1
|
Dasso ME, Centola CL, Galardo MN, Riera MF, Meroni SB. FSH increases lipid droplet content by regulating the expression of genes related to lipid storage in Rat Sertoli cells. Mol Cell Endocrinol 2025; 595:112403. [PMID: 39490730 DOI: 10.1016/j.mce.2024.112403] [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: 09/16/2024] [Revised: 10/21/2024] [Accepted: 10/26/2024] [Indexed: 11/05/2024]
Abstract
Sertoli cells (SCs) are essential for appropriate spermatogenesis. From a metabolic standpoint, they catabolize glucose and provide germ cells with lactate, which is their main energy source. SCs also oxidize fatty acids (FAs), which are stored as triacylglycerides (TAGs) within lipid droplets (LDs), to fulfill their own energy requirements. On the other hand, it has been demonstrated that FSH regulates some of SCs functions, but little is known about its effect on lipid metabolism. In the present study, we aimed to analyze FSH-mediated regulation of (1) lipid storage in LDs and (2) the expression of genes involved in FAs activation and TAG synthesis and storage in SCs. SCs obtained from 20-day-old rats were cultured for different incubation periods with FSH (100 ng/ml). It was observed that FSH increased LD content and TAG levels in SCs. There were also increments in the expression of Plin1, Fabp5, Acsl1, Acsl4, Gpat3, and Dgat1, which suggests that these proteins may mediate the increase in TAGs and LDs elicited by FSH. Regarding the signaling involved in FSH actions, it was observed that dbcAMP increased LD, and H89, a PKA inhibitor, inhibited FSH stimulus. Also, dbcAMP increased Plin2, Fabp5, Acsl1, Acsl4, and Dgat1 mRNA levels, confirming a role of the cAMP/PKA pathway in the regulation of lipid storage in SCs. Altogether, these results suggest that FSH, via the cAMP/PKA pathway, regulates lipid storage in SCs ensuring the availability of substrates to satisfy their energy requirements.
Collapse
Affiliation(s)
- Marina Ercilia Dasso
- Centro de Investigaciones Endocrinológicas, "Dr César Bergadá", CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Argentina
| | - Cecilia Lucia Centola
- Centro de Investigaciones Endocrinológicas, "Dr César Bergadá", CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Argentina
| | - Maria Noel Galardo
- Centro de Investigaciones Endocrinológicas, "Dr César Bergadá", CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Argentina
| | - Maria Fernanda Riera
- Centro de Investigaciones Endocrinológicas, "Dr César Bergadá", CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Argentina
| | - Silvina Beatriz Meroni
- Centro de Investigaciones Endocrinológicas, "Dr César Bergadá", CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Argentina.
| |
Collapse
|
2
|
Cannarella R, Condorelli RA, Gusmano C, Garofalo V, Aversa A, Calogero AE, La Vignera S. Reply of the Authors: Rossella Cannarella, Rosita Condorelli, Carmelo Gusmano, et al. Predictive role of 17α-hydroxy-progesterone serum levels before and after follicle-stimulating hormone administration in patients with abnormal sperm parameters. Fertil Steril 2024; 121:1076-1077. [PMID: 38307454 DOI: 10.1016/j.fertnstert.2024.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/04/2024]
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Carmelo Gusmano
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Vincenzo Garofalo
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Antonio Aversa
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
3
|
Li WX, Cai LT, Huang YP, Huang YQ, Pan SH, Liu ZL, Ndandala CB, Shi G, Deng SP, Shi HJ, Li GL, Jiang DN. Sequence identification and expression characterization of leptin in the spotted scat, Scatophagus argus. Comp Biochem Physiol B Biochem Mol Biol 2024; 269:110882. [PMID: 37562672 DOI: 10.1016/j.cbpb.2023.110882] [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: 04/23/2023] [Revised: 08/06/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023]
Abstract
Scatophagus argus is an important marine culture fish in South and South-East Asia, including Southeast coastal areas of China. Artificial propagation technology for S. argus is not optimum; thus further studies on its reproduction biology are required. Although previous studies have shown that leptin (Lep) can regulate fish reproduction, the role of lep genes in S. argus is unknown. Herein, in silico analysis showed that S. argus has two lep genes (lepa and lepb). Protein 3D-structure prediction showed that Lepa has four α-helices (similar to mammals), while Lepb only has three. Tissue distribution analysis showed that lepa is highly expressed in the liver, whereas lepb was not detected in any tissue. Notably, lepr was expressed in all tissues. Lepa mRNA expression levels in the liver and serum Lep, estradiol (E2) and vitellogenin (Vtg) levels of female fish were significantly higher in ovaries at stage IV than in ovaries at stage II. Serum E2 levels were significantly positively correlated with Vtg levels in female fish at different development stages, while serum E2 was not correlated with Lep levels. Consistently, in vitro incubation of the liver with E2 significantly up-regulated vtga, while it did not affect lepa expression. Recombinant Lep (10 nM) significantly up-regulated chicken gonadotropin-releasing hormone (cGnRH/GnRH-II) in the hypothalamus and GnRH receptor (GnRHR) and luteinizing hormone beta (Lhb) in the pituitary. These results suggest that lepa regulates female reproduction in S. argus.
Collapse
Affiliation(s)
- Wan-Xin Li
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Li-Ting Cai
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Yan-Ping Huang
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Yuan-Qing Huang
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Shu-Hui Pan
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Zhi-Long Liu
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Charles Brighton Ndandala
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Gang Shi
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Si-Ping Deng
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Hong-Juan Shi
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Guang-Li Li
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China
| | - Dong-Neng Jiang
- Fisheries College of Guangdong Ocean University, Guangdong Province Famous Fish Reproduction Regulation and Breeding Engineering Technology Research Center, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, 524088 Zhanjiang, China.
| |
Collapse
|
4
|
Danti L, Lundin K, Sepponen K, Yohannes DA, Kere J, Tuuri T, Tapanainen JS. CRISPR/Cas9-mediated activation of NR5A1 steers female human embryonic stem cell-derived bipotential gonadal-like cells towards a steroidogenic cell fate. J Ovarian Res 2023; 16:194. [PMID: 37726790 PMCID: PMC10510196 DOI: 10.1186/s13048-023-01264-5] [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: 03/31/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
The nuclear receptor subfamily 5 group A member 1 (NR5A1), encoding steroidogenic factor 1 (SF-1), has been identified as a critical factor in gonadal development in animal studies. A previous study of ours suggested that upregulation of NR5A1 during early gonadal differentiation in male (46,XY) human pluripotent stem cells steers the cells into a more mature gonadal cell type. However, the detailed role of NR5A1 in female gonadal differentiation has yet to be determined. In this study, by combining the processes of gonadal differentiation and conditional gene activation, we show that NR5A1 induction predominantly upregulates the female gonadal marker inhibin subunit α (INHA) and steroidogenic markers steroidogenic acute regulatory protein (STAR), cytochrome P450 family 11 subfamily A member 1 (CYP11A1), cytochrome P450 family 17 subfamily A member 1 (CYP17A1), hydroxy-delta-5-steroid dehydrogenase (HSD3B2) and hydroxysteroid 17-beta dehydrogenase 1 (HSD17B1). In contrast, NR5A1 induction did not seem to affect the bipotential gonadal markers gata binding protein 4 (GATA4) and Wilms' tumour suppressor 1 (WT1) nor the female gonadal markers r-spondin 1 (RSPO1) and wnt family member 4 (WNT4). Differentially expressed genes were highly associated with adrenal and ovarian steroidogenesis pathways. Moreover, time-series analysis revealed different dynamic changes between male and female induced samples, where continuously upregulated genes in female gonadal differentiation were mostly associated with adrenal steroidogenesis. Thus, in contrast to male gonadal differentiation, NR5A1 is necessary but not sufficient to steer human embryonic stem cell (hESC)-derived bipotential gonadal-like cells towards a more mature somatic, female cell fate. Instead, it seems to direct bipotential gonadal-like cells more towards a steroidogenic-like cell population. The information obtained in this study helps in elucidating the role of NR5A1 in gonadal differentiation of a female stem cell line.
Collapse
Affiliation(s)
- Laura Danti
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, 00290, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14183, Sweden
| | - Karolina Lundin
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, 00290, Finland
| | - Kirsi Sepponen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, 00290, Finland
| | - Dawit A Yohannes
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, 00290, Finland
- Research Programs Unit, Translational Immunology & Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, 00290, Finland
| | - Juha Kere
- Folkhälsan Research Centre, Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, 00290, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14183, Sweden
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, 00290, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, 00290, Finland.
- Department of Obstetrics and Gynecology, HFR - Cantonal Hospital of Fribourg and University of Fribourg, Fribourg, 1708, Switzerland.
| |
Collapse
|
5
|
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
|
6
|
Miller SC, MacDonald CC, Kellogg MK, Karamysheva ZN, Karamyshev AL. Specialized Ribosomes in Health and Disease. Int J Mol Sci 2023; 24:ijms24076334. [PMID: 37047306 PMCID: PMC10093926 DOI: 10.3390/ijms24076334] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Ribosomal heterogeneity exists within cells and between different cell types, at specific developmental stages, and occurs in response to environmental stimuli. Mounting evidence supports the existence of specialized ribosomes, or specific changes to the ribosome that regulate the translation of a specific group of transcripts. These alterations have been shown to affect the affinity of ribosomes for certain mRNAs or change the cotranslational folding of nascent polypeptides at the exit tunnel. The identification of specialized ribosomes requires evidence of the incorporation of different ribosomal proteins or of modifications to rRNA and/or protein that lead(s) to physiologically relevant changes in translation. In this review, we summarize ribosomal heterogeneity and specialization in mammals and discuss their relevance to several human diseases.
Collapse
Affiliation(s)
- Sarah C. Miller
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Clinton C. MacDonald
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Morgana K. Kellogg
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | - Andrey L. Karamyshev
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Correspondence: ; Tel.: +1-806-743-4102
| |
Collapse
|
7
|
Lustofin S, Kaminska A, Brzoskwinia M, Pardyak L, Pawlicki P, Szpregiel I, Bilinska B, Hejmej A. Follicle-stimulating hormone regulates Notch signalling in the seminiferous epithelium of continuously and seasonally breeding rodents. Reprod Fertil Dev 2022; 34:560-575. [PMID: 35143740 DOI: 10.1071/rd21237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/17/2022] [Indexed: 12/15/2022] Open
Abstract
CONTEXT Juxtacrine (contact-dependent) communication between the cells of seminiferous epithelium mediated by Notch signalling is of importance for the proper course of spermatogenesis in mammals. AIMS The present study was designed to evaluate the role of follicle-stimulating hormone (FSH) in the regulation of Notch signalling in rodent seminiferous epithelium. METHODS We explored the effects (1) of pharmacological inhibition of the hypothalamus-pituitary-gonadal (HPG) axis and FSH replacement in pubertal rats, and (2) of photoinhibition of HPG axis followed by FSH substitution in seasonally breeding rodents, bank voles, on Notch pathway activity. Experiments on isolated rat Sertoli cells exposed to FSH were also performed. Gene and protein expressions of Notch pathway components were analysed using RT-qPCR, western blot and immunohistochemistry/immunofluorescence. KEY RESULTS Distribution patterns of Notch pathway proteins in bank vole and rat seminiferous epithelium were comparable; however, levels of activated Notch1 and Notch3, hairy/enhancer of split 1 (HES1) and hairy/enhancer of split-related with YRPW motif 1 (HEY1) in bank voles were dependent on the length of the photoperiod. In response to FSH similar changes in these proteins were found in both species, indicating that FSH is a negative regulator of Notch pathway activity in seminiferous epithelium. CONCLUSIONS Our results support a common mechanism of FSH action on Notch pathway during onset and recrudescence of spermatogenesis in rodents. IMPLICATIONS Interaction between FSH signalling and Notch pathway in Sertoli cells may be involved in spermatogenic activity changes of the testes occurring during puberty or photoperiod shift in continuously and seasonally breeding rodents, respectively.
Collapse
Affiliation(s)
- Sylwia Lustofin
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland
| | - Alicja Kaminska
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland
| | - Malgorzata Brzoskwinia
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland
| | - Laura Pardyak
- Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, 30-248 Krakow, Poland
| | - Piotr Pawlicki
- Center of Experimental and Innovative Medicine, University of Agriculture in Krakow, 30-248 Krakow, Poland
| | - Izabela Szpregiel
- Department of Animal Physiology and Endocrinology, Faculty of Animal Science, University of Agriculture in Krakow, 30-059 Krakow, Poland
| | - Barbara Bilinska
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland
| | - Anna Hejmej
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, 30-387 Krakow, Poland
| |
Collapse
|
8
|
Long non-coding ROR promotes the progression of papillary thyroid carcinoma through regulation of the TESC/ALDH1A1/TUBB3/PTEN axis. Cell Death Dis 2022; 13:157. [PMID: 35173149 PMCID: PMC8850450 DOI: 10.1038/s41419-021-04210-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/02/2021] [Accepted: 09/21/2021] [Indexed: 12/16/2022]
Abstract
Papillary thyroidal carcinoma (PTC) is a common endocrine cancer that plagues people across the world. The potential roles of long non-coding RNAs (lncRNAs) in PTC have gained increasing attention. In this study, we aimed to explore whether lncRNA ROR affects the progression of PTC, with the involvement of tescalcin (TESC)/aldehyde dehydrogenase isoform 1A1 (ALDH1A1)/βIII-tubulin (TUBB3)/tensin homolog (PTEN) axis. PTC tumor and adjacent tissues were obtained, followed by measurement of lncRNA ROR and TESC, ALDH1A1, and TUBB3 expression. Interactions among lncRNA ROR, TESC, ALDH1A1, TUBB3, and PTEN were evaluated by ChIP assay, RT-qPCR, or western blot analysis. After ectopic expression and depletion experiments in PTC cells, MTT and colony formation assay, Transwell assay, and flow cytometry were performed to detect cell viability and colony formation, cell migration and invasion, and apoptosis, respectively. In addition, xenograft in nude mice was performed to test the effects of lncRNA ROR and PTEN on tumor growth in PTC in vivo. LncRNA ROR, TESC, ALDH1A1, and TUBB3 were highly expressed in PTC tissues and cells. Overexpression of lncRNA ROR activated TESC by inhibiting the G9a recruitment on the promoter of TESC and histone H3-lysine 9me methylation. Moreover, TESC upregulated ALDH1A1 expression to increase TUBB3 expression, which then reduced PTEN expression. Overexpression of lncRNA ROR, TESC, ALDH1A1 or TUBB3 and silencing of PTEN promoted PTC cell viability, colony formation, migration, and invasion while suppressing apoptosis. Moreover, overexpression of lncRNA ROR increased tumor growth by inhibiting PTEN in vivo. Taken together, the current study demonstrated that lncRNA ROR mediated TESC/ALDH1A1/TUBB3/PTEN axis, thereby facilitating the development of PTC.
Collapse
|
9
|
Follicle-stimulating Hormone (FSH) Action on Spermatogenesis: A Focus on Physiological and Therapeutic Roles. J Clin Med 2020; 9:jcm9041014. [PMID: 32260182 PMCID: PMC7230878 DOI: 10.3390/jcm9041014] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human reproduction is regulated by the combined action of the follicle-stimulating hormone (FSH) and the luteinizing hormone (LH) on the gonads. Although FSH is largely used in female reproduction, in particular in women attending assisted reproductive techniques to stimulate multi-follicular growth, its efficacy in men with idiopathic infertility is not clearly demonstrated. Indeed, whether FSH administration improves fertility in patients with hypogonadotropic hypogonadism, the therapeutic benefit in men presenting alterations in sperm production despite normal FSH serum levels is still unclear. In the present review, we evaluate the potential pharmacological benefits of FSH administration in clinical practice. METHODS This is a narrative review, describing the FSH physiological role in spermatogenesis and its potential therapeutic action in men. RESULTS The FSH role on male fertility is reviewed starting from the physiological control of spermatogenesis, throughout its mechanism of action in Sertoli cells, the genetic regulation of its action on spermatogenesis, until the therapeutic options available to improve sperm production. CONCLUSION FSH administration in infertile men has potential benefits, although its action should be considered by evaluating its synergic action with testosterone, and well-controlled, powerful trials are required. Prospective studies and new compounds could be developed in the near future.
Collapse
|
10
|
Fonseca PADS, dos Santos FC, Lam S, Suárez-Vega A, Miglior F, Schenkel FS, Diniz LDAF, Id-Lahoucine S, Carvalho MRS, Cánovas A. Genetic mechanisms underlying spermatic and testicular traits within and among cattle breeds: systematic review and prioritization of GWAS results. J Anim Sci 2018; 96:4978-4999. [PMID: 30304443 PMCID: PMC6276581 DOI: 10.1093/jas/sky382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/27/2018] [Indexed: 12/20/2022] Open
Abstract
Reduced bull fertility imposes economic losses in bovine herds. Specifically, testicular and spermatic traits are important indicators of reproductive efficiency. Several genome-wide association studies (GWAS) have identified genomic regions associated with these fertility traits. The aims of this study were as follows: 1) to perform a systematic review of GWAS results for spermatic and testicular traits in cattle and 2) to identify key functional candidate genes for these traits. The identification of functional candidate genes was performed using a systems biology approach, where genes shared between traits and studies were evaluated by a guilt by association gene prioritization (GUILDify and ToppGene software) in order to identify the best functional candidates. These candidate genes were integrated and analyzed in order to identify overlapping patterns among traits and breeds. Results showed that GWAS for testicular-related traits have been developed for beef breeds only, whereas the majority of GWAS for spermatic-related traits were conducted using dairy breeds. When comparing traits measured within the same study, the highest number of genes shared between different traits was observed, indicating a high impact of the population genetic structure and environmental effects. Several chromosomal regions were enriched for functional candidate genes associated with fertility traits. Moreover, multiple functional candidate genes were enriched for markers in a species-specific basis, taurine (Bos taurus) or indicine (Bos indicus). For the different candidate regions identified in the GWAS in the literature, functional candidate genes were detected as follows: B. Taurus chromosome X (BTX) (TEX11, IRAK, CDK16, ATP7A, ATRX, HDAC6, FMR1, L1CAM, MECP2, etc.), BTA17 (TRPV4 and DYNLL1), and BTA14 (MOS, FABP5, ZFPM2). These genes are responsible for regulating important metabolic pathways or biological processes associated with fertility, such as progression of spermatogenesis, control of ciliary activity, development of Sertoli cells, DNA integrity in spermatozoa, and homeostasis of testicular cells. This study represents the first systematic review on male fertility traits in cattle using a system biology approach to identify key candidate genes for these traits.
Collapse
Affiliation(s)
- Pablo Augusto de Souza Fonseca
- Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | | | - Stephanie Lam
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | - Aroa Suárez-Vega
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | - Filippo Miglior
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | - Flavio S Schenkel
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | | | - Samir Id-Lahoucine
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| | | | - Angela Cánovas
- Department of Animal Biosciences, Centre for Genetic Improvement of Livestock, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
11
|
Jia X, Liu Z, Lu X, Tang J, Wu Y, Du Q, He J, Zhang X, Jiang J, Liu W, Zheng Y, Ding Y, Zhu W, Zhang H. Effects of MCLR exposure on sex hormone synthesis and reproduction-related genes expression of testis in male Rana nigromaculata. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 236:12-20. [PMID: 29414332 DOI: 10.1016/j.envpol.2018.01.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 01/12/2018] [Accepted: 01/17/2018] [Indexed: 06/08/2023]
Abstract
Microcystin-leucine-arginine (MCLR) is the most popular and toxic variant among microcystins, which can cause severe reproductive toxicity to animals. However, the mechanisms of reproductive toxicity induced by MCLR in amphibians are still not entirely clear. In the current study, toxicity mechanisms of MCLR on the reproductive system of male Rana nigromaculata followed by low concentration (0, 0.1, 1, and 10 μg/L) and short-term (0, 7, and 14 days) MCLR exposure were shown. It was observed that MCLR could be bioaccumulated in the testes of male frogs, and the theoretical bioaccumulation factor values were 0.24 and 0.19 exposed to 1 μg/L and 10 μg/L MCLR for 14 days, respectively. MCLR exposure significantly decreased testosterone (T) concentrations and increased estradiol (E2) concentrations exposed to 1 and 10 μg/L MCLR for 14 days. The mRNA levels of HSD17B3 were downregulated, and HSD17B1 and CYP19A1 mRNA expression levels were upregulated, respectively. Only 10 μg/L MCLR group showed significant induction of follicle-stimulating hormone (FSH) levels and cyclic adenosine monophosphate (cAMP) content. Moreover, AR and ESR1 mRNA expression levels were significantly upregulated exposed to 1 and 10 μg/L MCLR for 14 days, respectively. Our results suggested that low-concentration MCLR induced transcription changes of CYP19A1, HSD17B3, and HSD17B1 led to endocrine disorders, and caused interference of spermatogenesis by the decrease of T and abnormal gene expressions of AR and ESR1 in the testes of R. nigromaculata.
Collapse
Affiliation(s)
- Xiuying Jia
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Zhengquan Liu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Xiangjun Lu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Juan Tang
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Yingzhu Wu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Qiongxia Du
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Jianbo He
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Xinyun Zhang
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Jinxiao Jiang
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Wenli Liu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Yuqing Zheng
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China
| | - Ying Ding
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China; Key Laboratory of Hangzhou City for Ecosystem Protection and Restoration, Hangzhou Normal University, Hangzhou 310036, China
| | - Weiqin Zhu
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China; Key Laboratory of Hangzhou City for Ecosystem Protection and Restoration, Hangzhou Normal University, Hangzhou 310036, China
| | - Hangjun Zhang
- College of Life and Environmental Sciences, Hangzhou Normal University, Xuelin Road 16#, Xiasha Gaojiao Dongqu, Hangzhou, Zhejiang Province, 310036, China; Guangzhou Key Laboratory of Environmental Exposure and Health, School of Environment, Jinan University, Guangzhou 510632, China; Key Laboratory of Hangzhou City for Ecosystem Protection and Restoration, Hangzhou Normal University, Hangzhou 310036, China.
| |
Collapse
|
12
|
Gautam M, Bhattacharya I, Rai U, Majumdar SS. Hormone induced differential transcriptome analysis of Sertoli cells during postnatal maturation of rat testes. PLoS One 2018; 13:e0191201. [PMID: 29342173 PMCID: PMC5771609 DOI: 10.1371/journal.pone.0191201] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 12/30/2017] [Indexed: 11/18/2022] Open
Abstract
Sertoli cells (Sc) are unique somatic cells of testis that are the target of both FSH and testosterone (T) and regulate spermatogenesis. Although Sc of neonatal rat testes are exposed to high levels of FSH and T, robust differentiation of spermatogonial cells becomes conspicuous only after 11-days of postnatal age. We have demonstrated earlier that a developmental switch in terms of hormonal responsiveness occurs in rat Sc at around 12 days of postnatal age during the rapid transition of spermatogonia A to B. Therefore, such “functional maturation” of Sc, during pubertal development becomes prerequisite for the onset of spermatogenesis. However, a conspicuous difference in robust hormone (both T and FSH) induced gene expression during the different phases of Sc maturation restricts our understanding about molecular events necessary for the spermatogenic onset and maintenance. Here, using microarray technology, we for the first time have compared the differential transcriptional profile of Sc isolated and cultured from immature (5 days old), maturing (12 days old) and mature (60 days old) rat testes. Our data revealed that immature Sc express genes involved in cellular growth, metabolism, chemokines, cell division, MAPK and Wnt pathways, while mature Sc are more specialized expressing genes involved in glucose metabolism, phagocytosis, insulin signaling and cytoskeleton structuring. Taken together, this differential transcriptome data provide an important resource to reveal the molecular network of Sc maturation which is necessary to govern male germ cell differentiation, hence, will improve our current understanding of the etiology of some forms of idiopathic male infertility.
Collapse
Affiliation(s)
- Mukesh Gautam
- Department of Zoology, University of Delhi, Delhi, India
| | | | - Umesh Rai
- Department of Zoology, University of Delhi, Delhi, India
| | - Subeer S. Majumdar
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, India
- National Institute of Animal Biotechnology, Hyderabad, India
- * E-mail:
| |
Collapse
|
13
|
Sominsky L, Goularte JF, Andrews ZB, Spencer SJ. Acylated Ghrelin Supports the Ovarian Transcriptome and Follicles in the Mouse: Implications for Fertility. Front Endocrinol (Lausanne) 2018; 9:815. [PMID: 30697193 PMCID: PMC6340924 DOI: 10.3389/fendo.2018.00815] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/27/2018] [Indexed: 12/16/2022] Open
Abstract
Ghrelin, an orexigenic gut-derived peptide, is gaining increasing attention due to its multifaceted role in a number of physiological functions, including reproduction. Ghrelin exists in circulation primarily as des-acylated and acylated ghrelin. Des-acyl ghrelin, until recently considered to be an inactive form of ghrelin, is now known to have independent physiological functionality. However, the relative contribution of acyl and des-acyl ghrelin to reproductive development and function is currently unknown. Here we used ghrelin-O-acyltransferase (GOAT) knockout (KO) mice that have no measurable levels of endogenous acyl ghrelin and chronically high levels of des-acyl ghrelin, to characterize how the developmental and life-long absence of acyl ghrelin affects ovarian development and reproductive capacity. We combined the assessment of markers of reproductive maturity and the capacity to breed with measures of ovarian morphometry, as well as with ovarian RNA sequencing analysis. Our data show that while GOAT KO mice retain the capacity to breed in young adulthood, there is a diminished number of ovarian follicles (per mm3) in the juvenile and adult ovaries, due to a significant reduction in the number of small follicles, particularly the primordial follicles. We also show pronounced specific changes in the ovarian transcriptome in the juvenile GOAT KO ovary, indicative of a potential for premature ovarian development. Collectively, these findings indicate that an absence of acyl ghrelin does not prevent reproductive success but that appropriate levels of acyl and des-acyl ghrelin may be necessary for optimal ovarian maturation.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
- *Correspondence: Luba Sominsky
| | - Jeferson F. Goularte
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Zane B. Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Sarah J. Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Nikbin S, Panandam JM, Yaakub H, Murugaiyah M. Association of novel SNPs in gonadotropin genes with sperm quality traits of Boer goats and Boer crosses. JOURNAL OF APPLIED ANIMAL RESEARCH 2018. [DOI: 10.1080/09712119.2017.1336441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Saeid Nikbin
- Department of Animal Science, Faculty of Agriculture, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Jothi Malar Panandam
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, Malaysia
| | - Halimatun Yaakub
- Department of Animal Science, Faculty of Agriculture, Universiti Putra Malaysia, Serdang, Malaysia
| | | |
Collapse
|
15
|
Follicle-Stimulating Hormone Receptor: Advances and Remaining Challenges. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:1-58. [DOI: 10.1016/bs.ircmb.2018.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
16
|
Chauvigné F, Ollé J, González W, Duncan N, Giménez I, Cerdà J. Toward developing recombinant gonadotropin-based hormone therapies for increasing fertility in the flatfish Senegalese sole. PLoS One 2017; 12:e0174387. [PMID: 28329024 PMCID: PMC5362233 DOI: 10.1371/journal.pone.0174387] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/08/2017] [Indexed: 11/18/2022] Open
Abstract
Captive flatfishes, such as the Senegalese sole, typically produce very low volumes of sperm. This situation is particularly prevalent in the first generation (F1) of reared sole males, which limits the development of artificial fertilization methods and the implementation of selective breeding programs. In this study, we investigated whether combined treatments with homologous recombinant follicle-stimulating (rFsh) and luteinizing (rLh) hormones, produced in a mammalian host system, could stimulate spermatogenesis and enhance sperm production in Senegalese sole F1 males. In an initial autumn/winter experiment, weekly intramuscular injections with increasing doses of rFsh over 9 weeks resulted in the stimulation of gonad weight, androgen release, germ cell proliferation and entry into meiosis, and the expression of different spermatogenesis-related genes, whereas a subsequent single rLh injection potentiated spermatozoa differentiation. In a second late winter/spring trial corresponding to the sole’s natural prespawning and spawning periods, we tested the effect of repeated rLh injections on the amount and quality of sperm produced by males previously treated with rFsh for 4, 6, 8 or 10 weeks. These latter results showed that the combination of rFsh and rLh treatments could increase sperm production up to 7 times, and slightly improve the motility of the spermatozoa, although a high variability in the response was found. However, sustained administration of rFsh during spawning markedly diminished Leydig cell survival and the steroidogenic potential of the testis. These data suggest that in vivo application of rFsh and rLh is effective at stimulating spermatogenesis and sperm production in Senegalese sole F1 males, setting the basis for the future establishment of recombinant gonadotropin-based hormone therapies to ameliorate reproductive dysfunctions of this species.
Collapse
Affiliation(s)
- François Chauvigné
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA)-Institut de Ciències del Mar, Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- * E-mail: (FC); (IG); (JC)
| | - Judith Ollé
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA)-Institut de Ciències del Mar, Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | | | - Neil Duncan
- IRTA, Sant Carles de la Ràpita, Tarragona, Spain
| | - Ignacio Giménez
- Rara Avis Biotec, S. L., Valencia, Spain
- * E-mail: (FC); (IG); (JC)
| | - Joan Cerdà
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA)-Institut de Ciències del Mar, Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- * E-mail: (FC); (IG); (JC)
| |
Collapse
|
17
|
Patel H, Bhartiya D. Testicular Stem Cells Express Follicle-Stimulating Hormone Receptors and Are Directly Modulated by FSH. Reprod Sci 2016; 23:1493-1508. [PMID: 27189070 DOI: 10.1177/1933719116643593] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Testicular spermatogonial stem cells (SSCs) are a heterogeneous population of stem cells, and definitive marker for the most primitive subset that undergoes asymmetric cell division remains to be identified. A novel subpopulation of pluripotent, very small embryonic-like stem cells (VSELs) has been reported in both human and mouse testes. Follicle-stimulating hormone (FSH) receptors (FSHRs) are expressed on Sertoli cells in testis and on granulosa cells in ovary, but recently FSHRs are reported on VSELs in ovaries, bone marrow, and cord blood. The present study was aimed to investigate whether FSHRs are also expressed on testicular stem cells (VSELs and SSCs) and their possible modulation by FSH using intact and chemoablated (25 mg/kg busulfan) mice. Chemoablated testis was a better model to study stem cell biology since quiescent stem cells survive along with the Sertoli cells in the tubules. Proliferating cell nuclear antigen-positive, small-sized cells presumed to be VSELs were clearly visualized, and flow cytometry analysis revealed an increase in LIN-/CD45-/SCA-1+ VSELs from 0.045±0.008% to 0.1±0.03% of total cells in chemoablated testis after FSH treatment. Very small embryonic-like stem cells expressing nuclear octamer-binding transcription factor 4 (OCT-4) and SSCs with cytoplasmic OCT-4 were detected. Very small embryonic-like stem cells (Oct-4A, Sca-1, Nanog), SSCs (Oct-4), and proliferation (Pcna) specific transcripts were upregulated on FSH treatment. Stem cells expressed FSHR and were stimulated by FSH, and Fshr3 was the predominant transcript maximally modulated by FSH. Nuclear OCT-4 and SCA-1 (stem cell antigen 1) positive VSELs are the most primitive stem cells in testis, and FSH stimulates them to undergo asymmetric cell division including self-renewal and give rise to SSCs, which in turn proliferate rapidly and undergo clonal expansion and further differentiation.
Collapse
Affiliation(s)
- Hiren Patel
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| | - Deepa Bhartiya
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Mumbai, India
| |
Collapse
|
18
|
Herrera-Luna CV, Scarlet D, Walter I, Aurich C. Effect of stallion age on the expression of LH and FSH receptors and aromatase P450 in equine male reproductive tissues. Reprod Fertil Dev 2016; 28:2016-2026. [DOI: 10.1071/rd15027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 06/06/2015] [Indexed: 11/23/2022] Open
Abstract
The aim of the present study was to characterise receptors for LH and FSH (LHR and FSHR, respectively) and aromatase in epididymal and testicular tissue from stallions of different ages (prepubertal, young, mature and old). Gene and protein expression were assessed by real-time quantitative polymerase chain reaction (real-time qPCR), immunohistochemistry and multiple immunofluorescence labelling. There were no differences in LHR mRNA expression in epididymal and testicular parenchyma in stallions of different age. In contrast, expression of FSHR and CYP19A1 in caput, corpus and cauda epididymis and in testicular parenchyma increased with age (P < 0.001). Immunolabelling for LHR, FSHR and aromatase was influenced by puberty. In postpubertal stallions, positive staining for LHR and aromatase was detected in Leydig cells, whereas protein expression of FSHR was present in Sertoli cells and primary spermatocytes. In prepubertal colts, staining for LHR, FSHR and aromatase was detected in seminiferous tubules. In epididymal tissue, aromatase was present in the cauda epididymis only, regardless of age. In conclusion, the results highlight the significance of gonadotropin action and oestrogen production for the maturation of male reproductive tissue in the horse. The presence of FSHR in the seminiferous tubules suggests effects of FSH on spermatogenesis in this species. The importance of oestrogen production for maintenance of testicular function in stallions was confirmed.
Collapse
|
19
|
Mazón MJ, Molés G, Rocha A, Crespo B, Lan-Chow-Wing O, Espigares F, Muñoz I, Felip A, Carrillo M, Zanuy S, Gómez A. Gonadotropins in European sea bass: Endocrine roles and biotechnological applications. Gen Comp Endocrinol 2015; 221:31-41. [PMID: 26002037 DOI: 10.1016/j.ygcen.2015.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 04/20/2015] [Accepted: 05/05/2015] [Indexed: 12/28/2022]
Abstract
Follicle stimulating hormone (Fsh) and luteinizing hormone (Lh) are central endocrine regulators of the gonadal function in vertebrates. They act through specific receptors located in certain cell types found in the gonads. In fish, the differential roles of these hormones are being progressively elucidated due to the development of suitable tools for their study. In European sea bass (Dicentrarchus labrax), isolation of the genes coding for the gonadotropin subunits and receptors allowed in first instance to conduct expression studies. Later, to overcome the limitation of using native hormones, recombinant dimeric gonadotropins, which show different functional characteristics depending on the cell system and DNA construct, were generated. In addition, single gonadotropin beta-subunits have been produced and used as antigens for antibody production. This approach has allowed the development of detection methods for native gonadotropins, with European sea bass being one of the few species where both gonadotropins can be detected in their native form. By administering recombinant gonadotropins to gonad tissues in vitro, we were able to study their effects on steroidogenesis and intracellular pathways. Their administration in vivo has also been tested for use in basic studies and as a biotechnological approach for hormone therapy and assisted reproduction strategies. In addition to the production of recombinant hormones, gene-based therapies using somatic gene transfer have been offered as an alternative. This approach has been tested in sea bass for gonadotropin delivery in vivo. The hormones produced by the genes injected were functional and have allowed studies on the action of gonadotropins in spermatogenesis.
Collapse
Affiliation(s)
- María José Mazón
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Gregorio Molés
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Ana Rocha
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Berta Crespo
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Olivier Lan-Chow-Wing
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Felipe Espigares
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Iciar Muñoz
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Alicia Felip
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Manuel Carrillo
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Silvia Zanuy
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Ana Gómez
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain.
| |
Collapse
|
20
|
Chauvigné F, Zapater C, Crespo D, Planas JV, Cerdà J. Fsh and Lh direct conserved and specific pathways during flatfish semicystic spermatogenesis. J Mol Endocrinol 2014; 53:175-90. [PMID: 25024405 DOI: 10.1530/jme-14-0087] [Citation(s) in RCA: 18] [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] [Indexed: 12/14/2022]
Abstract
The current view of the control of spermatogenesis by Fsh and Lh in non-mammalian vertebrates is largely based on studies carried out in teleosts with cystic and cyclic spermatogenesis. Much less is known concerning the specific actions of gonadotropins during semicystic germ cell development, a type of spermatogenesis in which germ cells are released into the tubular lumen where they transform into spermatozoa. In this study, using homologous gonadotropins and a candidate gene approach, for which the genes' testicular cell-type-specific expression was established, we investigated the regulatory effects of Fsh and Lh on gene expression during spermatogenesis in Senegalese sole (Solea senegalensis), a flatfish with asynchronous and semicystic germ cell development. During early spermatogenesis, Fsh and Lh upregulated steroidogenesis-related genes and nuclear steroid receptors, expressed in both somatic and germ cells, through steroid-dependent pathways, although Lh preferentially stimulated the expression of downstream genes involved in androgen and progestin syntheses. In addition, Lh specifically promoted the expression of spermatid-specific genes encoding spermatozoan flagellar proteins through direct interaction with the Lh receptor in these cells. Interestingly, at this spermatogenic stage, Fsh primarily regulated genes encoding Sertoli cell growth factors with potentially antagonistic effects on germ cell proliferation and differentiation through steroid mediation. During late spermatogenesis, fewer genes were regulated by Fsh or Lh, which was associated with a translational and posttranslational downregulation of the Fsh receptor in different testicular compartments. These results reveal that conserved and specialized gonadotropic pathways regulate semicystic spermatogenesis in flatfish, which may spatially adjust cell germ development to maintain a continuous reservoir of spermatids in the testis.
Collapse
Affiliation(s)
- François Chauvigné
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA)-Institut de Ciències del MarConsejo Superior de Investigaciones Científicas (CSIC), Passeig marítim 37-49, 08003 Barcelona, SpainDepartament de Fisiologia i ImmunologiaFacultat de Biologia, Universitat de Barcelona, i Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
| | - Cinta Zapater
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA)-Institut de Ciències del MarConsejo Superior de Investigaciones Científicas (CSIC), Passeig marítim 37-49, 08003 Barcelona, SpainDepartament de Fisiologia i ImmunologiaFacultat de Biologia, Universitat de Barcelona, i Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
| | - Diego Crespo
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA)-Institut de Ciències del MarConsejo Superior de Investigaciones Científicas (CSIC), Passeig marítim 37-49, 08003 Barcelona, SpainDepartament de Fisiologia i ImmunologiaFacultat de Biologia, Universitat de Barcelona, i Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
| | - Josep V Planas
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA)-Institut de Ciències del MarConsejo Superior de Investigaciones Científicas (CSIC), Passeig marítim 37-49, 08003 Barcelona, SpainDepartament de Fisiologia i ImmunologiaFacultat de Biologia, Universitat de Barcelona, i Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
| | - Joan Cerdà
- Institut de Recerca i Tecnologia Agroalimentàries (IRTA)-Institut de Ciències del MarConsejo Superior de Investigaciones Científicas (CSIC), Passeig marítim 37-49, 08003 Barcelona, SpainDepartament de Fisiologia i ImmunologiaFacultat de Biologia, Universitat de Barcelona, i Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain
| |
Collapse
|
21
|
Rebourcet D, O’Shaughnessy PJ, Monteiro A, Milne L, Cruickshanks L, Jeffrey N, Guillou F, Freeman TC, Mitchell RT, Smith LB. Sertoli cells maintain Leydig cell number and peritubular myoid cell activity in the adult mouse testis. PLoS One 2014; 9:e105687. [PMID: 25144714 PMCID: PMC4140823 DOI: 10.1371/journal.pone.0105687] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/23/2014] [Indexed: 01/08/2023] Open
Abstract
The Sertoli cells are critical regulators of testis differentiation and development. In the adult, however, their known function is restricted largely to maintenance of spermatogenesis. To determine whether the Sertoli cells regulate other aspects of adult testis biology we have used a novel transgenic mouse model in which Amh-Cre induces expression of the receptor for Diphtheria toxin (iDTR) specifically within Sertoli cells. This causes controlled, cell-specific and acute ablation of the Sertoli cell population in the adult animal following Diphtheria toxin injection. Results show that Sertoli cell ablation leads to rapid loss of all germ cell populations. In addition, adult Leydig cell numbers decline by 75% with the remaining cells concentrated around the rete and in the sub-capsular region. In the absence of Sertoli cells, peritubular myoid cell activity is reduced but the cells retain an ability to exclude immune cells from the seminiferous tubules. These data demonstrate that, in addition to support of spermatogenesis, Sertoli cells are required in the adult testis both for retention of the normal adult Leydig cell population and for support of normal peritubular myoid cell function. This has implications for our understanding of male reproductive disorders and wider androgen-related conditions affecting male health.
Collapse
Affiliation(s)
- Diane Rebourcet
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Peter J. O’Shaughnessy
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Garscube Campus, Glasgow, United Kingdom
| | - Ana Monteiro
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Garscube Campus, Glasgow, United Kingdom
| | - Laura Milne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Lyndsey Cruickshanks
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Nathan Jeffrey
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Florian Guillou
- Station de Physiologie de la Reproduction et des Comportements (PRC), Institut National de la Recherche Agronomique, UMR 6073 INRA-CNRS-Université de Tours, Nouzilly, France
| | - Tom C. Freeman
- The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, United Kingdom
| | - Rod T. Mitchell
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Lee B. Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom
- * E-mail:
| |
Collapse
|
22
|
O'Shaughnessy PJ. Hormonal control of germ cell development and spermatogenesis. Semin Cell Dev Biol 2014; 29:55-65. [DOI: 10.1016/j.semcdb.2014.02.010] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 01/27/2023]
|
23
|
De Gendt K, Verhoeven G, Amieux PS, Wilkinson MF. Genome-wide identification of AR-regulated genes translated in Sertoli cells in vivo using the RiboTag approach. Mol Endocrinol 2014; 28:575-91. [PMID: 24606126 DOI: 10.1210/me.2013-1391] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
An understanding of the molecular mechanisms by which androgens drive spermatogenesis has been thwarted by the fact that few consistent androgen receptor (AR) target genes have been identified. Here, we addressed this issue using next-generation sequencing coupled with the RiboTag approach, which purifies translated mRNAs expressed in cells that express cyclic recombinase (CRE). Using RiboTag mice expressing CRE in Sertoli cells (SCs), we identified genes expressed specifically in SCs in both prepubertal and adult mice. Unexpectedly, this analysis revealed that the SC-specific gene program is already largely defined at the initiation of spermatogenesis despite the subsequent dramatic maturational changes known to occur in SCs. To identify AR-regulated genes, we generated triple-mutant mice in which the SCs express the RiboTag but lack ARs. RNA sequencing analysis revealed hundreds of SC-expressed AR-regulated genes that had previously gone unnoticed, including suppressed genes involved in ovarian development. Comparison of the SC-enriched dataset with that from the whole testes allowed us to classify genes in terms of their degree of expression in SCs. This revealed that a greater fraction of AR-up-regulated genes than AR-down-regulated genes were expressed predominantly in SCs. Our results also revealed that AR signaling in SCs causes a large number of genes not detectably expressed in SCs to undergo altered expression, thereby providing genome-wide evidence for wide-scale communication between SCs and other cells. Taken together, our results identified novel classes of genes expressed in a hormone-dependent manner in different testicular cell subsets and highlight a new approach to analyze cell type-specific gene regulation.
Collapse
Affiliation(s)
- Karel De Gendt
- Department of Reproductive Medicine and Institute of Genomic Medicine (M.F.W.), University of California, La Jolla, California 92093 (K.D.G., M.F.W.); Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium (K.D.G., G.V.); and Department of Biology, Western Washington University, Bellingham, Washington 98225 (P.S.A.)
| | | | | | | |
Collapse
|
24
|
Mazón MJ, Gómez A, Yilmaz O, Carrillo M, Zanuy S. Administration of Follicle-Stimulating Hormone In Vivo Triggers Testicular Recrudescence of Juvenile European Sea Bass (Dicentrarchus labrax)1. Biol Reprod 2014; 90:6. [DOI: 10.1095/biolreprod.113.110569] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
25
|
RiboTag analysis of actively translated mRNAs in Sertoli and Leydig cells in vivo. PLoS One 2013; 8:e66179. [PMID: 23776628 PMCID: PMC3679032 DOI: 10.1371/journal.pone.0066179] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 05/02/2013] [Indexed: 01/06/2023] Open
Abstract
Male spermatogenesis is a complex biological process that is regulated by hormonal signals from the hypothalamus (GnRH), the pituitary gonadotropins (LH and FSH) and the testis (androgens, inhibin). The two key somatic cell types of the testis, Leydig and Sertoli cells, respond to gonadotropins and androgens and regulate the development and maturation of fertilization competent spermatozoa. Although progress has been made in the identification of specific transcripts that are translated in Sertoli and Leydig cells and their response to hormones, efforts to expand these studies have been restricted by technical hurdles. In order to address this problem we have applied an in vivo ribosome tagging strategy (RiboTag) that allows a detailed and physiologically relevant characterization of the "translatome" (polysome-associated mRNAs) of Leydig or Sertoli cells in vivo. Our analysis identified all previously characterized Leydig and Sertoli cell-specific markers and identified in a comprehensive manner novel markers of Leydig and Sertoli cells; the translational response of these two cell types to gonadotropins or testosterone was also investigated. Modulation of a small subset of Sertoli cell genes occurred after FSH and testosterone stimulation. However, Leydig cells responded robustly to gonadotropin deprivation and LH restoration with acute changes in polysome-associated mRNAs. These studies identified the transcription factors that are induced by LH stimulation, uncovered novel potential regulators of LH signaling and steroidogenesis, and demonstrate the effects of LH on the translational machinery in vivo in the Leydig cell.
Collapse
|
26
|
Das DS, Wadhwa N, Kunj N, Sarda K, Pradhan BS, Majumdar SS. Dickkopf homolog 3 (DKK3) plays a crucial role upstream of WNT/β-CATENIN signaling for Sertoli cell mediated regulation of spermatogenesis. PLoS One 2013; 8:e63603. [PMID: 23667645 PMCID: PMC3647036 DOI: 10.1371/journal.pone.0063603] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 04/07/2013] [Indexed: 12/20/2022] Open
Abstract
Testicular Sertoli cells (Sc) are main somatic component of seminiferous tubules that govern the differentiation of germ cells (Gc) and provide them physical support. Sc are the target of follicle stimulating hormone (FSH) and testosterone (T) which are known to regulate spermatogenesis. FSH and T levels in human and sub-human male primates remain high during infancy (4–6 months post birth), similar to those during puberty. Subsequently, juvenile phase is marked with low levels of these hormones. In spite of prolonged hormonal exposure, spermatogenesis is not discerned during infancy unlike that during puberty. Situation during infancy is similar to certain idiopathic male infertility, where prolonged hormone supplementation fails to initiate spermatogenesis. In our quest to determine non hormonal causes of idiopathic infertility which may reside within the Sc, we investigated the association between spermatogenesis and Sc specific gene(s) expressed differentially during puberty and infancy. Although products of several genes may be necessary for quantitatively normal spermatogenesis, one needs to investigate their roles one by one. Differential display and real time PCR analysis revealed higher expression of a known tumor suppressor, Dickkopf homolog 3 (DKK3), by pubertal monkey Sc as compared to infant Sc. To evaluate role of DKK3 in spermatogenesis, we generated DKK3 knock down mice (DKDM) using shRNA construct targeted to DKK3. In testis of adult DKDM, expression of DKK3 mRNA and protein were significantly (p<0.05) low and was associated with elevated WNT-4/β-CATENIN activity. Elevated β-CATENIN activity is known to restrict Sc maturation. Abundant expression of infant Sc marker, Mullerian inhibiting substance (MIS), in the testes of adult DKDM confirmed lack of Sc maturation in DKDM. Gc differentiation and fertility was severely compromised in DKDM. This is the first report of role of DKK3 in the testis and DKK3 mediated regulation of spermatogenesis via WNT-4/β-CATENIN modulation.
Collapse
Affiliation(s)
- Deepika Sharma Das
- Division of Cellular Endocrinology, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Neerja Wadhwa
- Division of Embryo Biotechnology, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Neetu Kunj
- Division of Embryo Biotechnology, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Kanchan Sarda
- Division of Cellular Endocrinology, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Bhola Shankar Pradhan
- Division of Cellular Endocrinology, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Subeer S. Majumdar
- Division of Cellular Endocrinology, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
- Division of Embryo Biotechnology, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
- * E-mail:
| |
Collapse
|
27
|
Karki R, Mariani M, Andreoli M, He S, Scambia G, Shahabi S, Ferlini C. βIII-Tubulin: biomarker of taxane resistance or drug target? Expert Opin Ther Targets 2013; 17:461-72. [PMID: 23379899 DOI: 10.1517/14728222.2013.766170] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION βIII-Tubulin (TUBB3) is predominantly expressed in neurons of the central and peripheral nervous systems, while in normal non-neoplastic tissues it is barely detectable. By contrast, this cytoskeletal protein is abundant in a wide range of tumors. βIII-Tubulin is linked to dynamic instability of microtubules (MTs), weakening the effects of agents interfering with MT polymerization. Based on this principle, early studies introduced the classical theory linking βIII-tubulin with a mechanism of counteracting taxane activity and accordingly, prompted its investigation as a predictive biomarker of taxane resistance. AREAS COVERED We reviewed 59 translational studies, including cohorts from lung, ovarian, breast, gastric, colorectal and various miscellaneous cancers subject to different chemotherapy regimens. EXPERT OPINION βIII-Tubulin functions more as a prognostic factor than as a predictor of response to chemotherapy. We believe this view can be explained by βIII-tubulin's association with prosurvival pathways in the early steps of the metastatic process. Its prognostic response increases if combined with additional biomarkers that regulate its expression, since βIII-tubulin can be expressed in conditions, such as estrogen exposure, unrelated to survival mechanisms and without any predictive activity. Additional avenues for therapeutic intervention could emerge if drugs are designed to directly target βIII-tubulin and its mechanism of regulation.
Collapse
Affiliation(s)
- Roshan Karki
- Reproductive Tumor Biology Research, Department of Obstetrics and Gynecology, Danbury Hospital, Biomedical Laboratory, Danbury, CT 06810, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Sambroni E, Rolland AD, Lareyre JJ, Le Gac F. FSH and LH have common and distinct effects on gene expression in rainbow trout testis. J Mol Endocrinol 2013; 50:1-18. [PMID: 23045716 DOI: 10.1530/jme-12-0197] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The general rules established from mammalian species for the regulation of spermatogenesis by gonadotropins may not be fully relevant in fish. Particularly, Fsh is as potent as Lh to stimulate steroidogenesis and the Fsh receptor is expressed in Leydig cells. In seasonal breeders, Fsh is likely the major gonadotropin involved in spermatogenesis onset and Lh is required to support spermatogenesis progression and gamete release. However, the genes that relay the action of Fsh and Lh have been poorly investigated in fish. The present study was aimed at identifying gonadotropin-dependent genes expressed in the testis during fish puberty. We cultured pubertal trout testicular explants for 96 h, with or without gonadotropin, and analyzed transcriptome variations using microarrays. Fsh and Lh had similar effects on a large group of genes while other genes were preferentially regulated by one or the other gonadotropin. We showed that most of the responsive genes were expressed in somatic cells and exhibited relevant patterns during the seasonal reproductive cycle. Some genes preferentially modulated by Lh could be involved in testicular cell fate (pvrl1 and bty) or sperm maturation (ehmt2 and racgap1) and will deserve further examination. Besides Fsh's effects on the steroidogenic pathway, our study demonstrates that Fsh coordinates relevant stimulatory and inhibitory paracrine factors known to regulate early germ cell proliferation and differentiation. Some of these genes belong to major regulatory pathways including the Igf pathway (igf1b/igf3 and igfbp6), the Tgfb pathway (amh, inha, inhba, and fstl3), the Wnt pathway (wisp1), and pleiotrophin (mdka).
Collapse
Affiliation(s)
- Elisabeth Sambroni
- INRA, UR1037, Laboratoire de Physiologie et Génomique des Poissons, Campus de Beaulieu, Testicular Physiology and Puberty, Biosit, Biogenouest, F-35000 Rennes, France
| | | | | | | |
Collapse
|
29
|
Migrenne S, Moreau E, Pakarinen P, Dierich A, Merlet J, Habert R, Racine C. Mouse testis development and function are differently regulated by follicle-stimulating hormone receptors signaling during fetal and prepubertal life. PLoS One 2012; 7:e53257. [PMID: 23300903 PMCID: PMC3531970 DOI: 10.1371/journal.pone.0053257] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 11/27/2012] [Indexed: 12/30/2022] Open
Abstract
It is currently admitted that Follicle-Stimulating Hormone (FSH) is physiologically involved in the development and function of fetal/neonatal Sertoli cells in the rat but not the mouse. However, FSH is produced by both species from late fetal life onwards. We thus reinvestigated the role of FSH in mouse testis development at day 0 (birth) 6, 8 and 10 post-partum (dpp) by using mice that lack functional FSH receptors (FSH-R−/−). At birth, the number and proliferative index of Sertoli cells were significantly lower in FSH-R−/− mice than in wild type neonates. Claudin 11 mRNA expression also was significantly reduced in FSH-R−/− testes at 0 and 8 dpp, whereas the mRNA levels of other Sertoli cell markers (Transferrin and Desert hedgehog) were comparable in FSH-R−/− and wild type testes. Conversely, AMH mRNA and protein levels were higher at birth, comparable at 6 dpp and then significantly lower in FSH-R−/− testes at 8–10 dpp in FSH-R−/− mice than in controls. Although the plasma concentration of LH and the number of Leydig cells were similar in FSH-R−/− and control (wild type), testosterone concentration and P450c17 mRNA expression were significantly increased in FSH-R−/− testes at birth. Conversely, at 10 dpp when adult Leydig cells appear, expression of the steroidogenic genes P450scc, P450c17 and StAR was lower in FSH-R−/− testes than in controls. In conclusion, our results show that 1) like in the rat, signaling via FSH-R controls Sertoli cell development and function during late fetal life in the mouse as well; 2) paracrine factors produced by Sertoli cells are involved in the FSH-R-dependent regulation of the functions of fetal Leydig cells in late fetal life; and 3) the role of FSH-R signaling changes during the prepubertal period.
Collapse
Affiliation(s)
- Stéphanie Migrenne
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay-aux-Roses, France
| | - Evelyne Moreau
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay-aux-Roses, France
| | - Pirjo Pakarinen
- University of Turku, Institute of Biomedicine, Department of Physiology, Turku, Finland
| | - Andrée Dierich
- CNRS 7104, IGBMC, Illkirch, France
- INSERM, U964, Illkirch , France
| | - Jorge Merlet
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay-aux-Roses, France
| | - René Habert
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay-aux-Roses, France
- * E-mail:
| | - Chrystèle Racine
- University Paris Diderot, Sorbonne Paris Cité, Laboratory of Development of the Gonads, Unit of Stem Cells and Radiation, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- INSERM, Unité 967, Fontenay-aux-Roses, France
| |
Collapse
|
30
|
Nicholls PK, Stanton PG, Chen JL, Olcorn JS, Haverfield JT, Qian H, Walton KL, Gregorevic P, Harrison CA. Activin signaling regulates Sertoli cell differentiation and function. Endocrinology 2012; 153:6065-77. [PMID: 23117933 DOI: 10.1210/en.2012-1821] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Throughout development, activin A signaling stimulates proliferation and inhibits differentiation of testicular Sertoli cells. A decline in activin levels at puberty corresponds with the differentiation of Sertoli cells that is required to sustain spermatogenesis. In this study, we consider whether terminally differentiated Sertoli cells can revert to a functionally immature phenotype in response to activin A. To increase systemic activin levels, the right tibialis anterior muscle of 7-wk-old C57BL/6J mice was transduced with an adeno-associated virus (rAAV6) expressing activin A. We show that chronic activin signaling reduces testis mass by 23.5% compared with control animals and induces a hypospermatogenic phenotype, consistent with a failure of Sertoli cells to support spermatogenesis. We use permeability tracers and transepithelial electrical resistance measurements to demonstrate that activin potently disrupts blood-testis-barrier function in adult mice and ablates tight junction formation in differentiated primary Sertoli cells, respectively. Furthermore, increased activin signaling reinitiates a program of cellular proliferation in primary Sertoli cells as determined by 5-ethynyl-2'-deoxyuridine incorporation. Proliferative cells reexpress juvenile markers, including cytokeratin-18, and suppress mature markers, including claudin-11. Thus, activin A is the first identified factor capable of reprogramming Sertoli cells to an immature, dedifferentiated phenotype. This study indicates that activin signaling must be strictly controlled in the adult in order to maintain Sertoli cell function in spermatogenesis.
Collapse
Affiliation(s)
- Peter K Nicholls
- Prince Henry's Institute of Medical Research, Clayton 3168, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
McCabe MJ, Allan CM, Foo CFH, Nicholls PK, McTavish KJ, Stanton PG. Androgen initiates Sertoli cell tight junction formation in the hypogonadal (hpg) mouse. Biol Reprod 2012; 87:38. [PMID: 22623623 DOI: 10.1095/biolreprod.111.094318] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Sertoli cell tight junctions (TJs) form at puberty as a major component of the blood-testis barrier (BTB), which is essential for spermatogenesis. This study characterized the hormonal induction of functional Sertoli cell TJ formation in vivo using the gonadotropin-deficient hypogonadal (hpg) mouse that displays prepubertal spermatogenic arrest. Androgen actions were determined in hpg mice treated for 2 or 10 days with dihydrotestosterone (DHT). Follicle-stimulating hormone (FSH) actions were studied in hpg mice expressing transgenic human FSH (hpg+tgFSH) with or without DHT treatment. TJ formation was examined by mRNA expression and immunolocalization of TJ proteins claudin-3 and claudin-11, and barrier functionality was examined by biotin tracer permeability. Immunolocalization of claudin-3 and claudin-11 was extensive at wild-type (wt) Sertoli cell TJs, which functionally excluded permeability tracer. In contrast, seminiferous tubules of hpg testes lacked claudin-3, but claudin-11 protein was present in adluminal regions of Sertoli cells. Biotin tracer permeated throughout these tubules, demonstrating dysfunctional TJs. In hpg+tgFSH testes, claudin-3 was generally absent, but claudin-11 had redistributed basally toward the TJs, where function was variable. In hpg testes, DHT treatment stimulated the redistribution of claudin-11 protein toward the basal region of Sertoli cells by Day 2, increased Cldn3 and Cldn11 mRNA expression, then induced the formation of functional TJs containing both proteins by Day 10. In hpg+tgFSH testes, TJ protein redistribution was accelerated and functional TJs formed by Day 2 of DHT treatment. We conclude that androgen stimulates initial Sertoli cell TJ formation and function in mice, whereas FSH activity is insufficient alone, but augments androgen-induced TJ function.
Collapse
Affiliation(s)
- Mark J McCabe
- Prince Henry's Institute, Monash Medical Centre, Clayton, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
32
|
Lee FY, Faivre EJ, Suzawa M, Lontok E, Ebert D, Cai F, Belsham DD, Ingraham HA. Eliminating SF-1 (NR5A1) sumoylation in vivo results in ectopic hedgehog signaling and disruption of endocrine development. Dev Cell 2011; 21:315-27. [PMID: 21820362 DOI: 10.1016/j.devcel.2011.06.028] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 03/23/2011] [Accepted: 06/24/2011] [Indexed: 11/16/2022]
Abstract
Sumoylation is generally considered a repressive mark for many transcription factors. However, the in vivo importance of sumoylation for any given substrate remains unclear and is questionable because the extent of sumoylation appears exceedingly low for most substrates. Here, we permanently eliminated SF-1/NR5A1 sumoylation in mice (Sf-1(K119R, K194R, or 2KR)) and found that Sf-1(2KR/2KR) mice failed to phenocopy a simple gain of SF-1 function or show elevated levels of well-established SF-1 target genes. Instead, mutant mice exhibited marked endocrine abnormalities and changes in cell fate that reflected an inappropriate activation of hedgehog signaling and other potential SUMO-sensitive targets. Furthermore, unsumoylatable SF-1 mutants activated Shh and exhibited preferential recruitment to Shh genomic elements in cells. We conclude that the sumoylation cycle greatly expands the functional capacity of transcription factors such as SF-1 and is leveraged during development to achieve cell-type-specific gene expression in multicellular organisms.
Collapse
Affiliation(s)
- Florence Y Lee
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Balasinor NH, D'Souza R, Nanaware P, Idicula-Thomas S, Kedia-Mokashi N, He Z, Dym M. Effect of high intratesticular estrogen on global gene expression and testicular cell number in rats. Reprod Biol Endocrinol 2010; 8:72. [PMID: 20573204 PMCID: PMC2906496 DOI: 10.1186/1477-7827-8-72] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Accepted: 06/23/2010] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND The identification of estrogen receptors alpha and beta and aromatase in the testis has highlighted the important role of estrogens in regulating spermatogenesis. There is a wealth of information on the deleterious effects of fetal and neonatal exposure of estrogens and xenoestrogens in the testis, including spermiation failure and germ cell apoptosis. However, very little is known about gene transcripts affected by exogenous estradiol exposure in the testis. The objective of the present study was to unveil global gene expression profiles and testicular cell number changes in rats after estradiol treatment. METHODS 17beta-estradiol was administered to adult male rats at a dose of 100 micrograms/kg body weight in saline daily for 10 days; male rats receiving only saline were used as controls. Microarray analysis was performed to examine global gene expression profiles with or without estradiol treatment. Real time RT-PCR was conducted to verify the microarray data. In silico promoter and estrogen responsive elements (EREs) analysis was carried out for the differentially expressed genes in response to estradiol. Quantitation of testicular cell number based on ploidy was also performed using flow cytometry in rats with or without estradiol treatment. RESULTS We found that 221 genes and expressed sequence tags (ESTs) were differentially expressed in rat testes treated with estradiol compared to the control; the microarray data were confirmed by real time RT-PCR. Gene Ontology analysis revealed that a number of the differentially expressed genes are involved in androgen and xenobiotic metabolism, maintenance of cell cytoskeleton, endocytosis, and germ cell apoptosis. A total of 33 up-regulated genes and 67 down-regulated genes showed the presence of EREs. Flow cytometry showed that estradiol induced a significant decrease in 2n cells (somatic and germ cells) and 4n cells (pachytene spermatocytes) and a marked increase in the number of elongated and elongating spermatids. CONCLUSIONS This study provides a novel insight into the molecular basis for spermiation failure and apoptosis caused by 17beta-estradiol and it also offers new mechanisms by which adult exposure to environmental estrogens can affect spermatogenesis and fertility.
Collapse
Affiliation(s)
- Nafisa H Balasinor
- Neuroendocrinology Division, National Institute for Research in Reproductive Health, Parel, Mumbai, India
| | - Ryan D'Souza
- Neuroendocrinology Division, National Institute for Research in Reproductive Health, Parel, Mumbai, India
| | - Padma Nanaware
- Biomedical Informatics Centre of ICMR, National Institute for Research in Reproductive Health, Parel, Mumbai, India
| | - Susan Idicula-Thomas
- Biomedical Informatics Centre of ICMR, National Institute for Research in Reproductive Health, Parel, Mumbai, India
| | - Neelam Kedia-Mokashi
- Neuroendocrinology Division, National Institute for Research in Reproductive Health, Parel, Mumbai, India
| | - Zuping He
- Georgetown University Medical Center, Department of Biochemistry and Molecular & Cellular Biology, 3900 Reservoir Road, NW, Washington, DC 20057, USA
| | - Martin Dym
- Georgetown University Medical Center, Department of Biochemistry and Molecular & Cellular Biology, 3900 Reservoir Road, NW, Washington, DC 20057, USA
| |
Collapse
|
34
|
Allan CM, Couse JF, Simanainen U, Spaliviero J, Jimenez M, Rodriguez K, Korach KS, Handelsman DJ. Estradiol induction of spermatogenesis is mediated via an estrogen receptor-{alpha} mechanism involving neuroendocrine activation of follicle-stimulating hormone secretion. Endocrinology 2010; 151:2800-10. [PMID: 20410197 PMCID: PMC2875821 DOI: 10.1210/en.2009-1477] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Both testosterone and its nonaromatizable metabolite dihydrotestosterone (DHT) induce spermatogenesis in gonadotropin-deficient hpg mice. Surprisingly, because aromatization is not required, estradiol (E2) also induces spermatogenesis and increases circulating FSH in hpg mice, but the mechanism remains unclear. We studied E2-induced spermatogenesis in hpg mice on an estrogen receptor (ER)-alpha (hpg/alphaERKO) or ERbeta (hpg/betaERKO) knockout or wild-type ER (hpg/WT) background treated with subdermal E2 or DHT implants for 6 wk. In hpg/WT and hpg/betaERKO, but not hpg/alphaERKO mice, E2 increased testis and epididymal weight, whereas DHT-induced increases were unaffected by ERalpha or ERbeta inactivation. E2 but not DHT treatment increased serum FSH (but not LH) in hpg/WT and hpg/betaERKO but not hpg/alphaERKO hpg mice. DHT or E2 alone increased (premeiotic) spermatogonia and (meiotic) spermatocytes without significant change in Sertoli cell numbers. DHT alone increased postmeiotic spermatids, regardless of ER presence, compared with variable ERalpha-dependent E2 postmeiotic responses. An ERalpha-mediated effect was confirmed by treating hpg mice for 6 wk by subdermal selective ER-alpha (16alpha-LE(2)) or ERbeta (8beta-VE(2)) agonist implants. ERalpha (but not ERbeta) agonist increased testis and epididymal weight, Sertoli cell, spermatogonia, meiotic, and postmeiotic germ cell numbers. Only ERalpha agonist markedly increased serum FSH, whereas either agonist induced small rises in serum LH. Administration of ERalpha agonist or E2 in the presence of functional ERalpha induced prominent gene expression of specific Sertoli (Eppin, Rhox5) and Leydig cell (Cyp11a1, Hsd3b1) markers. We conclude that E2-induced spermatogenesis in hpg mice involves an ERalpha-dependent neuroendocrine mechanism increasing blood FSH and Sertoli cell function.
Collapse
Affiliation(s)
- Charles M Allan
- ANZAC Research Institute, Sydney, New South Wales 2139, Australia
| | | | | | | | | | | | | | | |
Collapse
|
35
|
McCabe MJ, Tarulli GA, Meachem SJ, Robertson DM, Smooker PM, Stanton PG. Gonadotropins regulate rat testicular tight junctions in vivo. Endocrinology 2010; 151:2911-22. [PMID: 20357222 PMCID: PMC2875820 DOI: 10.1210/en.2009-1278] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Sertoli cell tight junctions (TJs) are an essential component of the blood-testis barrier required for spermatogenesis; however, the role of gonadotropins in their maintenance is unknown. This study aimed to investigate the effect of gonadotropin suppression and short-term replacement on TJ function and TJ protein (occludin and claudin-11) expression and localization, in an adult rat model in vivo. Rats (n = 10/group) received the GnRH antagonist, acyline, for 7 wk to suppress gonadotropins. Three groups then received for 7 d: 1) human recombinant FSH, 2) human chorionic gonadotropin (hCG) and rat FSH antibody (to study testicular androgen stimulation alone), and 3) hCG alone (to study testicular androgen and pituitary FSH production). TJ proteins were assessed by real-time PCR, Western blot analysis, and immunohistochemistry, whereas TJ function was assessed with a biotin permeation tracer. Acyline treatment significantly reduced testis weights, serum androgens, LH and FSH, and adluminal germ cells (pachytene spermatocyte, round and elongating spermatids). In contrast to controls, acyline induced seminiferous tubule permeability to biotin, loss of tubule lumens, and loss of occludin, but redistribution of claudin-11, immunostaining. Short-term hormone replacement stimulated significant recoveries in adluminal germ cell numbers. In hCG +/- FSH antibody-treated rats, occludin and claudin-11 protein relocalized at the TJ, but such relocalization was minimal with FSH alone. Tubule lumens also reappeared, but most tubules remained permeable to biotin tracer, despite the presence of occludin. It is concluded that gonadotropins maintain Sertoli cell TJs in the adult rat via a mechanism that includes the localization of occludin and claudin-11 at functional TJs.
Collapse
Affiliation(s)
- Mark J McCabe
- Prince Henry's Institute of Medical Research, P.O. Box 5152, Clayton 3168, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
36
|
Griffin D, Ellis P, Dunmore B, Bauer J, Abel M, Affara N. Transcriptional profiling of luteinizing hormone receptor-deficient mice before and after testosterone treatment provides insight into the hormonal control of postnatal testicular development and Leydig cell differentiation. Biol Reprod 2010; 82:1139-50. [PMID: 20164437 PMCID: PMC2874499 DOI: 10.1095/biolreprod.109.082099] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 11/12/2009] [Accepted: 01/15/2010] [Indexed: 01/07/2023] Open
Abstract
Luteinizing hormone (LH) is a key regulator of male fertility through its effects on testosterone secretion by Leydig cells. Transcriptional control of this is, however, currently poorly understood. Mice in which the LH receptor is knocked out (LuRKO) show reduced testicular size, reduced testosterone, elevated serum LH, and a spermatogenic arrest that can be rescued by the administration of testosterone. Using genome-wide transcription profiling of LuRKO and control testes during postnatal development and following testosterone treatment, we show that the transcriptional effects of LH insensitivity are biphasic, with an early testosterone-independent phase and a subsequent testosterone-dependent phase. Testosterone rescue re-enables the second, testosterone-dependent phase of the normal prepubertal transcription program and permits the continuation of spermatogenesis. Examination of the earliest responses to testosterone highlights six genes that respond rapidly in a dose-dependent fashion to the androgen and that are therefore candidate regulatory genes associated with the testosterone-driven progression of spermatogenesis. In addition, our transcriptional data suggest a model for the replacement of fetal-type Leydig cells by adult-type cells during testicular development in which a testosterone feedback switch is necessary for adult Leydig cell production. LH signaling affects the timing of the switch but is not a strict requirement for Leydig cell differentiation.
Collapse
Affiliation(s)
- D.K. Griffin
- Department of Biosciences, University of Kent, Canterbury, United Kingdom
| | - P.J. Ellis
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - B. Dunmore
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - J. Bauer
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - M.H. Abel
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, United Kingdom
| | - N.A. Affara
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
37
|
O'Shaughnessy PJ, Monteiro A, Verhoeven G, De Gendt K, Abel MH. Effect of FSH on testicular morphology and spermatogenesis in gonadotrophin-deficient hypogonadal mice lacking androgen receptors. Reproduction 2010; 139:177-84. [PMID: 19846485 PMCID: PMC2791495 DOI: 10.1530/rep-09-0377] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
FSH and androgen act to stimulate and maintain spermatogenesis. FSH acts directly on the Sertoli cells to stimulate germ cell number and acts indirectly to increase androgen production by the Leydig cells. In order to differentiate between the direct effects of FSH on spermatogenesis and those mediated indirectly through androgen action, we have crossed hypogonadal (hpg) mice, which lack gonadotrophins, with mice lacking androgen receptors (AR) either ubiquitously (ARKO) or specifically on the Sertoli cells (SCARKO). These hpg.ARKO and hpg.SCARKO mice were treated with recombinant FSH for 7 days and testicular morphology and cell numbers were assessed. In untreated hpg and hpg.SCARKO mice, germ cell development was limited and did not progress beyond the pachytene stage. In hpg.ARKO mice, testes were smaller with fewer Sertoli cells and germ cells compared to hpg mice. Treatment with FSH had no effect on Sertoli cell number but significantly increased germ cell numbers in all groups. In hpg mice, FSH increased the numbers of spermatogonia and spermatocytes, and induced round spermatid formation. In hpg.SCARKO and hpg.ARKO mice, in contrast, only spermatogonial and spermatocyte numbers were increased with no formation of spermatids. Leydig cell numbers were increased by FSH in hpg and hpg.SCARKO mice but not in hpg.ARKO mice. Results show that in rodents 1) FSH acts to stimulate spermatogenesis through an increase in spermatogonial number and subsequent entry of these cells into meiosis, 2) FSH has no direct effect on the completion of meiosis and 3) FSH effects on Leydig cell number are mediated through interstitial ARs.
Collapse
Affiliation(s)
- P J O'Shaughnessy
- Division of Cell Sciences, University of Glasgow Veterinary School, Bearsden Road, Glasgow G61 1QH, UK.
| | | | | | | | | |
Collapse
|