and 11-ketotestosterone (11-KT) levels compared to those of controls. Nevertheless, treatment with a high dose of L-ENK (200 µg) caused a significant reduction in the numbers of secondary spermatocytes as well as late spermatids associated with marginal immunolabelling of androgen receptors and significantly lower concentrations of serum 11-KT and LH compared to controls. In addition, the serum cortisol level was not affected in low-dose L-ENK-treated fish, but its level was significantly increased in the high-dose L-ENK-treated group. Together, these findings indicate that a low dose of L-ENK stimulates the germ cells at the meiosis stage and promotes further stages of spermatogenesis, whereas a high concentration of L-ENK inhibits spermatogenesis at the advanced stages. This effect appears to be mediated through the suppression of testicular steroidogenesis and the reduction of LH release in the pituitary gland of tilapia. The findings also suggest that elevated L-ENK levels in teleosts may exert their inhibitory influence on the hypophysial-testicular axis via glucocorticoids.
Collapse
Affiliation(s)
- Deepak Shinde
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad 580 003, India
| | - Shilpa K Bhat
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad 580 003, India
| | - C B Ganesh
- Neuroendocrinology Research Laboratory, Department of Studies in Zoology, Karnatak University, Dharwad 580 003, India.
| |
Collapse
|
5
|
Fernandes da Costa D, de Oliveira Ribeiro A, Morena Bonita Ricci J, da Silva Rodrigues M, Antonio de Oliveira M, Felipe da Rosa I, Benites Doretto L, Takahiro Nakajima R, Henrique Nóbrega R. A83-01 and DMH1 effects in the zebrafish spermatogonial niche: Unraveling the roles of TGF-β and BMP signaling in the Fsh-mediated spermatogonial fate. Gene 2024; 897:148082. [PMID: 38101710 DOI: 10.1016/j.gene.2023.148082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
Transforming growth factor-β (TGF-β) and bone morphogenetic protein (BMP) signaling has fundamental roles in the regulation of the stem cell niche for both embryonic and adult stem cells. In zebrafish, male germ stem cell niche is regulated by follicle-stimulating hormone (Fsh) through different members of the TGF-β superfamily. On the other hand, the specific roles of TGF-β and BMP signaling pathways are unknown in the zebrafish male germ stem cell niche. Considering this lack of information, the present study aimed to investigate the pharmacological inhibition of TGF-β (A83-01) and BMP (DMH1) signaling pathways in the presence of recombinant zebrafish Fsh using testicular explants. We also reanalyzed single cell-RNA sequencing (sc-RNA-seq) dataset from adult zebrafish testes to identify the testicular cellular sites of smad expression, and to understand the physiological significance of the changes in smad transcript levels after inhibition of TGF-β or BMP pathways. Our results showed that A83-01 potentiated the pro-stimulatory effects of Fsh on spermatogonial differentiation leading to an increase in the proportion area occupied by differentiated spermatogonia with concomitant reduction of type A undifferentiated (Aund) spermatogonia. In agreement, expression analysis showed lower mRNA levels for the pluripotency gene pou5f3, and increased expression of dazl (marker of type B spermatogonia and spermatocyte) and igf3 (pro-stimulatory growth factor) following the co-treatment with TGF-β inhibitor and Fsh. Contrariwise, the inhibition of BMP signaling nullified the pro-stimulatory effects of Fsh, resulting in a reduction of differentiated spermatogonia and increased proportion area occupied by type Aund spermatogonia. Supporting this evidence, BMP signaling inhibition increased the mRNA levels of pluripotency genes nanog and pou5f3, and decreased dazl levels when compared to control. The sc-RNA-seq data unveiled a distinctive pattern of smad expression among testicular cells, primarily observed in spermatogonia (smad 2, 3a, 3b, 8), spermatocytes (smad 2, 3a, 8), Sertoli cells (smad 1, 3a, 3b), and Leydig cells (smad 1, 2). This finding supports the notion that inhibition of TGF-β and BMP signaling pathways may predominantly impact cellular components within the spermatogonial niche, namely spermatogonia, Sertoli, and Leydig cells. In conclusion, our study demonstrated that TGF-β and BMP signaling pathways exert antagonistic roles in the zebrafish germ stem cell niche. The members of the TGF-β subfamily are mainly involved in maintaining the undifferentiated state of spermatogonia, while the BMP subfamily promotes spermatogonial differentiation. Therefore, in the complex regulation of the germ stem cell niche by Fsh, members of the BMP subfamily (pro-differentiation) should be more predominant in the niche than those belonging to the TGF-β (anti-differentiation). Overall, these findings are not only relevant for understanding the regulation of germ stem cell niche but may also be useful for expanding in vitro the number of undifferentiated spermatogonia more efficiently than using recombinant hormones or growth factors.
Collapse
Affiliation(s)
- Daniel Fernandes da Costa
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970 Botucatu, São Paulo, Brazil
| | - Amanda de Oliveira Ribeiro
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970 Botucatu, São Paulo, Brazil
| | - Juliana Morena Bonita Ricci
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970 Botucatu, São Paulo, Brazil
| | - Maira da Silva Rodrigues
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970 Botucatu, São Paulo, Brazil
| | - Marcos Antonio de Oliveira
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970 Botucatu, São Paulo, Brazil
| | - Ivana Felipe da Rosa
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970 Botucatu, São Paulo, Brazil
| | - Lucas Benites Doretto
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970 Botucatu, São Paulo, Brazil
| | - Rafael Takahiro Nakajima
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970 Botucatu, São Paulo, Brazil
| | - Rafael Henrique Nóbrega
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970 Botucatu, São Paulo, Brazil; South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, Faculty of Fisheries and Protection of Waters, University of South Bohemia in Ceske Budejovice, 389 25 Vodňany, Czech Republic.
| |
Collapse
|
6
|
Andersson E, Schulz RW, Almeida F, Kleppe L, Skaftnesmo KO, Kjærner-Semb E, Crespo D, Fjelldal PG, Hansen TJ, Norberg B, Edvardsen RB, Wargelius A. Loss of Fshr Prevents Testicular Maturation in Atlantic Salmon (Salmo salar L.). Endocrinology 2024; 165:bqae013. [PMID: 38298132 PMCID: PMC10878062 DOI: 10.1210/endocr/bqae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/02/2024]
Abstract
Early puberty poses a significant challenge for male Atlantic salmon in aquaculture due to its negative impact on growth and welfare. The regulation of puberty in vertebrates involves 2 key reproductive hormones: follicle-stimulating hormone (FSH) and luteinizing hormone (LH) and their gonadal receptors. In male mice lacking FSH receptor, testes size is reduced, but fertility is maintained, while medaka and zebrafish with a disrupted fshr gene exhibit near normal testis size and fertility. In these fishes both Fsh and Lh are present during puberty and Lh may rescue fertility, while in salmonid fish only Fsh is present in the circulation during puberty. Using CRISPR-Cas9, we produced crispants with a high prevalence of fshr mutations at the target site, which remained fertile, although more than half showed a testis development deviating from wild-type (wt) males. Crossing out these F0 crispants to each other produced a viable F1 generation showing frameshift (fshr-/-) or in-frame mutations (fshrif/if). Nearly all wt males matured while all fshr-/- males remained immature with small testes containing A spermatogonia as the furthest developed germ cell type and prepubertal plasma androgen levels. Also, the pituitary transcript levels of gnrhr2bba and lhb, but not for fshb, were reduced in the fshr-/- males compared with maturing males. More than half of the fshrif/if mutant males showed no or a delayed maturation. In conclusion, Atlantic salmon show the unique characteristic that loss of Fshr function alone results in male infertility, offering new opportunities to control precocious puberty or fertility in salmon.
Collapse
Affiliation(s)
- Eva Andersson
- Institute of Marine Research, NO-5817 Bergen, Norway
| | - Rüdiger W Schulz
- Institute of Marine Research, NO-5817 Bergen, Norway
- Science Faculty, Department Biology, Utrecht University, NL-3584 CH Utrecht, The Netherlands
| | | | - Lene Kleppe
- Institute of Marine Research, NO-5817 Bergen, Norway
| | | | | | - Diego Crespo
- Institute of Marine Research, NO-5817 Bergen, Norway
| | | | | | | | | | | |
Collapse
|
7
|
Liao X, Tao B, Zhang X, Chen L, Chen J, Song Y, Hu W. Loss of gdnfa disrupts spermiogenesis and male courtship behavior in zebrafish. Mol Cell Endocrinol 2023; 576:112010. [PMID: 37419437 DOI: 10.1016/j.mce.2023.112010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/19/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Spermatogenesis is essential for establishment and maintenance of reproduction in male vertebrates. Spermatogenesis, which is mainly regulated by the combined action of hormones, growth factors, and epigenetic factors, is highly conserved. Glial cell line-derived neurotrophic factor (GDNF) is a member of the transforming growth factor-β superfamily. In this study, global gdnfa knockout and Tg (gdnfa: mcherry) transgenic zebrafish lines were generated. Loss of gdnfa resulted in disorganized testes, decreased gonadosomatic index, and low percentage of mature spermatozoa. In the Tg (gdnfa: mcherry) zebrafish line, we found that gdnfa was expressed in Leydig cells. The mutation in gdnfa significantly decreased Leydig cell marker gene expression and androgen secretion in Leydig cells. In addition, courtship behavior was disrupted in the male mutants. We present in vivo data showing that global knockout of gdnfa disrupts spermiogenesis and male courtship behavior in zebrafish. The first viable vertebrate model with a global gdnfa knockout may be valuable for studying the role of GDNF in animal reproduction.
Collapse
Affiliation(s)
- Xianyao Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Binbin Tao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China.
| | - Xiya Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lu Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ji Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Yanlong Song
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China
| | - Wei Hu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Hubei Hongshan Laboratory, Wuhan, 430072, China; Guangdong Laboratory for Lingnan Modem Agriculture, China; Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
8
|
Sposato AL, Llewellyn DR, Weber JM, Hollins HL, Schrock MN, Farrell JA, Gagnon JA. Germ cells do not progress through spermatogenesis in the infertile zebrafish testis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.05.556432. [PMID: 37732254 PMCID: PMC10508784 DOI: 10.1101/2023.09.05.556432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Vertebrate spermatogonial stem cells maintain sperm production over the lifetime of an animal but fertility declines with age. While morphological studies have greatly informed our understanding of typical spermatogenesis, the molecular and cellular mechanisms underlying spermatogenesis are not yet understood, particularly with respect to the onset of fertility. We used single-cell RNA sequencing to generate a developmental atlas of the zebrafish testis. Using 5 timepoints across the adult life of a zebrafish, we described cellular profiles in the testis during and after fertility. While all germ cell stages of spermatogenesis are detected in testes from fertile adult zebrafish, testes from older infertile males only contained spermatogonia and a reduced population of spermatocytes. These remaining germ cells are transcriptionally distinct from fertile spermatogonia. Immune cells including macrophages and lymphocytes drastically increase in abundance in infertile testes. Our developmental atlas reveals the cellular changes as the testis ages and defines a molecular roadmap for the regulation of male fertility.
Collapse
Affiliation(s)
- Andrea L. Sposato
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112
| | | | - Jenna M. Weber
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112
| | - Hailey L. Hollins
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112
| | - Madison N. Schrock
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112
| | - Jeffrey A. Farrell
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814
| | - James A. Gagnon
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112
- Henry Eyring Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
9
|
Zupa R, Duncan N, Giménez I, Mylonas CC, Pousis C, Passantino L, Cuko R, Corriero A. Male germ cell proliferation and apoptosis in sexually immature meagre Argyrosomus regius (Asso, 1801) treated with recombinant follicle stimulating hormone. Sci Rep 2023; 13:7013. [PMID: 37117257 PMCID: PMC10147655 DOI: 10.1038/s41598-023-34102-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/24/2023] [Indexed: 04/30/2023] Open
Abstract
The meagre Argyrosomus regius (Asso, 1801) is a marine fish species that has an increasing aquaculture production in Europe. Lowering the age at maturity of hatchery-produced juveniles would support meagre aquaculture by reducing time between generations in selective breeding programs and reducing industrial costs for broodstock maintenance. The aim of this work was to assess the effects of a treatment with recombinant follicle stimulating hormone (rFsh), produced in ovarian cells of Chinese hamsters, on male germ cell proliferation and apoptosis in sexually immature meagre. The rFsh-treated fish had higher gonadosomatic index, larger seminiferous tubules, more abundant luminal spermatozoa, a lower density of anti-PCNA positive single A spermatogonia, a higher density of anti-PCNA positive spermatocysts and a lower incidence of germ cell apoptosis than control groups. The present study demonstrated the effectiveness of the produced rFsh in stimulating testis development and spermatogenesis in pre-pubertal meagre. Moreover, the rFsh treatment proved to be highly efficient in removing the apoptotic block of spermatogenesis observed in juvenile meagre, allowing spermatogonial survival and progress towards meiosis. The administration of rFsh did not stimulate spermatogonial self-renewal, a process whose control still needs to be elucidated.
Collapse
Affiliation(s)
- Rosa Zupa
- Department of Veterinary Medicine, University of Bari Aldo Moro, S.P. per Casamassima km.3, 70010, Valenzano, Bari, Italy
| | - Neil Duncan
- IRTA, Ctra. de Poble Nou km. 5.5, 43540, La Ràpita, Tarragona, Spain
| | - Ignacio Giménez
- Rara Avis Biotec, S. L., Calle Moratín 17, 46002, Valencia, Spain
| | - Constantinos C Mylonas
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research, 71003, Heraklion, Crete, Greece
| | - Chrysovalentinos Pousis
- Department of Veterinary Medicine, University of Bari Aldo Moro, S.P. per Casamassima km.3, 70010, Valenzano, Bari, Italy
| | - Letizia Passantino
- DiMePRe-J, University of Bari Aldo Moro, S.P. per Casamassima km.3, 70010, Valenzano, Bari, Italy
| | - Rezart Cuko
- Department of Veterinary Medicine, University of Bari Aldo Moro, S.P. per Casamassima km.3, 70010, Valenzano, Bari, Italy
| | - Aldo Corriero
- Department of Veterinary Medicine, University of Bari Aldo Moro, S.P. per Casamassima km.3, 70010, Valenzano, Bari, Italy.
| |
Collapse
|
10
|
Rodrigues MS, Tovo-Neto A, Rosa IF, Doretto LB, Fallah HP, Habibi HR, Nóbrega RH. Thyroid Hormones Deficiency Impairs Male Germ Cell Development: A Cross Talk Between Hypothalamic-Pituitary-Thyroid, and—Gonadal Axes in Zebrafish. Front Cell Dev Biol 2022; 10:865948. [PMID: 35646887 PMCID: PMC9133415 DOI: 10.3389/fcell.2022.865948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/21/2022] [Indexed: 11/18/2022] Open
Abstract
In vertebrates, thyroid hormones are critical players in controlling different physiological processes such as development, growth, metabolism among others. There is evidence in mammals that thyroid hormones are also an important component of the hormonal system that controls reproduction, although studies in fish remain poorly investigated. Here, we tested this hypothesis by investigating the effects of methimazole-induced hypothyroidism on the testicular function in adult zebrafish. Treatment of fish with methimazole, in vivo, significantly altered zebrafish spermatogenesis by inhibiting cell differentiation and meiosis, as well as decreasing the relative number of spermatozoa. The observed impairment of spermatogenesis by methimazole was correlated with significant changes in transcript levels for several genes implicated in the control of reproduction. Using an in vitro approach, we also demonstrated that in addition to affecting the components of the brain-pituitary-peripheral axis, T3 (triiodothyronine) also exerts direct action on the testis. These results reinforce the hypothesis that thyroid hormones are an essential element of multifactorial control of reproduction and testicular function in zebrafish and possibly other vertebrate species.
Collapse
Affiliation(s)
- Maira S. Rodrigues
- Aquaculture Program (CAUNESP), São Paulo State University (UNESP), São Paulo, Brazil
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Aldo Tovo-Neto
- Aquaculture Program (CAUNESP), São Paulo State University (UNESP), São Paulo, Brazil
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Ivana F. Rosa
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Lucas B. Doretto
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Hamideh P. Fallah
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Hamid R. Habibi
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Rafael H. Nóbrega
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
- *Correspondence: Rafael H. Nóbrega,
| |
Collapse
|
11
|
Transcriptomes of testis and pituitary from male Nile tilapia (O. niloticus L.) in the context of social status. PLoS One 2022; 17:e0268140. [PMID: 35544481 PMCID: PMC9094562 DOI: 10.1371/journal.pone.0268140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/22/2022] [Indexed: 11/19/2022] Open
Abstract
African cichlids are well established models for studying social hierarchies in teleosts and elucidating the effects social dominance has on gene expression. Ascension in the social hierarchy has been found to increase plasma levels of steroid hormones, follicle stimulating hormone (Fsh) and luteinizing hormone (Lh) as well as gonadosomatic index (GSI). Furthermore, the expression of genes related to gonadotropins and steroidogenesis and signaling along the brain-pituitary-gonad axis (BPG-axis) is affected by changes of an animal’s social status. In this study, we use RNA-sequencing to obtain an in-depth look at the transcriptomes of testes and pituitaries from dominant and subordinate male Nile tilapia living in long-term stable social hierarchies. This allows us to draw conclusions about factors along the brain-pituitary-gonad axis that are involved in maintaining dominance over weeks or even months. We identify a number of genes that are differentially regulated between dominant and subordinate males and show that in high-ranking fish this subset of genes is generally upregulated. Genes differentially expressed between the two social groups comprise growth factors, related binding proteins and receptors, components of Wnt-, Tgfβ- and retinoic acid-signaling pathway, gonadotropin signaling and steroidogenesis pathways. The latter is backed up by elevated levels of 11-ketotestosterone, testosterone and estradiol in dominant males. Luteinizing hormone (Lh) is found in higher concentration in the plasma of long-term dominant males than in subordinate animals. Our results both strengthen the existing models and propose new candidates for functional studies to expand our understanding of social phenomena in teleost fish.
Collapse
|
12
|
Zhao H, Zhang L, Li Q, Zhao Z, Duan Y, Huang Z, Ke H, Liu C, Li H, Liu L, Du J, Wei Z, Mou C, Zhou J. Integrated analysis of the miRNA and mRNA expression profiles in Leiocassis longirostris at gonadal maturation. Funct Integr Genomics 2022; 22:655-667. [PMID: 35467220 DOI: 10.1007/s10142-022-00857-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 03/09/2022] [Accepted: 03/22/2022] [Indexed: 11/04/2022]
Abstract
Leiocassis longirostris is a commercially important fish species that shows a sexually dimorphic growth pattern. A lack of molecular data from the gonads of this species has hindered research and selective breeding efforts. In this study, we conducted a comprehensive analysis of the expression profile of miRNA and mRNA to explore their regulatory roles in the gonadal maturation stage of L. longirostris. We identified 60 differentially expressed miRNAs and 20,752 differentially expressed genes by sequencing. A total of 90 miRNAs and 21 target genes involved in gonad development and sex determination were identified. Overall, the results of this study enhance our understanding of the molecular mechanisms underlying sex determination and differentiation and provide valuable genomic information for the selective breeding of L. longirostris.
Collapse
Affiliation(s)
- Han Zhao
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Lu Zhang
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Qiang Li
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Zhongmeng Zhao
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Yuanliang Duan
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Zhipeng Huang
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Hongyu Ke
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Chao Liu
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Huadong Li
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Lu Liu
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Jun Du
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Zhen Wei
- Leiocassis Longirostris Foundation Seed Farm, Sichuan Province, China
| | - Chengyan Mou
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Jian Zhou
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China.
| |
Collapse
|
13
|
Gdnf Acts as a Germ Cell-Derived Growth Factor and Regulates the Zebrafish Germ Stem Cell Niche in Autocrine- and Paracrine-Dependent Manners. Cells 2022; 11:cells11081295. [PMID: 35455974 PMCID: PMC9030868 DOI: 10.3390/cells11081295] [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: 12/24/2021] [Revised: 03/27/2022] [Accepted: 04/06/2022] [Indexed: 11/30/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) and its receptor (GDNF Family Receptor α1-GFRα1) are well known to mediate spermatogonial stem cell (SSC) proliferation and survival in mammalian testes. In nonmammalian species, Gdnf and Gfrα1 orthologs have been found but their functions remain poorly investigated in the testes. Considering this background, this study aimed to understand the roles of the Gdnf-Gfrα1 signaling pathway in zebrafish testes by combining in vivo, in silico and ex vivo approaches. Our analysis showed that zebrafish exhibit two paralogs for Gndf (gdnfa and gdnfb) and its receptor, Gfrα1 (gfrα1a and gfrα1b), in accordance with a teleost-specific third round of whole genome duplication. Expression analysis further revealed that both ligands and receptors were expressed in zebrafish adult testes. Subsequently, we demonstrated that gdnfa is expressed in the germ cells, while Gfrα1a/Gfrα1b was detected in early spermatogonia (mainly in types Aund and Adiff) and Sertoli cells. Functional ex vivo analysis showed that Gdnf promoted the creation of new available niches by stimulating the proliferation of both type Aund spermatogonia and their surrounding Sertoli cells but without changing pou5f3 mRNA levels. Strikingly, Gdnf also inhibited late spermatogonial differentiation, as shown by the decrease in type B spermatogonia and down-regulation of dazl in a co-treatment with Fsh. Altogether, our data revealed that a germ cell-derived factor is involved in maintaining germ cell stemness through the creation of new available niches, supporting the development of spermatogonial cysts and inhibiting late spermatogonial differentiation in autocrine- and paracrine-dependent manners.
Collapse
|
14
|
Shinde D, Ganesh CB. Chronic exposure to aquacultural stressors affects pituitary-testis axis in the Mozambique tilapia Oreochromis mossambicus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:437-448. [PMID: 35201519 DOI: 10.1007/s10695-022-01061-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/12/2022] [Indexed: 06/14/2023]
Abstract
Reproduction in fish is modulated by several factors that include environmental and endocrine components. The aim of this study was to elucidate the effect of aquacultural stressors along the pituitary-testis axis in a continuously breeding cichlid fish Oreochromis mossambicus. The fish (35.05 ± 2.20 g) were divided into three groups (n = 10 in each group, n = 5 in each replicate), namely initial controls (euthanized on the day of initiation of experiment), time-matched controls (kept undisturbed), and stressed fish, which were subjected to different kinds of randomized aquacultural stressors such as handling, chasing, frequent netting, and low water levels, daily for a period of 21 days. Although the gonadosomatic index and the mean numbers of spermatogonia-A and spermatogonia-B did not differ significantly among different experimental groups, significant decrease was observed in the mean numbers of primary spermatocytes, secondary spermatocytes, early spermatids, and late spermatids in fish exposed to stressors compared to those of initial controls and time-matched controls. While the diameter of the seminiferous lobule was significantly lower, the size of the lumen and the serum levels of cortisol were significantly increased in stressed fish compared with initial controls and time-matched controls. Furthermore, weak androgen receptor immunoreactivity was observed in the Sertoli cells of the testis in contrast to the strongly immunoreactive androgen receptors in initial controls and time-matched controls. Concomitant with this, there was a significant decrease in the percent area and the intensity of luteinizing hormone (LH) immunoreactive content in the proximal pars distalis (PPD) region of the pituitary gland in stressed fish compared with initial controls and time-matched controls. Overall, these results suggest that exposure to chronic aquacultural stressors causes suppression of LH synthesis in the pituitary gland concomitant with decreased androgen receptor expression and blockade of recruitment of germline cells at the meiosis stage. This inhibition appears to be mediated through the hypothalamic-pituitary-interrenal axis in the tilapia O. mossambicus.
Collapse
Affiliation(s)
- Deepak Shinde
- Department of Studies in Zoology, Karnatak University, Dharwad, 580 003, India
| | - C B Ganesh
- Department of Studies in Zoology, Karnatak University, Dharwad, 580 003, India.
| |
Collapse
|
15
|
Roles of Gonadotropin Receptors in Sexual Development of Medaka. Cells 2022; 11:cells11030387. [PMID: 35159197 PMCID: PMC8834109 DOI: 10.3390/cells11030387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 12/10/2022] Open
Abstract
The gonadotropins, follicle-stimulating hormone (FSH) and luteinizing hormone (LH), are secreted from the pituitary and bind to the FSH receptor (FSHR) and LH receptor (LHR) to regulate gonadal development in vertebrates. Previously, using fshr-knockout (KO) medaka (Oryzias latipes), we demonstrated that FSH regulates ovarian development by elevating estrogen levels. However, the lhr-KO phenotype in medaka is poorly characterized. Here, we generated lhr-KO medaka using the transcription activator-like effector nuclease (TALEN) technique. We analyzed its phenotype and that of fshr-KO, lhr;fshr double-heterozygotes (double-hetero), and double-KO fish. All genetically male medaka displayed normal testes and were fertile, whereas fshr-KO and double-KO genetically female fish displayed small ovaries containing many early pre-vitellogenic oocytes and were infertile. Although lhr-KO genetically female fish had normal ovaries with full-grown oocytes, ovulation did not occur. Levels of 17α,20β-dihydroxy-4-pregnen-3-one, which is required for meiotic maturation of oocytes and sperm maturation in teleost fish, were significantly decreased in all KO female medaka ovaries except for double-heteros. Further, 17β-estradiol levels in fshr-KO and double-KO ovaries were significantly lower than those in double-heteros. These findings indicate that LH is necessary for oocyte maturation and FSH is necessary for follicle development, but that neither are essential for spermatogenesis in medaka.
Collapse
|
16
|
Evidence-based hormonal, mutational, and endocrine-disrupting chemical-induced zebrafish as an alternative model to study PCOS condition similar to mammalian PCOS model. Life Sci 2022; 291:120276. [PMID: 34990650 DOI: 10.1016/j.lfs.2021.120276] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022]
Abstract
Polycystic ovarian syndrome (PCOS) causes swollen ovaries in women at reproductive age due to hormonal disorder with small cysts on the outer edges. The cause of the disorder is still yet to be found. Multiple factors have increased PCOS prevalence, hyperandrogenism, oxidative stress, inflammation, and insulin resistance. Various animal PCOS models have been developed to imitate the pathophysiology of PCOS in humans. Zebrafish is one of the most versatile animal experimental models because of the transparency of the embryos, small size, and rapid growth. The zebrafish similarity to higher vertebrates made it a useful non-mammalian model for PCOS drug testing and screening. This review provides an insight into the usage of zebrafish, a non-mammalian model for PCOS, as an opportunity for evaluating future initiatives in such a research domain.
Collapse
|
17
|
Crespo D, Skaftnesmo KO, Kjærner-Semb E, Yilmaz O, Norberg B, Olausson S, Vogelsang P, Bogerd J, Kleppe L, Edvardsen RB, Andersson E, Wargelius A, Hansen TJ, Fjelldal PG, Schulz RW. Pituitary Gonadotropin Gene Expression During Induced Onset of Postsmolt Maturation in Male Atlantic Salmon: In Vivo and Tissue Culture Studies. Front Endocrinol (Lausanne) 2022; 13:826920. [PMID: 35370944 PMCID: PMC8964956 DOI: 10.3389/fendo.2022.826920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/17/2022] [Indexed: 12/25/2022] Open
Abstract
Precocious male maturation causes reduced welfare and increased production costs in Atlantic salmon (Salmo salar) aquaculture. The pituitary produces and releases follicle-stimulating hormone (Fsh), the gonadotropin triggering puberty in male salmonids. However, little is known about how Fsh production is regulated in Atlantic salmon. We examined, in vivo and ex vivo, transcriptional changes of gonadotropin-related genes accompanying the initial steps of testis maturation, in pituitaries of males exposed to photoperiod and temperature conditions promoting maturation (constant light and 16°C). Pituitary fshb, lhb and gnrhr2bba transcripts increased in vivo in maturing males (gonado-somatic index > 0.1%). RNA sequencing (RNAseq) analysis using pituitaries from genetically similar males carrying the same genetic predisposition to mature, but differing by responding or not responding to stimulatory environmental conditions, revealed 144 differentially expressed genes, ~2/3rds being up-regulated in responders, including fshb and other pituitary hormones, steroid-related and other puberty-associated transcripts. Functional enrichment analyses confirmed gene involvement in hormone/steroid production and gonad development. In ex vivo studies, whole pituitaries were exposed to a selection of hormones and growth factors. Gonadotropin-releasing hormone (Gnrh), 17β-estradiol (E2) and 11-ketotestosterone (11-KT) up-regulated gnrhr2bba and lhb, while fshb was up-regulated by Gnrh but down-regulated by 11-KT in pituitaries from immature males. Also pituitaries from maturing males responded to Gnrh and sex steroids by increased gnrhr2bba and lhb transcript levels, but fshb expression remained unchanged. Growth factors (inhibin A, activin A and insulin-like growth factor 1) did not change gnrhr2bba, lhb or fshb transcript levels in pituitaries either from immature or maturing males. Additional pituitary ex vivo studies on candidates identified by RNAseq showed that these transcripts were preferentially regulated by Gnrh and sex steroids, but not by growth factors, and that Gnrh/sex steroids were less effective when incubating pituitaries from maturing males. Our results suggest that a yet to be characterized mechanism up-regulating fshb expression in the salmon pituitary is activated in response to stimulatory environmental conditions prior to morphological signs of testis maturation, and that the transcriptional program associated with this mechanism becomes unresponsive or less responsive to most stimulators ex vivo once males had entered pubertal developmental in vivo.
Collapse
Affiliation(s)
- Diego Crespo
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
- *Correspondence: Diego Crespo,
| | - Kai Ove Skaftnesmo
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Erik Kjærner-Semb
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Ozlem Yilmaz
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | - Birgitta Norberg
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | - Sara Olausson
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | - Petra Vogelsang
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Jan Bogerd
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht, Netherlands
| | - Lene Kleppe
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Rolf B. Edvardsen
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Eva Andersson
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Anna Wargelius
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Tom J. Hansen
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Matre Research Station, Matredal, Norway
| | - Per Gunnar Fjelldal
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Matre Research Station, Matredal, Norway
| | - Rüdiger W. Schulz
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
18
|
Zhang Y, Guo J, Chen Y, Wang J, Zhou Q, Chen M, Wang C. Embryonic exposure to fenbuconazole inhibits gametogenesis in adult zebrafish by targeting gonads not brain. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:112967. [PMID: 34773848 DOI: 10.1016/j.ecoenv.2021.112967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 06/13/2023]
Abstract
Fenbuconazole (FBZ) is widely used in agriculture. The current study was conducted to evaluate the influence of embryonic exposure to FBZ on reproduction in adult zebrafish. Embryos were exposed to 5, 50 and 500 ng/L FBZ for 72 h and then raised in clean water until adulthood. The result showed that the percentage of mature gametes was significantly reduced in adult zebrafish. The fertilization rate and survival rate of F1 embryos were decreased when the exposed fish were mated with untreated fish. The transcription of brain gnrh3, fshβ and lhγ in adult fish was upregulated, while the levels of 17β-estradiol and testosterone were not significantly changed in all treated groups, indicating that the reproduction-related genes in brain was not responsible for the reduced reproductive ability. The downregulated transcription of fshr, lhr, ar and esr2 in the gonads indicated the dysfunction of Sertoli and Leydig cells. Notably, downregulated transcription and upregulated methylation levels of genes related to germ cells were observed in treated F0 larvae and adult gonads. The elevated methylation levels of piwil1 and dnmt6 in the testes and vasa and dazl in the ovary were matched with the alterations in the expression of these genes, suggesting that germ cells are the main targets of FBZ. These results provide new mechanism underlying reproductive toxicity in fish caused by chemicals, and give potential retroactive biomarkers for monitoring reproductive toxic pollutants.
Collapse
Affiliation(s)
- Ying Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Jiaojiao Guo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Ying Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Jiangwei Wang
- Key Laboratory of the Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Qian Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Meng Chen
- Key Laboratory of the Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian 361005, PR China.
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China.
| |
Collapse
|
19
|
Rodrigues MS, Fallah HP, Zanardini M, Malafaia G, Habibi HR, Nóbrega RH. Interaction between thyroid hormones and gonadotropin inhibitory hormone in ex vivo culture of zebrafish testis: An approach to study multifactorial control of spermatogenesis. Mol Cell Endocrinol 2021; 532:111331. [PMID: 34038752 DOI: 10.1016/j.mce.2021.111331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022]
Abstract
Reproduction is under multifactorial control of neurohormones, pituitary gonadotropins, as well as of local gonadal signaling systems including sex steroids, growth factors and non-coding RNAs. Among the factors, gonadotropin-inhibitory hormone (Gnih) is a novel RFamide neuropeptide which directly modulates gonadotropin synthesis and release from pituitary, and in the gonads, Gnih mediated inhibitory actions on gonadotropin response of zebrafish spermatogenesis. Thyroid hormones are peripheral hormones which are also known to interact with reproductive axis, in particular, regulating testicular development and function. This study investigated the interaction between Gnih and thyroid hormones in zebrafish spermatogenesis using in vivo and ex vivo approaches. Three experimental groups were established: "control" (non-treated fish), "methimazole" and "methimazole + T4". Fish were exposed to goitrogen methimazole for 3 weeks; T4 (100 μg/L) was added in the water from the second week only in the "reversal treatment" group. After exposure, testes were dissected out and immediately incubated in Leibovitz's L-15 culture medium containing hCG, Gnih or hCG + Gnih for 7 days. Germ cell cysts and haploid cell population were evaluated by histomorphometry and flow cytometry, respectively. Our results showed that hypothyroidism affected germ cell development in basal and gonadotropin-induced spermatogenesis, in particular, meiosis and spermiogenesis. Hypothyroid testes showed lower amount of spermatozoa, and decreased potency of hCG. We also showed that goitrogen treatment nullified the inhibitory actions of Gnih on the gonadotropin-induced spermatogenesis. This study provided evidences that thyroid hormones are important regulatory factors for hCG- and Gnih-mediated functions in zebrafish spermatogenesis.
Collapse
Affiliation(s)
- Maira S Rodrigues
- Aquaculture Program (CAUNESP), São Paulo State University (UNESP), 14884-900, Jaboticabal, São Paulo, Brazil; Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970, Botucatu, São Paulo, Brazil
| | - Hamideh P Fallah
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Maya Zanardini
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Guilherme Malafaia
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970, Botucatu, São Paulo, Brazil; Biological Research Laboratory, Goiano Federal Institution, Urata Campus, Rodovia Geraldo Silva Nascimento, 2,5 km, Zona Rural, Urutaí, Goiás, Brazil
| | - Hamid R Habibi
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada.
| | - Rafael H Nóbrega
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), 18618-970, Botucatu, São Paulo, Brazil.
| |
Collapse
|
20
|
Cortisol modulates calcium release-activated calcium channel gating in fish hepatocytes. Sci Rep 2021; 11:9621. [PMID: 33953236 PMCID: PMC8100157 DOI: 10.1038/s41598-021-88957-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Glucocorticoids (GCs) are rapidly released in response to stress and play an important role in the physiological adjustments to re-establish homeostasis. The mode of action of GCs for stress coping is mediated largely by the steroid binding to the glucocorticoid receptor (GR), a ligand-bound transcription factor, and modulating the expression of target genes. However, GCs also exert rapid actions that are independent of transcriptional regulation by modulating second messenger signaling. However, a membrane-specific protein that transduces rapid GCs signal is yet to be characterized. Here, using freshly isolated hepatocytes from rainbow trout (Oncorhynchus mykiss) and fura2 fluorescence microscopy, we report that stressed levels of cortisol rapidly stimulate the rise in cytosolic free calcium ([Ca2+]i). Pharmacological manipulations using specific extra- and intra-cellular calcium chelators, plasma membrane and endoplasmic reticulum channel blockers and receptors, indicated extracellular Ca2+ entry is required for the cortisol-mediated rise in ([Ca2+]i). Particularly, the calcium release-activated calcium (CRAC) channel gating appears to be a key target for the rapid action of cortisol in the ([Ca2+]i) rise in trout hepatocytes. To test this further, we carried out in silico molecular docking studies using the Drosophila CRAC channel modulator 1 (ORAI1) protein, the pore forming subunit of CRAC channel that is highly conserved. The result predicts a putative binding site on CRAC for cortisol to modulate channel gating, suggesting a direct, as well as an indirect regulation (by other membrane receptors) of CRAC channel gating by cortisol. Altogether, CRAC channel may be a novel cortisol-gated Ca2+ channel transducing rapid nongenomic signalling in hepatocytes during acute stress.
Collapse
|
21
|
Oliveira MA, Martinez ERM, Butzge AJ, Doretto LB, Ricci JMB, Rodrigues MS, Vigoya AAA, Gómez-González NE, Stewart AB, Nóbrega RH. Molecular characterization and expression analysis of anti-Müllerian hormone in common carp (Cyprinus carpio) adult testes. Gene Expr Patterns 2021; 40:119169. [PMID: 33667682 DOI: 10.1016/j.gep.2021.119169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/11/2021] [Accepted: 02/26/2021] [Indexed: 11/24/2022]
Abstract
Anti-Müllerian hormone (Amh) is a member of the transforming growth factor-β (Tgf-β) superfamily required in the regression of Müllerian ducts during gonadal sex differentiation of higher vertebrates. Teleost fish lack Müllerian ducts, but identified Amh orthologs have been shown to exert crucial functions during sex determination and differentiation of several species of teleosts. However, the function of Amh during gametogenesis in adult fish remains poorly investigated. Therefore, to expand present knowledge on the role of Amh in teleosts, the present study aimed to isolate and clone full-length amh cDNA in the common carp, Cyprinus carpio, and examine its expression levels throughout the male reproductive cycle and in response to different hormone treatments of testicular explants. Molecular cloning and characterization showed that the common carp Amh precursor amino acid sequence shared common features to other fish Amh precursors, including a conserved C-terminus (Tgf-β domain) and a double proteolytic cleavage site (R-X-X-R-X-X-R) upstream to the Tgf-β domain. Expression analysis showed amh dimorphic expression in the adult gonads with higher expression in the testes than ovaries. In testes, amh mRNA was detected in Sertoli cells contacting different types of germ cells, although the expression was greatest in Sertoli cells associated with type A undifferentiated spermatogonia. Expression analysis during the reproductive cycle showed that amh transcripts were down-regulated during the developing phase, which is characterized by an increased proliferation of type A undifferentiated spermatogonia and Sertoli cells and appearance of spermatocytes (meiosis) in the testes. Furthermore, ex vivo experiments showed that a 7 day exposure to Fsh or estrogens was required to decrease amh mRNA levels in common carp testicular explants. In summary, this study provided information on the molecular characterization and transcript abundance of amh in common carp adult testes. Altogether, these data will be useful for further investigations on sex determination and differentiation in this species, and also to improved strategies for improved carp aquaculture, such as inhibiting precocious maturation of males.
Collapse
Affiliation(s)
- Marcos A Oliveira
- Aquaculture Program (CAUNESP), São Paulo State University (UNESP), Jaboticabal, São Paulo, Brazil; Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Emanuel R M Martinez
- Aquaculture Program (CAUNESP), São Paulo State University (UNESP), Jaboticabal, São Paulo, Brazil; Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Arno J Butzge
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Lucas B Doretto
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Juliana M B Ricci
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Maira S Rodrigues
- Aquaculture Program (CAUNESP), São Paulo State University (UNESP), Jaboticabal, São Paulo, Brazil; Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Angel A A Vigoya
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil; Faculty of Veterinary Medicine and Animal Science, San Martín University Foundation (FUSM), Bogotá, Colombia
| | - Núria E Gómez-González
- Department of Cell Biology and Histology, Faculty of Biology, Universidad de Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Amanda B Stewart
- Department of Orthopaedics Muscle skeletal Research, West Virginia University, USA
| | - Rafael H Nóbrega
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil.
| |
Collapse
|
22
|
Crespo D, Assis LHC, Zhang YT, Safian D, Furmanek T, Skaftnesmo KO, Norberg B, Ge W, Choi YC, den Broeder MJ, Legler J, Bogerd J, Schulz RW. Insulin-like 3 affects zebrafish spermatogenic cells directly and via Sertoli cells. Commun Biol 2021; 4:204. [PMID: 33589679 PMCID: PMC7884674 DOI: 10.1038/s42003-021-01708-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/13/2021] [Indexed: 01/31/2023] Open
Abstract
Pituitary hormones can use local signaling molecules to regulate target tissue functions. In adult zebrafish testes, follicle-stimulating hormone (Fsh) strongly increases the production of insulin-like 3 (Insl3), a Leydig cell-derived growth factor found in all vertebrates. Little information is available regarding Insl3 function in adult spermatogenesis. The Insl3 receptors Rxfp2a and 2b were expressed by type A spermatogonia and Sertoli and myoid cells, respectively, in zebrafish testis tissue. Loss of insl3 increased germ cell apoptosis in males starting at 9 months of age, but spermatogenesis appeared normal in fully fertile, younger adults. Insl3 changed the expression of 409 testicular genes. Among others, retinoic acid (RA) signaling was up- and peroxisome proliferator-activated receptor gamma (Pparg) signaling was down-regulated. Follow-up studies showed that RA and Pparg signaling mediated Insl3 effects, resulting in the increased production of differentiating spermatogonia. This suggests that Insl3 recruits two locally active nuclear receptor pathways to implement pituitary (Fsh) stimulation of spermatogenesis.
Collapse
Affiliation(s)
- Diego Crespo
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.10917.3e0000 0004 0427 3161Present Address: Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Luiz H. C. Assis
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Yu Ting Zhang
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Fujian, PR China ,grid.449133.80000 0004 1764 3555Present Address: Institute of Oceanography, Minjiang University, Fuzhou, PR China
| | - Diego Safian
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.4818.50000 0001 0791 5666Present Address: Experimental Zoology Group and Aquaculture and Fisheries Group, Department of Animal Science, Wageningen University, Wageningen, The Netherlands
| | - Tomasz Furmanek
- grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Kai Ove Skaftnesmo
- grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| | - Birgitta Norberg
- grid.10917.3e0000 0004 0427 3161Institute of Marine Research, Austevoll Research Station, Storebø, Norway
| | - Wei Ge
- grid.437123.00000 0004 1794 8068Center of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Yung-Ching Choi
- grid.437123.00000 0004 1794 8068Center of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau China
| | - Marjo J. den Broeder
- grid.5477.10000000120346234Division of Toxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Juliette Legler
- grid.5477.10000000120346234Division of Toxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jan Bogerd
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Rüdiger W. Schulz
- grid.5477.10000000120346234Reproductive Biology Group, Division Developmental Biology, Department of Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands ,grid.10917.3e0000 0004 0427 3161Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen, Norway
| |
Collapse
|
23
|
Fallah HP, Rodrigues MS, Zanardini M, Nóbrega RH, Habibi HR. Effects of gonadotropin-inhibitory hormone on early and late stages of spermatogenesis in ex-vivo culture of zebrafish testis. Mol Cell Endocrinol 2021; 520:111087. [PMID: 33249103 DOI: 10.1016/j.mce.2020.111087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/27/2020] [Accepted: 11/16/2020] [Indexed: 11/29/2022]
Abstract
Gonadotropin-inhibitory hormone (Gnih) is known to play a role in the regulation of reproduction in vertebrates by influencing gonadotropin release and synthesis. While the endocrine actions of Gnih have been identified in several species, its paracrine/autocrine effects in the control of spermatogenesis are less defined. We have used ex vivo culture of zebrafish testis to investigate the role of gonadal zebrafish Gnih (zGnih) in the regulation of the spermatogenic process. We used FACScan cell cycle analysis, morphometric quantifications, BrdU incorporation and caspase-3 activity assays as well as measuring 11-Ketotestosterone (11-KT) level in the culture media. FACScan analysis and morphometric quantification results demonstrated direct action of zGnih on basal and gonadotropin (Lh and Fsh)-induced spermatogenesis. Treatment with zGnih (10 nM) significantly decreased the number of G0/G1 cells after 7-days of culture while no significant changes were found in the proportion area of spermatogonia cell types. Investigation of DNA synthesis using BrdU (5-Bromo-2'-Deoxyuridine) labeling showed that treatment with zGnih (10 nM) significantly decreased proliferative activity of type A spermatogonia, while increased the mitotic activity of type B spermatogonia. We also showed that treatment with zGnih (100 nM) completely eliminated 11-KT release induced by 100 ng/ml Fsh. Treatment with zGnih (10 and 100 nM) also inhibited both hCG and Fsh-induced spermatogenesis. These results, plus our previous findings, demonstrate that zGnih produced locally in the testis is a component of a complex multifactorial system that regulates testicular function in zebrafish.
Collapse
Affiliation(s)
- Hamideh P Fallah
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Maira S Rodrigues
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada; Department of Morphology, Reproductive and Molecular Biology Group, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Maya Zanardini
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Rafael H Nóbrega
- Department of Morphology, Reproductive and Molecular Biology Group, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Hamid R Habibi
- Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada.
| |
Collapse
|
24
|
Jorge BC, Reis ACC, Sterde ÉT, Balin PDS, Scarano WR, Hisano H, Arena AC. Exposure to benzo(a)pyrene from juvenile period to peripubertal impairs male reproductive parameters in adult rats. CHEMOSPHERE 2021; 263:128016. [PMID: 33297042 DOI: 10.1016/j.chemosphere.2020.128016] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 06/12/2023]
Abstract
Benzo(a)pyrene (BaP) is a persistent organic pollutant and endocrine disruptor that can compromise the steroidogenesis process by interacting with the StAR protein, causing adverse effects on male reproduction. However, consequences of prepubertal BaP exposure and its impacts on adult life are yet unknown. This study investigated the effects of BaP exposure from the juvenile period to peripubertal on reproductive parameters in adult male rats. Males were exposed to 0; 0.1; 1 or 10 μg/kg/day of BaP from post-natal (PND) 23 to PND 53 (by gavage). The lowest dose of BaP was able to compromise the male copulatory behavior, as evidenced by the delay in the first mount, intromission and ejaculation. Furthermore, BaP-treated groups showed lower sperm quality (disrupted motility and morphology) and quantity, reduced relative weights of the thyroid and seminal gland. Serum testosterone levels and the Leydig cells nuclei volume were decreased by BaP exposure whereas the StAR expression was increased. Histopathological changes in the testis also were detected in the males exposed to BaP. These results showed that prepubertal BaP-exposure adversely influenced the male reproductive system in the adult life, indicating that a comprehensive risk assessment of BaP-exposure on prepubertal period is necessary.
Collapse
Affiliation(s)
- Bárbara Campos Jorge
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | - Ana Carolina Casali Reis
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | - Érika Tissiana Sterde
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | - Paola da Silva Balin
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | - Wellerson Rodrigo Scarano
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil
| | | | - Arielle Cristina Arena
- Department of Structural and Functional Biology, Institute of Biosciences of Botucatu, Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil; Center of Toxicological Assistance (CEATOX), Institute of Biosciences of Botucatu, Univ. Estadual Paulista - Botucatu (UNESP), São Paulo, Brazil.
| |
Collapse
|
25
|
Molés G, Hausken K, Carrillo M, Zanuy S, Levavi-Sivan B, Gómez A. Generation and use of recombinant gonadotropins in fish. Gen Comp Endocrinol 2020; 299:113555. [PMID: 32687933 DOI: 10.1016/j.ygcen.2020.113555] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 07/11/2020] [Accepted: 07/14/2020] [Indexed: 02/09/2023]
Abstract
Understanding the differential roles of the pituitary gonadotropins Fsh and Lh in gonad maturation is crucial for a successful manipulation of the reproductive process in fish, and requires species-specific tools and appropriate active hormones. With the increasing availability of fish cDNAs coding for gonadotropin subunits, the production of recombinant hormones in heterologous systems has gradually substituted the approach of isolating native hormones. These recombinant hormones can be continually produced without depending on the fish as starting material and no cross-contamination with other pituitary glycoproteins is assured. Recombinant gonadotropins should be produced in eukaryotic cells, which have glycosylation capacity, but this post-translational modification varies greatly depending on the cell system, influencing hormone activity and stability. The production of recombinant gonadotropin beta-subunits to be used as antigens for antibody production has allowed the development of immunoassays for quantification of gonadotropins in some fish species. The administration in vivo of dimeric homologous recombinant gonadotropins has been used in basic studies and as a biotechnological approach to induce gametogenesis. In addition, gene-based therapies using somatic transfer of the gonadotropin genes have been tested as an alternative for hormone delivery in vivo. In summary, the use of homologous hormonal treatments can open new strategies in aquaculture to solve reproductive problems or develop out-of-season breeding programs.
Collapse
Affiliation(s)
- G Molés
- Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (CSIC), Ribera de Cabanes s/n, 12595 Castelló, Spain
| | - K Hausken
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - M Carrillo
- Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (CSIC), Ribera de Cabanes s/n, 12595 Castelló, Spain
| | - S Zanuy
- Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (CSIC), Ribera de Cabanes s/n, 12595 Castelló, Spain
| | - B Levavi-Sivan
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel.
| | - A Gómez
- Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas (CSIC), Ribera de Cabanes s/n, 12595 Castelló, Spain.
| |
Collapse
|
26
|
Li M, Liu X, Dai S, Xiao H, Qi S, Li Y, Zheng Q, Jie M, Cheng CHK, Wang D. Regulation of spermatogenesis and reproductive capacity by Igf3 in tilapia. Cell Mol Life Sci 2020; 77:4921-4938. [PMID: 31955242 PMCID: PMC11104970 DOI: 10.1007/s00018-019-03439-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 11/11/2019] [Accepted: 12/23/2019] [Indexed: 02/01/2023]
Abstract
A novel insulin-like growth factor (igf3), which is exclusively expressed in the gonads, has been widely identified in fish species. Recent studies have indicated that Igf3 regulates spermatogonia proliferation and differentiation in zebrafish; however, detailed information on the role of this Igf needs further in vivo investigation. Here, using Nile tilapia (Oreochromis niloticus) as an animal model, we report that igf3 is required for spermatogenesis and reproduction. Knockout of igf3 by CRISPR/Cas9 severely inhibited spermatogonial proliferation and differentiation at 90 days after hatching, the time critical for meiosis initiation, and resulted in less spermatocytes in the mutants. Although spermatogenesis continued to occur later, more spermatocytes and less spermatids were observed in the igf3-/- testes when compared with wild type of testes at adults, indicating that Igf3 regulates spermatocyte to spermatid transition. Importantly, a significantly increased occurrence of apoptosis in spermatids was observed after loss of Igf3. Therefore, igf3-/- males were subfertile with drastically reduced semen volume and sperm count. Conversely, the overexpression of Igf3 in XY tilapia enhanced spermatogenesis leading to more spermatids and sperm count. Transcriptomic analysis revealed that the absence of Igf3 resulted in dysregulation of many genes involved in cell cycle, meiosis and pluripotency regulators that are critical for spermatogenesis. In addition, in vitro gonadal culture with 17α-methyltetosterone (MT) and 11-ketotestosterone (11-KT) administration and in vivo knockout of cyp11c1 demonstrated that igf3 expression is regulated by androgens, suggesting that Igf3 acts downstream of androgens in fish spermatogenesis. Notably, the igf3 knockout did not affect body growth, indicating that this Igf specifically functions in reproduction. Taken together, our data provide genetic evidence for fish igf3 in the regulation of reproductive capacity by controlling spermatogenesis.
Collapse
Affiliation(s)
- Minghui Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Xingyong Liu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Shengfei Dai
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Hesheng Xiao
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Shuangshuang Qi
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Yibing Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Qiaoyuan Zheng
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Mimi Jie
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Christopher H K Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Deshou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
27
|
Xie X, Nóbrega R, Pšenička M. Spermatogonial Stem Cells in Fish: Characterization, Isolation, Enrichment, and Recent Advances of In Vitro Culture Systems. Biomolecules 2020; 10:E644. [PMID: 32331205 PMCID: PMC7226347 DOI: 10.3390/biom10040644] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Spermatogenesis is a continuous and dynamic developmental process, in which a single diploid spermatogonial stem cell (SSC) proliferates and differentiates to form a mature spermatozoon. Herein, we summarize the accumulated knowledge of SSCs and their distribution in the testes of teleosts. We also reviewed the primary endocrine and paracrine influence on spermatogonium self-renewal vs. differentiation in fish. To provide insight into techniques and research related to SSCs, we review available protocols and advances in enriching undifferentiated spermatogonia based on their unique physiochemical and biochemical properties, such as size, density, and differential expression of specific surface markers. We summarize in vitro germ cell culture conditions developed to maintain proliferation and survival of spermatogonia in selected fish species. In traditional culture systems, sera and feeder cells were considered to be essential for SSC self-renewal, in contrast to recently developed systems with well-defined media and growth factors to induce either SSC self-renewal or differentiation in long-term cultures. The establishment of a germ cell culture contributes to efficient SSC propagation in rare, endangered, or commercially cultured fish species for use in biotechnological manipulation, such as cryopreservation and transplantation. Finally, we discuss organ culture and three-dimensional models for in vitro investigation of fish spermatogenesis.
Collapse
Affiliation(s)
- Xuan Xie
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, University of South Bohemia in Ceske Budejovice, Zátiší 728/II, 389 25 Vodňany, Czech Republic;
| | - Rafael Nóbrega
- Reproductive and Molecular Biology Group, Department of Morphology, Institute of Biosciences, São Paulo State University, Botucatu, SP 18618-970, Brazil;
| | - Martin Pšenička
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, University of South Bohemia in Ceske Budejovice, Zátiší 728/II, 389 25 Vodňany, Czech Republic;
| |
Collapse
|
28
|
Cortisol Directly Stimulates Spermatogonial Differentiation, Meiosis, and Spermiogenesis in Zebrafish ( Danio rerio) Testicular Explants. Biomolecules 2020; 10:biom10030429. [PMID: 32164184 PMCID: PMC7175196 DOI: 10.3390/biom10030429] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 12/21/2022] Open
Abstract
Cortisol is the major endocrine factor mediating the inhibitory effects of stress on vertebrate reproduction. It is well known that cortisol affects reproduction by interacting with the hypothalamic–pituitary–gonads axis, leading to downstream inhibitory and stimulatory effects on gonads. However, the mechanisms are not fully understood. In this study, we provide novel data demonstrating the stimulatory effects of cortisol on spermatogenesis using an ex vivo organ culture system. The results revealed that cortisol treatment did not modulate basal androgen production, but it influenced transcript levels of a selected number of genes involved in the zebrafish testicular function ar (androgen receptor), star (steroidogenic acute regulatory), cyp17a1 (17α-hydroxylase/17,20 lyase/17,20 desmolase), cyp11a2 (cytochrome P450, family 11, subfamily A, polypeptide 2), hsd11b2 (11-beta hydroxysteroid dehydrogenase), cyp2k22 (cytochrome P450, family 2, subfamily K, polypeptide 22), fkbp5 (FKBP prolyl isomerase 5), grα (glucocorticoid receptor alpha), and grβ (glucocorticoid receptor beta) in a short-term culture. We also showed that cortisol stimulates spermatogonial proliferation and differentiation in an androgen independent manner as well as promoting meiosis and spermiogenesis by increasing the number of spermatozoa in the testes. Moreover, we demonstrated that concomitant treatment with RU 486, a potent glucocorticoid receptor (Gr) antagonist, did not affect the cortisol effects on spermatogonial differentiation but blocked the induced effects on meiosis and spermiogenesis. Supporting the Gr-mediated effects, RU 486 nullified the cortisol-induced expression of sycp3l (synaptonemal complex protein 3), a marker for the meiotic prophase that encodes a component of the synaptonemal complex. This is consistent with in silico analysis that found 10 putative GREs (glucocorticoid response elements) upstream of the zebrafish sycp3l. Finally, we also showed that grα mRNA is expressed in Sertoli and Leydig cells, but also in several types of germ cells, including spermatogonia and spermatocytes. Altogether, this evidence indicates that cortisol exerts paracrine roles in the zebrafish testicular function and spermatogenesis, highlighting its effects on spermatogonial differentiation, meiosis, and spermiogenesis.
Collapse
|
29
|
Fallah HP, Rodrigues MS, Corchuelo S, Nóbrega RH, Habibi HR. Role of GnRH Isoforms in Paracrine/Autocrine Control of Zebrafish (Danio rerio) Spermatogenesis. Endocrinology 2020; 161:5701481. [PMID: 31930304 DOI: 10.1210/endocr/bqaa004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/10/2020] [Indexed: 12/25/2022]
Abstract
Abstract
It is well established that hypothalamic GnRH (gonadotropin-releasing hormone) is one of the key peptides involved in the neuroendocrine control of testicular development and spermatogenesis. However, the role of GnRH as a paracrine regulator of testicular function has not been fully investigated. The present study demonstrates the presence of GnRH and its receptors in the zebrafish (Danio rerio) testis, and provides information on direct action of native GnRH isoforms (GnRH2 and GnRH3) on different stages of spermatogenesis in this model. Both GnRH2 and GnRH3 stimulated basal spermatogenesis by increasing numbers of type Aund spermatogonia, spermatozoa, and testosterone release, and in this study GnRH2 exerted higher relative activity than GnRH3. Next, we evaluated the effects of GnRH isoforms on human chorionic gonadotropin (hCG)- and follicle-stimulating hormone (Fsh)-induced spermatogenesis. The 2 GnRH isoforms were found to have different effects on Fsh- and hCG-induced response depending on the stage of spermatogenesis and concentration of the peptides. The results provide strong support for the hypothesis that locally produced GnRH2 and GnRH3 are important components of the complex multifactorial system that regulates testicular germinal cell development and function in adult zebrafish.
Collapse
Affiliation(s)
- Hamideh P Fallah
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Maira S Rodrigues
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Department of Morphology, Reproductive and Molecular Biology Group, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Sheryll Corchuelo
- Department of Morphology, Reproductive and Molecular Biology Group, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Rafael H Nóbrega
- Department of Morphology, Reproductive and Molecular Biology Group, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Hamid R Habibi
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
30
|
Thönnes M, Vogt M, Steinborn K, Hausken KN, Levavi-Sivan B, Froschauer A, Pfennig F. An ex vivo Approach to Study Hormonal Control of Spermatogenesis in the Teleost Oreochromis niloticus. Front Endocrinol (Lausanne) 2020; 11:443. [PMID: 32793114 PMCID: PMC7366826 DOI: 10.3389/fendo.2020.00443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 06/05/2020] [Indexed: 11/13/2022] Open
Abstract
As the male reproductive organ, the main task of the testis is the production of fertile, haploid spermatozoa. This process, named spermatogenesis, starts with spermatogonial stem cells, which undergo a species-specific number of mitotic divisions until starting meiosis and further morphological maturation. The pituitary gonadotropins, luteinizing hormone, and follicle stimulating hormone, are indispensable for vertebrate spermatogenesis, but we are still far from fully understanding the complex regulatory networks involved in this process. Therefore, we developed an ex vivo testis cultivation system which allows evaluating the occurring changes in histology and gene expression. The experimental circulatory flow-through setup described in this work provides the possibility to study the function of the male tilapia gonads on a cellular and transcriptional level for at least 7 days. After 1 week of culture, tilapia testis slices kept their structure and all stages of spermatogenesis could be detected histologically. Without pituitary extract (tilPE) however, fibrotic structures appeared, whereas addition of tilPE preserved spermatogenic cysts and somatic interstitium completely. We could show that tilPE has a stimulatory effect on spermatogonia proliferation in our culture system. In the presence of tilPE or hCG, the gene expression of steroidogenesis related genes (cyp11b2 and stAR2) were notably increased. Other testicular genes like piwil1, amh, or dmrt1 were not expressed differentially in the presence or absence of gonadotropins or gonadotropin containing tilPE. We established a suitable system for studying tilapia spermatogenesis ex vivo with promise for future applications.
Collapse
Affiliation(s)
- Michelle Thönnes
- Faculty of Biology, School of Science, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Marlen Vogt
- Faculty of Biology, School of Science, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Katja Steinborn
- Faculty of Biology, School of Science, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Krist N. Hausken
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Berta Levavi-Sivan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Alexander Froschauer
- Faculty of Biology, School of Science, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
| | - Frank Pfennig
- Faculty of Biology, School of Science, Institute of Zoology, Technische Universität Dresden, Dresden, Germany
- *Correspondence: Frank Pfennig
| |
Collapse
|
31
|
Suzuki H, Kazeto Y, Gen K, Ozaki Y. Functional analysis of recombinant single-chain Japanese eel Fsh and Lh produced in FreeStyle 293-F cell lines: Binding specificities to their receptors and differential efficacy on testicular steroidogenesis. Gen Comp Endocrinol 2020; 285:113241. [PMID: 31400434 DOI: 10.1016/j.ygcen.2019.113241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/11/2019] [Accepted: 08/06/2019] [Indexed: 01/02/2023]
Abstract
Pituitary gonadotropins, follicle-stimulating hormone (Fsh) and luteinizing hormone (Lh), play central roles in the control of gonadal development of vertebrates. In mammals, Fsh and Lh exclusively activate their respective cognate receptors: Fsh receptor (Fshr) in the Sertoli cell and Lh/choriogonadotropin receptor (Lhcgr) in the Leydig cell. In teleosts, the distinct functions of Fsh and Lh and information on cellular localization of their receptors are still poorly understood. Recently we established FreeStyle 293-F cell lines producing recombinant Japanese eel Fsh and Lh (reFsh and reLh), which form a single chain consisting of a common α-subunit and β-subunits. In this study, we conducted functional analyses of reFsh and reLh, focusing on the binding specificities to their receptors and effects on testicular steroidogenesis in vitro. Assays with gonadotropin receptors-expressing COS-7 cells indicated reFsh stimulated its cognate receptor, meanwhile reLh activated both receptors. Although results of in vitro incubations showed that reFsh and reLh induced testicular 11-ketotestosterone production in a dose and time-dependent manner by upregulating expression of steroidogenic enzymes, the effective doses of reLh were apparently lower and the effects of reLh emerged faster in comparison with reFsh. Results of quantitative real-time PCR using testicular cell fractions showed that fshr and lhcgr1 mRNA were detected both in Sertoli and Leydig cells. These analyses revealed that reFsh and reLh were biologically active and hence will be useful for future studies. Moreover, our data showed that both eel Fsh and Lh acted as steroidogenic hormones through their receptors in testicular somatic cells; however, Lh was more potent on androgen production, implying differential functions on spermatogenesis.
Collapse
Affiliation(s)
- Hiroshi Suzuki
- Graduate School of Marine Science and Technology, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato-ku, Tokyo 108-8477, Japan; National Research Institute of Aquaculture, Japan Fisheries Research and Education Agency, 224-1 Hiruda, Tamaki, Watarai, Mie 519-0423, Japan.
| | - Yukinori Kazeto
- National Research Institute of Aquaculture, Japan Fisheries Research and Education Agency, Tsuiura, Kamiura, Saiki, Oita 879-2602, Japan.
| | - Koichiro Gen
- Seikai National Fisheries Research Institute, Japan Fisheries Research and Education Agency, 1551-8 Taira, Nagasaki 851-2213, Japan.
| | - Yuichi Ozaki
- National Research Institute of Aquaculture, Japan Fisheries Research and Education Agency, 224-1 Hiruda, Tamaki, Watarai, Mie 519-0423, Japan.
| |
Collapse
|
32
|
Safian D, Bogerd J, Schulz RW. Regulation of spermatogonial development by Fsh: The complementary roles of locally produced Igf and Wnt signaling molecules in adult zebrafish testis. Gen Comp Endocrinol 2019; 284:113244. [PMID: 31415728 DOI: 10.1016/j.ygcen.2019.113244] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/28/2022]
Abstract
Spermatogenesis is a cellular developmental process characterized by the coordinated proliferation and differentiation activities of somatic and germ cells in order to produce a large number of spermatozoa, the cellular basis of male fertility. Somatic cells in the testis, such as Leydig, peritubular myoid and Sertoli cells, provide structural and metabolic support and contribute to the regulatory microenvironment required for proper germ cell survival and development. The pituitary follicle-stimulating hormone (Fsh) is a major endocrine regulator of vertebrate spermatogenesis, targeting somatic cell functions in the testes. In fish, Fsh regulates Leydig and Sertoli cell functions, such as sex steroid and growth factor production, processes that also control the development of spermatogonia, the germ cell stages at the basis of the spermatogenic process. Here, we summarize recent advances in our understanding of mechanisms used by Fsh to regulate the development of spermatogonia. This involves discussing the roles of insulin-like growth factor (Igf) 3 and canonical and non-canonical Wnt signaling pathways. We will also discuss how these locally active regulatory systems interact to maintain testis tissue homeostasis.
Collapse
Affiliation(s)
- Diego Safian
- Reproductive Biology Group, Division Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, University of Utrecht, 3584 CH Utrecht, The Netherlands
| | - Jan Bogerd
- Reproductive Biology Group, Division Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, University of Utrecht, 3584 CH Utrecht, The Netherlands
| | - Rüdiger W Schulz
- Reproductive Biology Group, Division Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, University of Utrecht, 3584 CH Utrecht, The Netherlands; Reproduction and Developmental Biology Group, Institute of Marine Research, P.O. Box 1870, Nordnes, 5817 Bergen, Norway.
| |
Collapse
|
33
|
Crespo D, Assis LHC, van de Kant HJG, de Waard S, Safian D, Lemos MS, Bogerd J, Schulz RW. Endocrine and local signaling interact to regulate spermatogenesis in zebrafish: follicle-stimulating hormone, retinoic acid and androgens. Development 2019; 146:dev.178665. [PMID: 31597660 DOI: 10.1242/dev.178665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 10/01/2019] [Indexed: 01/07/2023]
Abstract
Retinoic acid (RA) is crucial for mammalian spermatogonia differentiation, and stimulates Stra8 expression, a gene required for meiosis. Certain fish species, including zebrafish, have lost the stra8 gene. While RA still seems important for spermatogenesis in fish, it is not known which stage(s) respond to RA or whether its effects are integrated into the endocrine regulation of spermatogenesis. In zebrafish, RA promoted spermatogonia differentiation, supported androgen-stimulated meiosis, and reduced spermatocyte and spermatid apoptosis. Follicle-stimulating hormone (Fsh) stimulated RA production. Expressing a dominant-negative RA receptor variant in germ cells clearly disturbed spermatogenesis but meiosis and spermiogenesis still took place, although sperm quality was low in 6-month-old adults. This condition also activated Leydig cells. Three months later, spermatogenesis apparently had recovered, but doubling of testis weight demonstrated hypertrophy, apoptosis/DNA damage among spermatids was high and sperm quality remained low. We conclude that RA signaling is important for zebrafish spermatogenesis but is not of crucial relevance. As Fsh stimulates androgen and RA production, germ cell-mediated, RA-dependent reduction of Leydig cell activity may form a hitherto unknown intratesticular negative-feedback loop.
Collapse
Affiliation(s)
- Diego Crespo
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Luiz H C Assis
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Henk J G van de Kant
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Sjors de Waard
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Diego Safian
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Moline S Lemos
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Jan Bogerd
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Rüdiger W Schulz
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht 3584 CH, The Netherlands .,Research Group Reproduction and Developmental Biology, Institute of Marine Research, Bergen NO-5817, Norway
| |
Collapse
|
34
|
Li J, Cheng CHK. Evolution of gonadotropin signaling on gonad development: insights from gene knockout studies in zebrafish. Biol Reprod 2019; 99:686-694. [PMID: 29718109 DOI: 10.1093/biolre/ioy101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/25/2018] [Indexed: 11/13/2022] Open
Abstract
Gonadal development is precisely regulated by the two gonadotropins luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Much progress on understanding the functions of LH and FSH signaling on gonad development has been achieved in the past decades, mostly from studies in mammals, especially genetic studies in both mouse and human. The functions of both LH and FSH signaling in nonmammalian species are still largely unknown. In recent years, using zebrafish, a teleost phylogenetically distant from mammals, we and others have genetically analyzed the functions of gonadotropins and their receptors through gene knockout studies. In this review, we will summarize the pertinent findings and discuss how the actions of gonadotropin signaling on gonad development have evolved during evolution from fish to mammals.
Collapse
Affiliation(s)
- Jianzhen Li
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| | - Christopher H K Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| |
Collapse
|
35
|
Ozaki Y, Damsteegt EL, Setiawan AN, Miura T, Lokman PM. Expressional regulation of gonadotropin receptor genes and androgen receptor genes in the eel testis. Gen Comp Endocrinol 2019; 280:123-133. [PMID: 31009604 DOI: 10.1016/j.ygcen.2019.04.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 11/17/2022]
Abstract
Receptors for follicle-stimulating hormone (Fshr), luteinizing hormone (Lhcgr1 and Lhcgr2) and androgens (Ara and Arb) transduce the hormonal signals that coordinate spermatogenesis, but the factors that regulate the abundance of these transducers in fish testes remain little-understood. To mend this paucity of information, we first determined changes in transcript abundance for these receptors (fshr, lhcgr1, ara and arb) during spermatogenesis induced by human chorionic gonadotropin (hCG) injection in the eel, Anguilla australis. We related our findings to testicular production of the fish androgen, 11-ketotestosterone (11-KT), and to the levels of the transcripts encoding steroidogenic acute regulatory protein (star) and 11β-hydroxylase (cyp11b), and subsequently evaluated the effects of hCG or 11-KT on mRNA levels of these target genes in vitro. Testicular 11-KT production was greatly increased by hCG treatment, both in vivo and in vitro, and associated with up-regulation of star and cyp11b transcripts. In situ hybridization indicated that testicular fshr mRNA levels were higher in the early stages of hCG-induced spermatogenesis, while lhcgr1 transcripts were most abundant later, once spermatids were observed. In vitro experiments further showed that hCG and its steroidal mediator 11-KT significantly increased fshr transcript abundance. These data provide new angles on the interactions between gonadotropin and androgen signaling during early spermatogenesis. Increases in levels of 11-KT following hCG injection elevated testicular fshr mRNA levels augmenting Fsh sensitivity in the testis. This evidence is suggestive of a positive feedback loop between gonadotropins and 11-KT that may be key to regulating early spermatogenesis in fish.
Collapse
MESH Headings
- Androgens/metabolism
- Anguilla/blood
- Anguilla/genetics
- Animals
- Chorionic Gonadotropin/administration & dosage
- Chorionic Gonadotropin/pharmacology
- Gene Expression Regulation/drug effects
- Humans
- Male
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, FSH/genetics
- Receptors, FSH/metabolism
- Receptors, Gonadotropin/genetics
- Receptors, Gonadotropin/metabolism
- Receptors, LH/genetics
- Receptors, LH/metabolism
- Spermatogenesis/drug effects
- Spermatogenesis/genetics
- Steroid 11-beta-Hydroxylase/genetics
- Steroid 11-beta-Hydroxylase/metabolism
- Testis/drug effects
- Testis/metabolism
- Testosterone/analogs & derivatives
- Testosterone/blood
Collapse
Affiliation(s)
- Yuichi Ozaki
- Department of Zoology, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand
| | - Erin L Damsteegt
- Department of Zoology, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand.
| | - Alvin N Setiawan
- Department of Zoology, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand
| | - Takeshi Miura
- Graduate School of Agriculture, Ehime University, 3-5-7 Tarumi, Matsuyama, Ehime 790 8566, Japan
| | - P Mark Lokman
- Department of Zoology, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand
| |
Collapse
|
36
|
Li X, Zhang B, Li N, Ji X, Liu K, Jin M. Zebrafish neurobehavioral phenomics applied as the behavioral warning methods for fingerprinting endocrine disrupting effect by lead exposure at environmentally relevant level. CHEMOSPHERE 2019; 231:315-325. [PMID: 31132538 DOI: 10.1016/j.chemosphere.2019.05.146] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 06/09/2023]
Abstract
Environmental lead (Pb) exposure is a great hazard to the public health. Although environmentally relevant Pb poisoning is preventable, insidious Pb contaminants are still a major threat to human health. Herein, we reported that exposure to Pb at environmentally relevant concentration level (1 μg/L, 10 μg/L and 100 μg/L), disturbed the courtship behavior of adult male zebrafish and further altered the transcriptional patterns of key genes involved in testicular steroidogenesis (igf3, amh, piwil1, lhcgr, fshr, cyp11c1, star, cyp19a1a, cyp19a1b) and apoptosis (bax, cytoC, caspase 9, caspase 3, puma). Both the behavioral and the transcriptional profiles share a similar biphasic dose response, with stimulatory effects after low-level exposure and inhibitory effects after high-level exposure. This results revealed the endocrine disrupting effects of Pb even at an environmentally relevant level within the concentration range of ambient water quality criteria (AWQC) and the reliability of locomotion fingerprint as the indicator for detecting the risk induced by Pb pollution. Current research, for the first time, employed the ZebraTower system as the biological early warning system (BEWS) to find that Pb exerted biphasic effects on the courtship behavior and endocrine regulation of male adult zebrafish. Methodologically, we firstly propose an efficient solution to monitor and assess the risk of Pb exposure by combining the (BEWS) and data analyzing methods such as zebrafish phenomics, which would make a contribution to the detection and prevention of environmentally relevant Pb poisoning.
Collapse
Affiliation(s)
- Xiang Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No.44 West Culture Road, Ji'nan 250012, Shandong Province, PR China
| | - Baoyue Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Ning Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Xiuna Ji
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China.
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China.
| |
Collapse
|
37
|
Schulz RW, Taranger GL, Bogerd J, Nijenhuis W, Norberg B, Male R, Andersson E. Entry into puberty is reflected in changes in hormone production but not in testicular receptor expression in Atlantic salmon (Salmo salar). Reprod Biol Endocrinol 2019; 17:48. [PMID: 31226998 PMCID: PMC6588918 DOI: 10.1186/s12958-019-0493-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/14/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Puberty in male Atlantic salmon in aquaculture can start as early as after the first winter in seawater, stunts growth and entails welfare problems due to the maturation-associated loss of osmoregulation capacity in seawater. A better understanding of the regulation of puberty is the basis for developing improved cultivation approaches that avoid these problems. Our aim here was to identify morphological and molecular markers signaling the initiation of, and potential involvement in, testis maturation. METHODS In the first experiment, we monitored for the first time in large Atlantic salmon males several reproductive parameters during 17 months including the first reproductive cycle. Since testicular growth accelerated after the Winter solstice, we focused in the second experiment on the 5 months following the winter solstice, exposing fish from February 1 onwards to the natural photoperiod (NL) or to continuous additional light (LL). RESULTS In the first experiment, testis weight, plasma androgens and pituitary gonadotropin transcript levels increased with the appearance of type B spermatogonia in the testis, but testicular transcript levels for gonadotropin or androgen receptors did not change while being clearly detectable. In the second experiment, all males kept under NL had been recruited into puberty until June. However, recruitment into puberty was blocked in ~ 40% of the males exposed to LL. The first morphological sign of recruitment was an increased proliferation activity of single spermatogonia and Sertoli cells. Irrespective of the photoperiod, this early sign of testis maturation was accompanied by elevated pituitary gnrhr4 and fshb and testicular igf3 transcript levels as well as increased plasma androgen levels. The transition into puberty occurred again with stable testicular gonadotropin and androgen receptor transcript levels. CONCLUSIONS The sensitivity to reproductive hormones is already established before puberty starts and up-regulation of testicular hormone receptor expression is not required to facilitate entry into puberty. The increased availability of receptor ligands, on the other hand, may result from an up-regulation of pituitary Gnrh receptor expression, eventually activating testicular growth factor and sex steroid release and driving germ and Sertoli cell proliferation and differentiation.
Collapse
Affiliation(s)
- Rüdiger W Schulz
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, P.O.Box 1870 Nordnes, 5817, Bergen, Norway
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Geir Lasse Taranger
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, P.O.Box 1870 Nordnes, 5817, Bergen, Norway
| | - Jan Bogerd
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Wouter Nijenhuis
- Reproductive Biology Group, Division Developmental Biology, Department Biology, Science Faculty, Utrecht University, Utrecht, The Netherlands
| | - Birgitta Norberg
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, P.O.Box 1870 Nordnes, 5817, Bergen, Norway
| | - Rune Male
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Eva Andersson
- Research Group Reproduction and Developmental Biology, Institute of Marine Research, P.O.Box 1870 Nordnes, 5817, Bergen, Norway.
| |
Collapse
|
38
|
The initiation of puberty in Atlantic salmon brings about large changes in testicular gene expression that are modulated by the energy status. BMC Genomics 2019; 20:475. [PMID: 31185904 PMCID: PMC6558769 DOI: 10.1186/s12864-019-5869-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 05/31/2019] [Indexed: 02/08/2023] Open
Abstract
Background When puberty starts before males reach harvest size, animal welfare and sustainability issues occur in Atlantic salmon (Salmo salar) aquaculture. Hallmarks of male puberty are an increased proliferation activity in the testis and elevated androgen production. Examining transcriptional changes in salmon testis during the transition from immature to maturing testes may help understanding the regulation of puberty, potentially leading to procedures to modulate its start. Since differences in body weight influence, via unknown mechanisms, the chances for entering puberty, we used two feed rations to create body weight differences. Results Maturing testes were characterized by an elevated proliferation activity of Sertoli cells and of single undifferentiated spermatogonia. Pituitary gene expression data suggest increased Gnrh receptor and gonadotropin gene expression, potentially responsible for the elevated circulating androgen levels in maturing fish. Transcriptional changes in maturing testes included a broad variety of signaling systems (e.g. Tgfβ, Wnt, insulin/Igf, nuclear receptors), but also, activation of metabolic pathways such as anaerobic metabolism and protection against ROS. Feed restriction lowered the incidence of puberty. In males maturing despite feed restriction, plasma androgen levels were higher than in maturing fish receiving the full ration. A group of 449 genes that were up-regulated in maturing fully fed fish, was up-regulated more prominently in testis from fish maturing under caloric restriction. Moreover, 421 genes were specifically up-regulated in testes from fish maturing under caloric restriction, including carbon metabolism genes, a pathway relevant for nucleotide biosynthesis and for placing epigenetic marks. Conclusions Undifferentiated spermatogonia and Sertoli cell populations increased at the beginning of puberty, which was associated with the up-regulation of metabolic pathways (e.g. anaerobic and ROS pathways) known from other stem cell systems. The higher androgen levels in males maturing under caloric restriction may be responsible for the stronger up-regulation of a common set of (449) maturation-associated genes, and the specific up-regulation of another set of (421) genes. The latter opened regulatory and/or metabolic options for initiating puberty despite feed restriction. As a means to reduce the incidence of male puberty in salmon, however, caloric restriction seems unsuitable. Electronic supplementary material The online version of this article (10.1186/s12864-019-5869-9) contains supplementary material, which is available to authorized users.
Collapse
|
39
|
Roles of piwil1 gene in gonad development and gametogenesis in Japanese flounder, Paralichthys olivaceus. Gene 2019; 701:104-112. [PMID: 30905810 DOI: 10.1016/j.gene.2019.03.045] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/05/2019] [Accepted: 03/20/2019] [Indexed: 11/23/2022]
Abstract
PIWI family member piwil1, which associates with Piwi-interacting RNA (piRNA), is responsible in regulation of germ cell differentiation and maintenance of reproductive stem cells. In this study, we analyzed the piwil1 gene in Paralichthys olivaceus. Bioinformatics analysis and structure prediction showed that piwil1 had the conserved domains: PAZ domain and PIWI domain. Expression analysis during embryonic development implied that piwil1 gene was maternally inherited. The tissue distribution showed a sexually dimorphic gene expression pattern, with higher expression level in testis than ovary. In situ hybridization results demonstrated that piwil1 was predominantly distributed in oogonia, oocytes, sertoli cells and spermatocytes. A CpG island was predicted in the 5'-flanking region of piwil1 gene, and its methylation levels showed significant disparity between males and females, indicating that the sexually dimorphic expression of piwil1 gene might be regulated by methylation. Furthermore, we explored the distinct roles of human chorionic gonadotropin and 17α-methyltestosterone in regulating the expression of piwil1, and found that piwil1 was interacting with the HPG axis hormones. These results indicated that piwil1 might play a crucial role in gonadal development and gametogenesis in Paralichthys olivaceus.
Collapse
|
40
|
Safian D, Ryane N, Bogerd J, Schulz RW. Fsh stimulates Leydig cell Wnt5a production, enriching zebrafish type A spermatogonia. J Endocrinol 2018; 239:351-363. [PMID: 30400013 DOI: 10.1530/joe-18-0447] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 09/14/2018] [Indexed: 12/13/2022]
Abstract
Follicle-stimulating hormone (Fsh) modulates vertebrate spermatogenesis by regulating somatic cell functions in the testis. We have found previously that zebrafish Fsh stimulated the differentiating proliferation of type A undifferentiated spermatogonia (Aund) in an androgen-independent manner by regulating the production of growth factors and other signaling molecules in both Sertoli (SCs) and Leydig cells (LCs). For example, Fsh triggered the release of Igf3 that subsequently activated β-catenin signaling to promote the differentiating proliferation of Aund. In the present study, we report that Fsh moreover uses the non-canonical Wnt pathway to promote the proliferation and accumulation of Aund. Initially, we found that the stimulatory effect of Fsh on the proliferation activity of Aund was further strengthened when β-catenin signaling was inhibited, resulting in an accumulation of Aund. We then showed that this Fsh-induced accumulation of Aund was associated with increased transcript levels of the non-canonical Wnt ligand, wnt5a. In situ hybridization of insl3 mRNA, a gene expressed in LCs, combined with Wnt5a immunocytochemistry identified LCs as the cellular source of Wnt5a in the adult zebrafish testis. Addition of an antagonist of Wnt5a to incubations with Fsh decreased both the proliferation activity and the relative section area occupied by Aund, while an agonist of Wnt5a increased these same parameters for Aund. Taken together, our data suggest that Fsh triggered LCs to release Wnt5a, which then promoted the proliferation and accumulation of Aund. Hence, Fsh uses non-canonical Wnt signaling to ensure the production of Aund, while also triggering β-catenin signaling via Igf3 to ensure spermatogonial differentiation.
Collapse
Affiliation(s)
- Diego Safian
- Reproductive Biology Group, Division Developmental Biology, Department of Biology, Institute of Biodynamics and Biocomplexity, Faculty of Science, University of Utrecht, NL-3584 CH Utrecht, The Netherlands
| | - Najoua Ryane
- Reproductive Biology Group, Division Developmental Biology, Department of Biology, Institute of Biodynamics and Biocomplexity, Faculty of Science, University of Utrecht, NL-3584 CH Utrecht, The Netherlands
| | - Jan Bogerd
- Reproductive Biology Group, Division Developmental Biology, Department of Biology, Institute of Biodynamics and Biocomplexity, Faculty of Science, University of Utrecht, NL-3584 CH Utrecht, The Netherlands
| | - Rüdiger W Schulz
- Reproductive Biology Group, Division Developmental Biology, Department of Biology, Institute of Biodynamics and Biocomplexity, Faculty of Science, University of Utrecht, NL-3584 CH Utrecht, The Netherlands
- Reproduction and Developmental Biology Group, Institute of Marine Research, Nordnes, Bergen, Norway
| |
Collapse
|
41
|
Casarini L, Santi D, Brigante G, Simoni M. Two Hormones for One Receptor: Evolution, Biochemistry, Actions, and Pathophysiology of LH and hCG. Endocr Rev 2018; 39:549-592. [PMID: 29905829 DOI: 10.1210/er.2018-00065] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/08/2018] [Indexed: 01/03/2023]
Abstract
LH and chorionic gonadotropin (CG) are glycoproteins fundamental to sexual development and reproduction. Because they act on the same receptor (LHCGR), the general consensus has been that LH and human CG (hCG) are equivalent. However, separate evolution of LHβ and hCGβ subunits occurred in primates, resulting in two molecules sharing ~85% identity and regulating different physiological events. Pituitary, pulsatile LH production results in an ~90-minute half-life molecule targeting the gonads to regulate gametogenesis and androgen synthesis. Trophoblast hCG, the "pregnancy hormone," exists in several isoforms and glycosylation variants with long half-lives (hours) and angiogenic potential and acts on luteinized ovarian cells as progestational. The different molecular features of LH and hCG lead to hormone-specific LHCGR binding and intracellular signaling cascades. In ovarian cells, LH action is preferentially exerted through kinases, phosphorylated extracellular-regulated kinase 1/2 (pERK1/2) and phosphorylated AKT (also known as protein kinase B), resulting in irreplaceable proliferative/antiapoptotic signals and partial agonism on progesterone production in vitro. In contrast, hCG displays notable cAMP/protein kinase A (PKA)-mediated steroidogenic and proapoptotic potential, which is masked by estrogen action in vivo. In vitro data have been confirmed by a large data set from assisted reproduction, because the steroidogenic potential of hCG positively affects the number of retrieved oocytes, and LH affects the pregnancy rate (per oocyte number). Leydig cell in vitro exposure to hCG results in qualitatively similar cAMP/PKA and pERK1/2 activation compared with LH and testosterone. The supposed equivalence of LH and hCG has been disproved by such data, highlighting their sex-specific functions and thus deeming it an oversight caused by incomplete understanding of clinical data.
Collapse
Affiliation(s)
- Livio Casarini
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniele Santi
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Giulia Brigante
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Manuela Simoni
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Modena, Italy
| |
Collapse
|
42
|
Safian D, Bogerd J, Schulz RW. Igf3 activates β-catenin signaling to stimulate spermatogonial differentiation in zebrafish. J Endocrinol 2018; 238:245-257. [PMID: 29941503 DOI: 10.1530/joe-18-0124] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023]
Abstract
Follicle-stimulating hormone (Fsh) is a major regulator of spermatogenesis, targeting somatic cell functions in the testes. We reported previously that zebrafish Fsh promoted the differentiation of type A undifferentiated spermatogonia (Aund) by stimulating the production of factors that advance germ cell differentiation, such as androgens, insulin-like peptide 3 (Insl3) and insulin-like growth factor 3 (Igf3). In addition, Fsh also modulated the transcript levels of several other genes, including some belonging to the Wnt signaling pathway. Here, we evaluated if and how Fsh utilizes part of the canonical Wnt pathway to regulate the development of spermatogonia. We quantified the proliferation activity and relative section areas occupied by Aund and type A differentiating (Adiff) spermatogonia and we analyzed the expression of selected genes in response to recombinant proteins and pharmacological inhibitors. We found that from the three downstream mediators of Fsh activity we examined, Igf3, but not 11-ketotestosterone or Insl3, modulated the transcript levels of two β-catenin sensitive genes (cyclinD1 and axin2). Using a zebrafish β-catenin signaling reporter line, we showed that Igf3 activated β-catenin signaling in type A spermatogonia and that this activation did not depend on the release of Wnt ligands. Pharmacological inhibition of the β-catenin or of the phosphoinositide 3-kinase (PI3K) pathways revealed that Igf3 activated β-catenin signaling in a manner involving PI3K to promote the differentiation of Aund to Adiff spermatogonia. This mechanism represents an intriguing example for a pituitary hormone like Fsh using Igf signaling to recruit the evolutionary conserved, local β-catenin signaling pathway to regulate spermatogenesis.
Collapse
Affiliation(s)
- Diego Safian
- Reproductive Biology GroupDivision Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology , Faculty of Science, University of Utrecht, Utrecht, The Netherlands
| | - Jan Bogerd
- Reproductive Biology GroupDivision Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology , Faculty of Science, University of Utrecht, Utrecht, The Netherlands
| | - Rüdiger W Schulz
- Reproductive Biology GroupDivision Developmental Biology, Institute of Biodynamics and Biocomplexity, Department of Biology , Faculty of Science, University of Utrecht, Utrecht, The Netherlands
- Reproduction and Developmental Biology GroupInstitute of Marine Research, Nordnes, Bergen, Norway
| |
Collapse
|
43
|
Basak R, Roy A, Rai U. In silico analysis, temporal expression and gonadotropic regulation of receptors for follicle-stimulating hormone and luteinizing hormone in testis of spotted snakehead Channa punctata. JOURNAL OF FISH BIOLOGY 2018; 93:53-71. [PMID: 29931764 DOI: 10.1111/jfb.13727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 04/19/2018] [Indexed: 06/08/2023]
Abstract
This study in spotted snakehead Channa punctata was aimed to develop a comprehensive understanding of testicular gonadotropin receptors, from their sequence characterization, temporal expression to gonadotropic regulation, in seasonally breeding teleosts. A single form of follicle-stimulating hormone receptor (cpfshra) and luteinizing hormone/choriogonadotropin receptor (cplhcgr), was identified from testicular transcriptome data of C. punctata. Although deduced full-length protein sequence for cpFshra (694 amino acids) and cpLhcgr (691 amino acids) showed homology with their counterparts of other vertebrates, multiple insertion-deletion-substitution of residues suggest marked alterations in their structure and ligand specificity. The absolute quantification of testicular cpfshra and cplhcgr was estimated along the reproductive cycle following real-time PCR. The temporal expression profile showed highest testicular expression of both the gonadotropin receptors during resting phase. Their expression progressively decreased during preparatory and spawning phases concomitant with spermatogonial proliferation and differentiation and spermiogenesis. However, levels of cpfshra and cplhcgr sharply increased during post-spawning when seminiferous lobules were largely devoid of germ cells. To explore gonadotropic regulation of testicular cpfshra and cplhcgr, one group of fish of resting phase was administered with single dose of human chorionic gonadotropin (hCG; 5,000 IU/kg body mass) on day 0 and sacrificed on day 3 and day 5, while another group receiving two injections of hCG (day 0 and day 7) was sacrificed on day 14. The expression pattern of testicular gonadotropin receptors in hCG-treated fish sacrificed after 3, 5 and 14 days was similar to that of preparatory, spawning and postspawning phases, respectively. Likewise, testicular histology of hCG-treated fish sacrificed on day 3, day 5 and day 14 was comparable with that of preparatory, early spawning and late spawning phases, respectively. In light of the fact that gonadotropin receptors are largely expressed on somatic cells, an apparent decrease in testicular cpfshra and cplhcgr levels during preparatory and spawning phases or after 3 and 5 days from first hCG injection might not be due to downregulation of their expression. Rather, this could be due to dilution of somatic cell mRNA by large amount of germ cell mRNA. To verify this assumption, effect of hCG on plasma level of androgens was investigated employing enzyme-linked immunosorbent assay. A marked increase in plasma level of testosterone and 11-ketotestosterone was observed after hCG treatment in C. punctata. This would have been possible only when hCG upregulated the expression of testicular gonadotropin receptors.
Collapse
Affiliation(s)
| | - Alivia Roy
- Department of Zoology, University of Delhi, Delhi, India
| | - Umesh Rai
- Department of Zoology, University of Delhi, Delhi, India
| |
Collapse
|
44
|
de Castro Assis LH, de Nóbrega RH, Gómez-González NE, Bogerd J, Schulz RW. Estrogen-induced inhibition of spermatogenesis in zebrafish is largely reversed by androgen. J Mol Endocrinol 2018; 60:273-284. [PMID: 29476039 DOI: 10.1530/jme-17-0177] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/20/2022]
Abstract
The hormonal regulation of spermatogenesis involves both gonadotropins and steroid hormones. Long-term in vivo exposure of adult zebrafish to estrogen impaired spermatogenesis associated with an androgen insufficiency, possibly induced by inhibiting gonadotropin release. Using this experimental model, we investigated if androgen treatment could enhance spermatogenesis, while maintaining the inhibition of gonadotropin release through continued estrogen exposure. Moreover, we also exposed animals to androgen alone, in order to examine androgen effects in the absence of estrogen-induced gonadotropin inhibition. Estrogen exposure depleted type B spermatogonia, meiotic and postmeiotic germ cells from the adult testis, but promoted the proliferation of type A undifferentiated spermatogonia, which accumulated in the testis. This change in germ cell composition was accompanied by reduced mRNA levels of those growth factors (e.g. insl3 and igf3) expressed by testicular somatic cells and known to stimulate spermatogonial differentiation in zebrafish. Additional androgen (11-ketoandrostenedione, which is converted to 11-ketotestosterone) treatment in vivo reversed most of the effects of estrogen exposure on spermatogenesis while insl3 and igf3 transcript levels remained suppressed. When androgen treatment was given alone, it promoted the production of haploid cells at the expense of spermatogonia, and increased transcript levels of some growth factor and hormone receptor genes, but not those of insl3 or igf3 We conclude that estrogen exposure efficiently inhibits spermatogenesis because it induces androgen insufficiency and suppresses gonadotropin-regulated growth factors known to stimulate germ cell differentiation. Moreover, our results suggest that androgens and the growth factors Insl3 and Igf3 stimulate spermatogenesis via independent pathways.
Collapse
Affiliation(s)
- Luiz Henrique de Castro Assis
- Reproductive Biology GroupDivision of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, The Netherlands
| | - Rafael Henrique de Nóbrega
- Reproductive and Molecular Biology GroupDepartment of Morphology, Institute of Bioscience of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Nuria Esther Gómez-González
- Department of Cell Biology and HistologyFaculty of Biology, University of Murcia, IMIB-Arrixaca, Murcia, Spain
| | - Jan Bogerd
- Reproductive Biology GroupDivision of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, The Netherlands
| | - Rüdiger Winfried Schulz
- Reproductive Biology GroupDivision of Developmental Biology, Department of Biology, Faculty of Science, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
45
|
Graziano M, Benito R, Planas JV, Palstra AP. Swimming exercise to control precocious maturation in male seabass (Dicentrarchus labrax). BMC DEVELOPMENTAL BIOLOGY 2018; 18:10. [PMID: 29649968 PMCID: PMC5897932 DOI: 10.1186/s12861-018-0170-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 04/08/2018] [Indexed: 01/19/2023]
Abstract
Background Male European seabass, already predominant (~ 70%) in cultured stocks, show a high incidence (20–30%) of precocious sexual maturation under current aquaculture practices, leading to important economic losses for the industry. In view of the known modulation of reproductive development by swimming exercise in other teleost species, we aimed at investigating the effects of sustained swimming on reproductive development in seabass males during the first year of life in order to determine if swimming could potentially reduce precocious sexual maturation. Methods Pre-pubertal seabass (3.91 ± 0.22 g of body weight (BW)) were subjected to a 10 week swimming regime at their optimal swimming speed (Uopt) in an oval-shaped Brett-type flume or kept at rest during this period. Using Blazka-type swim tunnels, Uopt was determined three times during the course of the experiment: 0.66 m s− 1 at 19 ± 1 g BW, 10.2 ± 0.2 cm of standard length (SL) (week 1); 0.69 m s− 1 at 38 ± 3 g BW, 12.7 ± 0.3 cm SL (week 5), and also 0.69 m s− 1 at 77 ± 7 g BW, 15.7 ± 0.5 cm SL (week 9). Every 2 weeks, size and gonadal weight were monitored in the exercised (N = 15) and non-exercised fish (N = 15). After 10 weeks, exercised and non-exercised males were sampled to determine plasma 11-ketotestosterone levels, testicular mRNA expression levels of genes involved in steroidogenesis and gametogenesis by qPCR, as well as the relative abundance of germ cells representing the different spermatogenic stages by histological examination. Results Our results indicate that sustained swimming exercise at Uopt delays testicular development in male European seabass as evidenced by decreased gonado-somatic index, slower progression of testicular development and by reduced mRNA expression levels of follicle stimulating hormone receptor (fshR), 3-beta-hydroxysteroid dehydrogenase (3βhsd), 11-beta hydroxysteroid dehydrogenase (11βhsd), estrogen receptor-beta (erβ2), anti-mullerian hormone (amh), structural maintenance of chromosomes protein 1B (smc1β), inhibin beta A (inhba) and gonado-somal derived factor 1 (gsdf1) in exercised males as compared with the non-exercised males. Conclusions Swimming exercise may represent a natural and non-invasive tool to reduce the incidence of sexually precocious males in seabass aquaculture.
Collapse
Affiliation(s)
- Marco Graziano
- Department of Physiology and Immunology, School of Biology, University of Barcelona, Diagonal 643, 08028, Barcelona, Spain.,Wageningen Marine Research, Wageningen University & Research, Korringaweg 5, 4401, NT, Yerseke, The Netherlands
| | - Raul Benito
- Department of Physiology and Immunology, School of Biology, University of Barcelona, Diagonal 643, 08028, Barcelona, Spain.,Wageningen Marine Research, Wageningen University & Research, Korringaweg 5, 4401, NT, Yerseke, The Netherlands
| | - Josep V Planas
- Department of Physiology and Immunology, School of Biology, University of Barcelona, Diagonal 643, 08028, Barcelona, Spain
| | - Arjan P Palstra
- Wageningen Marine Research, Wageningen University & Research, Korringaweg 5, 4401, NT, Yerseke, The Netherlands. .,Wageningen Livestock Research, Wageningen University & Research Animal Breeding and Genomics, PO Box 338, 6700, AH, Wageningen, The Netherlands.
| |
Collapse
|
46
|
Zebrafish androgen receptor is required for spermatogenesis and maintenance of ovarian function. Oncotarget 2018; 9:24320-24334. [PMID: 29849943 PMCID: PMC5966271 DOI: 10.18632/oncotarget.24407] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/24/2018] [Indexed: 11/25/2022] Open
Abstract
The androgen receptor (AR) is a nuclear receptor protein family member and inducible transcription factor that modulates androgen target gene expression. Studies using a mouse model confirmed the need for ar in reproductive development, particularly spermatogenesis. Here, we investigated the role of ar in zebrafish using CRISPR/Cas9 gene targeting technology. Targeted disruption of ar in zebrafish increases the number of female offspring and increases offspring weight. In addition, ar-null male zebrafish have female secondary sex characteristics. More importantly, targeted disruption of ar in zebrafish causes male infertility via defective spermatogenesis and female premature ovarian failure during growth. Mechanistic assays suggest that these effects are caused by fewer proliferated cells and more apoptotic cells in ar-null testes. Moreover, genes involved in reproductive development, estradiol induction and hormone synthesis were dys-regulated in testes and ovaries and the reproductive-endocrine axis was disordered. Our data thus suggest that the zebrafish ar is required for spermatogenesis and maintenance of ovarian function, which confirms evolutionarily conserved functions of ar in vertebrates, as well as indicates that ar-null zebrafish are a suitable model for studying pathologic mechanisms related to androgen disorders.
Collapse
|
47
|
Using specific recombinant gonadotropins to induce spermatogenesis and spermiation in the European eel (Anguilla anguilla). Theriogenology 2018; 107:6-20. [DOI: 10.1016/j.theriogenology.2017.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/25/2017] [Accepted: 11/01/2017] [Indexed: 01/10/2023]
|
48
|
Pinto CL, Markey K, Dix D, Browne P. Identification of candidate reference chemicals for in vitro steroidogenesis assays. Toxicol In Vitro 2017; 47:103-119. [PMID: 29146384 DOI: 10.1016/j.tiv.2017.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/19/2017] [Accepted: 11/11/2017] [Indexed: 11/15/2022]
Abstract
The Endocrine Disruptor Screening Program (EDSP) is transitioning from traditional testing methods to integrating ToxCast/Tox21 in vitro high-throughput screening assays for identifying chemicals with endocrine bioactivity. The ToxCast high-throughput H295R steroidogenesis assay may potentially replace the low-throughput assays currently used in the EDSP Tier 1 battery to detect chemicals that alter the synthesis of androgens and estrogens. Herein, we describe an approach for identifying in vitro candidate reference chemicals that affect the production of androgens and estrogens in models of steroidogenesis. Candidate reference chemicals were identified from a review of H295R and gonad-derived in vitro assays used in methods validation and published in the scientific literature. A total of 29 chemicals affecting androgen and estrogen levels satisfied all criteria for positive reference chemicals, while an additional set of 21 and 15 chemicals partially fulfilled criteria for positive reference chemicals for androgens and estrogens, respectively. The identified chemicals included pesticides, pharmaceuticals, industrial and naturally-occurring chemicals with the capability to increase or decrease the levels of the sex hormones in vitro. Additionally, 14 and 15 compounds were identified as potential negative reference chemicals for effects on androgens and estrogens, respectively. These candidate reference chemicals will be informative for performance-based validation of in vitro steroidogenesis models.
Collapse
Affiliation(s)
- Caroline Lucia Pinto
- U.S. EPA, Office of Science Coordination and Policy, Washington, D.C. 20004, United States; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37831-0117, United States.
| | - Kristan Markey
- U.S. EPA, Office of Science Coordination and Policy, Washington, D.C. 20004, United States
| | - David Dix
- U.S. EPA, Office of Chemical Safety and Pollution Prevention, Washington, D.C. 20004, United States
| | - Patience Browne
- U.S. EPA, Office of Science Coordination and Policy, Washington, D.C. 20004, United States
| |
Collapse
|
49
|
Morais RDVS, Crespo D, Nóbrega RH, Lemos MS, van de Kant HJG, de França LR, Male R, Bogerd J, Schulz RW. Antagonistic regulation of spermatogonial differentiation in zebrafish (Danio rerio) by Igf3 and Amh. Mol Cell Endocrinol 2017. [PMID: 28645700 DOI: 10.1016/j.mce.2017.06.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fsh-mediated regulation of zebrafish spermatogenesis includes modulating the expression of testicular growth factors. Here, we study if and how two Sertoli cell-derived Fsh-responsive growth factors, anti-Müllerian hormone (Amh; inhibiting steroidogenesis and germ cell differentiation) and insulin-like growth factor 3 (Igf3; stimulating germ cell differentiation), cooperate in regulating spermatogonial development. In dose response and time course experiments with primary testis tissue cultures, Fsh up-regulated igf3 transcript levels and down-regulated amh transcript levels; igf3 transcript levels were more rapidly up-regulated and responded to lower Fsh concentrations than were required to decrease amh mRNA levels. Quantification of immunoreactive Amh and Igf3 on testis sections showed that Fsh increased slightly Igf3 staining but decreased clearly Amh staining. Studying the direct interaction of the two growth factors showed that Amh compromised Igf3-stimulated proliferation of type A (both undifferentiated [Aund] and differentiating [Adiff]) spermatogonia. Also the proliferation of those Sertoli cells associated with Aund spermatogonia was reduced by Amh. To gain more insight into how Amh inhibits germ cell development, we examined Amh-induced changes in testicular gene expression by RNA sequencing. The majority (69%) of the differentially expressed genes was down-regulated by Amh, including several stimulators of spermatogenesis, such as igf3 and steroidogenesis-related genes. At the same time, Amh increased the expression of inhibitory signals, such as inha and id3, or facilitated prostaglandin E2 (PGE2) signaling. Evaluating one of the potentially inhibitory signals, we indeed found in tissue culture experiments that PGE2 promoted the accumulation of Aund at the expense of Adiff and B spermatogonia. Our data suggest that an important aspect of Fsh bioactivity in stimulating spermatogenesis is implemented by restricting the different inhibitory effects of Amh and by counterbalancing them with stimulatory signals, such as Igf3.
Collapse
Affiliation(s)
- R D V S Morais
- Reproductive Biology Group (R.D.V.S.M., D.C., R.H.N., H.J.G.v.d.K., J.B., R.W.S.), Division of Developmental Biology, Institute for Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - D Crespo
- Reproductive Biology Group (R.D.V.S.M., D.C., R.H.N., H.J.G.v.d.K., J.B., R.W.S.), Division of Developmental Biology, Institute for Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - R H Nóbrega
- Reproductive Biology Group (R.D.V.S.M., D.C., R.H.N., H.J.G.v.d.K., J.B., R.W.S.), Division of Developmental Biology, Institute for Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands; Department of Morphology (R.H.N.), Institute of Bioscience, São Paulo State University, 18618-970 Botucatu, Brazil
| | - M S Lemos
- Laboratory of Cellular Biology (L.R.F., M.S.L.), Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - H J G van de Kant
- Reproductive Biology Group (R.D.V.S.M., D.C., R.H.N., H.J.G.v.d.K., J.B., R.W.S.), Division of Developmental Biology, Institute for Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - L R de França
- Laboratory of Cellular Biology (L.R.F., M.S.L.), Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, 31270-901 Belo Horizonte, Brazil; National Institute of Amazonian Research (L.R.F.), Manaus, Brazil
| | - R Male
- Department of Molecular Biology (R.M.), University of Bergen, 5020 Bergen, Norway
| | - J Bogerd
- Reproductive Biology Group (R.D.V.S.M., D.C., R.H.N., H.J.G.v.d.K., J.B., R.W.S.), Division of Developmental Biology, Institute for Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands.
| | - R W Schulz
- Reproductive Biology Group (R.D.V.S.M., D.C., R.H.N., H.J.G.v.d.K., J.B., R.W.S.), Division of Developmental Biology, Institute for Biodynamics and Biocomplexity, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands; Research Group Reproduction and Developmental Biology (R.W.S.), Institute of Marine Research, 5817 Bergen, Norway.
| |
Collapse
|
50
|
Xie Y, Chu L, Liu Y, Sham KWY, Li J, Cheng CHK. The highly overlapping actions of Lh signaling and Fsh signaling on zebrafish spermatogenesis. J Endocrinol 2017; 234:233-246. [PMID: 28611209 DOI: 10.1530/joe-17-0079] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 06/09/2017] [Indexed: 01/30/2023]
Abstract
Gonadotropin signaling plays a pivotal role in the spermatogenesis of vertebrates, but exactly how gonadotropins regulate the process in non-mammalian species remains elusive. Using a gene knockout approach in zebrafish, we have previously demonstrated the non-canonical action of gonadotropin signaling on spermatogenesis by analyzing four single mutant lines (lhb, lhr, fshb and fshr) and three double mutant lines (lhb;fshb, lhr;fshr and fshb;lhr). In this study, we further investigated the actions of gonadotropins on the testis by establishing three other double-mutant zebrafish lines (lhb;lhr, fshb;fshr and lhb;fshr). All lhb;lhr and fshb;fshr mutant males were fertile. Analysis on the gonadosomatic index and testicular histology in these lhb;lhr and fshb;fshr mutants demonstrated that Lh signaling and Fsh signaling could functionally compensate each other in the testis. Intriguingly, it was found that the lhb;fshr mutant male fish were also morphologically and histologically normal and functionally fertile, a phenomenon which could be explained by the cross-activation of Lhr by Fsh. We have demonstrated this cross-reactivity for the first time in zebrafish. Fsh was shown to activate Lhr using three different assay systems, in which Lh-Fshr activation was also confirmed. Taken together, we conclude that the action of Lh signaling and Fsh signaling is redundant in that either alone can support zebrafish spermatogenesis based on two observations. First, that either Lh signaling or Fsh signaling alone is sufficient to support male fertility. Second, that the two gonadotropin ligands could promiscuously activate both receptors. Apart from revealing the complexity of gonadotropin signaling in controlling male reproduction in zebrafish, this study also shed light toward a better understanding on the evolution of gonadotropin signaling in vertebrates from fish to mammals.
Collapse
Affiliation(s)
- Yuxin Xie
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Lianhe Chu
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Yun Liu
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Kathy W Y Sham
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Jianzhen Li
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
- College of Life SciencesNorthwest Normal University, Lanzhou, China
| | - Christopher H K Cheng
- School of Biomedical SciencesThe Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| |
Collapse
|