1
|
Giudice MG, Kanbar M, Poels J, Duquenne A, Wyns C. Long-term culture of human Sertoli cells from adult Klinefelter patients as a first step to develop new tools for unravelling the testicular physiopathology. Hum Reprod 2024; 39:2400-2410. [PMID: 39237101 DOI: 10.1093/humrep/deae201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/29/2024] [Indexed: 09/07/2024] Open
Abstract
STUDY QUESTION Are Sertoli cells (SCs) from adult Klinefelter men (47,XXY) capable of proliferating in vitro and maintaining their main phenotypical and functional characteristics as do SCs from adult 46,XY patients? SUMMARY ANSWER Isolated SCs from patients with Klinefelter syndrome (KS) can be expanded in vitro while maintaining their characteristics and a stable karyotype, similar to SCs from 46,XY patients. WHAT IS KNOWN ALREADY The mechanism leading to testicular tissue degeneration in KS is still unknown. A few recent studies highlight the main role played by SCs in the physiopathology of the disease, but new study models based on co-culture or testicular organoids are needed to further understand the SC's involvement in the mechanism of testicular degeneration and fibrosis, and to find therapeutical targets. KS SC expansion could be the first step towards developing such in vitro study models. SCs have been isolated from 46,XY men and expanded in vitro while maintaining the expression of phenotypical and functional markers, but propagation of SCs from KS men has not been achieved yet. STUDY DESIGN, SIZE, DURATION Testicular tissue was obtained during a testicular sperm extraction procedure for infertility treatment between 2019 and 2021 from three azoospermic adult KS (47,XXY) men (33±3.6 years old) and from three control patients (46,XY) (36±2 years old) presenting with obstructive azoospermia. SCs isolated from frozen-thawed tissue of KS and 46,XY patients were cultured for 60 days and compared. All patients signed an informed consent according to the ethical board approval of the study protocol. PARTICIPANTS/MATERIALS, SETTING, METHODS Testicular biopsies obtained from KS (n = 3) and 46,XY (n = 3) adult patients were slow-frozen. After tissue thawing SCs were isolated using a double-step enzymatic digestion and differential plating, and cultured for 60 days in DMEM medium containing FBS. Analyses were performed at different culture times (passages 5 (P5) and 10 (P10)). Quantification of cells using immunofluorescence (IF) for cell type-specific markers (Sox9, GATA4, ACTA2, INSL3, MAGEA4), SCs characterization using both IF and quantitative real-time PCR for GDNF, BMP4, AR and CLDN11 and cells karyotyping were performed. MAIN RESULTS AND THE ROLE OF CHANCE We demonstrate for the first time that a small population of human SCs isolated from frozen-thawed testis of adult KS patients can be expanded in vitro while retaining expression of characteristic markers of SCs and the 47,XXY karyotype, and exhibiting cell-specific functional proteins and gene expression (GDNF, BMP4, AR, and CLDN11) after 60 days in culture. At P10, 83.39 ± 4.2% of cultured cells from KS men and 85.34 ± 4.1% from 46,XY men expressed Sox9, and 88.8 ± 3.9% of KS cells versus 82.9 ± 3.2% of the control cells were positive for GATA4 without any differences between two groups; both Sox9 and GATA4 are typical SC markers. No differences were found between KS and 46,XY SCs in vitro in terms of cells expansion (exponential growth between P1 and P10 with an average cell count of 2.8±1.5×107 versus 3.8±1.2×107 respectively for the KS and control groups at P10). There was no significant statistical difference for functional proteins and genes expressions (GDNF, BMP4, AR, and CLDN11) neither between KS SCs and control SCs nor between P5 and P10. LIMITATIONS, REASONS FOR CAUTION The small number of donor samples is a limitation but it is due to limited availability of tissue for research in KS populations. Although no differences were observed in SCs function in the culture of isolated SCs after 60 days, the possibility of a SCs dysfunction needs to be investigated in more complex 3-dimensional models allowing the establishment of a proper cell organization and further analyses of cell functions and interactions during longer culture periods. WIDER IMPLICATIONS OF THE FINDINGS The demonstration of the possibility to propagate KS SCs in vitro could be useful to build new in vitro models for deciphering testicular cell interactions, determining deficient signalling pathways involved in impaired spermatogenesis, and identifying targets for infertility treatment in KS. As the cell numbers achieved in this study are higher than cell numbers used to develop testicular organoids, we may expect to be able to understand the behaviour and physiopathology of SCs in the disease during the long-term culture of these organoids. Such models could be further applied to understand other causes of deficiencies in seminiferous tubules. STUDY FUNDING/COMPETING INTEREST(S) M.G.G is funded by a grant from the Cliniques Universitaires Saint-Luc (FRC) for the research project on Klinefelter Syndrome Physiopathology. The authors declare no conflicts of interest. TRIAL REGISTRATION NUMBER NCT05997706.
Collapse
Affiliation(s)
- Maria Grazia Giudice
- Pôle de recherche en Physiologie de la Reproduction (REPR), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Pôle de recherche en Physiologie de la Reproduction (REPR), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jonathan Poels
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Armelle Duquenne
- Center for Human Genetic, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christine Wyns
- Pôle de recherche en Physiologie de la Reproduction (REPR), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
2
|
Hau RK, Wright SH, Cherrington NJ. Drug Transporters at the Human Blood-Testis Barrier. Drug Metab Dispos 2023; 51:560-571. [PMID: 36732077 PMCID: PMC10158500 DOI: 10.1124/dmd.122.001186] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/06/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Transporters are involved in the movement of many physiologically important molecules across cell membranes and have a substantial impact on the pharmacological and toxicological effect of xenobiotics. Many transporters have been studied in the context of disposition to, or toxicity in, organs such as the kidney and liver; however, transporters in the testes are increasingly gaining recognition for their role in drug transport across the blood-testis barrier (BTB). The BTB is an epithelial membrane barrier formed by adjacent Sertoli cells (SCs) in the seminiferous tubules that form intercellular junctional complexes to protect developing germ cells from the external environment. Consequently, many charged or large polar molecules cannot cross this barrier without assistance from a transporter. SCs express a variety of drug uptake and efflux transporters to control the flux of endogenous and exogenous molecules across the BTB. Recent studies have identified several transport pathways in SCs that allow certain drugs to circumvent the human BTB. These pathways may exist in other species, such as rodents and nonhuman primates; however, there is (1) a lack of information on their expression and/or localization in these species, and (2) conflicting reports on localization of some transporters that have been evaluated in rodents compared with humans. This review outlines the current knowledge on the expression and localization of pharmacologically relevant drug transporters in human testes and calls attention to the insufficient and contradictory understanding of testicular transporters in other species that are commonly used in drug disposition and toxicity studies. SIGNIFICANCE STATEMENT: While the expression, localization, and function of many xenobiotic transporters have been studied in organs such as the kidney and liver, the characterization of transporters in the testes is scarce. This review summarizes the expression and localization of common pharmacologically-relevant transporters in human testes that have significant implications for the development of drugs that can cross the blood-testis barrier. Potential expression differences between humans and rodents highlighted here suggest rodents may be inappropriate for some testicular disposition and toxicity studies.
Collapse
Affiliation(s)
- Raymond K Hau
- College of Pharmacy, Department of Pharmacology & Toxicology (R.K.H., N.J.C.) and College of Medicine, Department of Physiology (S.H.W.), The University of Arizona, Tucson, Arizona
| | - Stephen H Wright
- College of Pharmacy, Department of Pharmacology & Toxicology (R.K.H., N.J.C.) and College of Medicine, Department of Physiology (S.H.W.), The University of Arizona, Tucson, Arizona
| | - Nathan J Cherrington
- College of Pharmacy, Department of Pharmacology & Toxicology (R.K.H., N.J.C.) and College of Medicine, Department of Physiology (S.H.W.), The University of Arizona, Tucson, Arizona
| |
Collapse
|
3
|
Martinez-Fabregas J, Tamargo-Azpilicueta J, Diaz-Moreno I. Lysosomes: Multifunctional compartments ruled by a complex regulatory network. FEBS Open Bio 2022; 12:758-774. [PMID: 35218162 PMCID: PMC8972048 DOI: 10.1002/2211-5463.13387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/08/2022] [Accepted: 02/24/2022] [Indexed: 11/23/2022] Open
Abstract
More than 50 years have passed since Nobel laureate Cristian de Duve described for the first time the presence of tiny subcellular compartments filled with hydrolytic enzymes: the lysosome. For a long time, lysosomes were deemed simple waste bags exerting a plethora of hydrolytic activities involved in the recycling of biopolymers, and lysosomal genes were considered to just be simple housekeeping genes, transcribed in a constitutive fashion. However, lysosomes are emerging as multifunctional signalling hubs involved in multiple aspects of cell biology, both under homeostatic and pathological conditions. Lysosomes are involved in the regulation of cell metabolism through the mTOR/TFEB axis. They are also key players in the regulation and onset of the immune response. Furthermore, it is becoming clear that lysosomal hydrolases can regulate several biological processes outside of the lysosome. They are also implicated in a complex communication network among subcellular compartments that involves intimate organelle‐to‐organelle contacts. Furthermore, lysosomal dysfunction is nowadays accepted as the causative event behind several human pathologies: low frequency inherited diseases, cancer, or neurodegenerative, metabolic, inflammatory, and autoimmune diseases. Recent advances in our knowledge of the complex biology of lysosomes have established them as promising therapeutic targets for the treatment of different pathologies. Although recent discoveries have started to highlight that lysosomes are controlled by a complex web of regulatory networks, which in some cases seem to be cell‐ and stimuli‐dependent, to harness the full potential of lysosomes as therapeutic targets, we need a deeper understanding of the little‐known signalling pathways regulating this subcellular compartment and its functions.
Collapse
Affiliation(s)
- Jonathan Martinez-Fabregas
- Instituto de Investigaciones Químicas (IIQ) - Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla - CSIC, Avda. Américo Vespucio 49, 41092, Sevilla, Spain
| | - Joaquin Tamargo-Azpilicueta
- Instituto de Investigaciones Químicas (IIQ) - Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla - CSIC, Avda. Américo Vespucio 49, 41092, Sevilla, Spain
| | - Irene Diaz-Moreno
- Instituto de Investigaciones Químicas (IIQ) - Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla - CSIC, Avda. Américo Vespucio 49, 41092, Sevilla, Spain
| |
Collapse
|
4
|
Moreira BP, Silva AM, Martins AD, Monteiro MP, Sousa M, Oliveira PF, Alves MG. Effect of Leptin in Human Sertoli Cells Mitochondrial Physiology. Reprod Sci 2020; 28:920-931. [DOI: 10.1007/s43032-020-00328-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/20/2020] [Indexed: 12/12/2022]
|
5
|
Carrageta DF, Bernardino RL, Alves MG, Oliveira PF. CFTR regulation of aquaporin-mediated water transport. VITAMINS AND HORMONES 2020; 112:163-177. [PMID: 32061340 DOI: 10.1016/bs.vh.2019.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is an anion channel responsible for the direct transport of bicarbonate and chloride. CFTR-dependent ionic transport is crucial for pH regulation and fluid homeodynamics among epithelial surfaces. Particularly, CFTR performs an essential role in the male reproductive tract, which requires a tight regulation of water and electrolytes in order to produce healthy spermatozoa. The absence or malfunction of CFTR results in cystic fibrosis, the most common lethal disease among Caucasians, that is characterized by an impaired fluid and ionic homeostasis in the whole organism. Due to the wide expression and importance of CFTR, the male reproductive tract is highly affected by cystic fibrosis, resulting in male infertility. Although CFTR is not permeable to water, this protein acts as a regulator of other protein channels, such as aquaporins. In fact, CFTR acts as a molecular partner of aquaporins in epithelial cells, regulating fluid homeodynamics. Herein, up-to-date data concerning the regulation of aquaporin-mediated water transport by CFTR will be discussed, highlighting the role of both channels in the male reproductive tract.
Collapse
Affiliation(s)
- David F Carrageta
- Department of Microscopy, Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Raquel L Bernardino
- Department of Microscopy, Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Marco G Alves
- Department of Microscopy, Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Pedro F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal; i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.
| |
Collapse
|
6
|
Moreira BP, Silva JF, Jarak I, de Lourdes Pereira M, Oliveira PF, Alves MG. Technical-grade chlordane compromises rat Sertoli cells proliferation, viability and metabolic activity. Toxicol In Vitro 2019; 63:104673. [PMID: 31704469 DOI: 10.1016/j.tiv.2019.104673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 01/08/2023]
Abstract
Environmental contaminants are a daily presence in human routine. Multiple studies highlight the obesogenic activity of some chemicals. Moreover, these compounds have been suggested as a cause of male subfertility and/or infertility. Technical-grade chlordane (TGC) is classified as an endocrine-disruptor chemical, while its classification as obesogen is controversial. Herein, we studied the influence of TGC on Sertoli cells (SCs) metabolism. Rat Sertoli cells (rSCs) were cultured without and in the presence of increasing concentrations (1, 10 and 1000 nM) of TGC. The viability, proliferation, metabolic activity and the metabolic profile of rSCs was assessed. Expression of key glycolysis-related enzymes, transporters and biomarkers of oxidative damage were also evaluated. Our results show that exposure to higher concentrations of TGC decreases SCs proliferation and viability, which was accompanied by increased glucose consumption associated with an upregulation of Glut3 levels. As a result, pyruvate/lactate production were enhanced thus increasing the glycolytic flux in cells exposed to 1000 nM TGC, although lactate dehydrogenase expression and activity did not increase. Notably, biomarkers associated with oxidative damage remained unchanged after exposure to TGC. This is the first report showing that TGC alters glucose rSCs metabolism and the nutritional support of spermatogenesis with consequences for male fertility.
Collapse
Affiliation(s)
- Bruno P Moreira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Juliana F Silva
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Ivana Jarak
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Maria de Lourdes Pereira
- Department of Medical Sciences & CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Pedro F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Department of Genetics, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Marco G Alves
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
7
|
Bernardino RL, Dias TR, Moreira BP, Cunha M, Barros A, Oliveira E, Sousa M, Alves MG, Oliveira PF. Carbonic anhydrases are involved in mitochondrial biogenesis and control the production of lactate by human Sertoli cells. FEBS J 2019; 286:1393-1406. [PMID: 30724485 DOI: 10.1111/febs.14779] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 01/04/2019] [Accepted: 02/04/2019] [Indexed: 01/04/2023]
Abstract
The process that allows cells to control their pH and bicarbonate levels is essential for ionic and metabolic equilibrium. Carbonic anhydrases (CAs) catalyse the conversion of CO2 to HCO 3 - and H+ and are thus essential for this process. Herein, we inhibited CAs with acetazolamide - ACT and SLC-0111 - to study their involvement in the metabolism, mitochondrial potential, mitochondrial biogenesis and lipid metabolism of human Sertoli cells (hSCs), obtained from biopsies from men with conserved spermatogenesis. We were able to identify three isoforms of CAs, one mitochondrial isoform (CA VB) and two cell membrane-bound isoforms (CA IX and CA XII) in hSCs. When assessing the expression of markers for mitochondrial biogenesis, we observed a decrease in HIF-1α, SIRT1, PGC1α and NRF-1 mRNAs after all CAs were inhibited, resulting in decreased mitochondrial DNA copy numbers. This was followed by an increased production of lactate and alanine in the same conditions. In addition, consumption of glucose was maintained after inhibition of all CAs in hSCs. These results indicate a reduced conversion of pyruvate to acetyl-coA, possibly due to decreased mitochondrial function, caused by CA inhibition in hSCs. Inhibition of CAs also caused alterations in lipid metabolism, since we detected an increased expression of hormone-sensitive lipase (HSL) in hSCs. Our results suggest that CAs are essential for mitochondrial biogenesis, glucose and lipid metabolism in hSCs. This is the first report showing that CAs play an essential role in hSC metabolic dynamics, being involved in mitochondrial biogenesis and controlling lactate production.
Collapse
Affiliation(s)
- Raquel L Bernardino
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences, Abel Salazar (ICBAS) and Multidisciplinary Unit for Biomedical Research (UMIB), University of Porto, Portugal
| | - Tânia R Dias
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences, Abel Salazar (ICBAS) and Multidisciplinary Unit for Biomedical Research (UMIB), University of Porto, Portugal.,University of Beira Interior, Covilhã, Portugal.,LAQV/REQUINTE - Laboratory of Bromatology and Hydrology, Faculty of Pharmacy, University of Porto, Portugal
| | - Bruno P Moreira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences, Abel Salazar (ICBAS) and Multidisciplinary Unit for Biomedical Research (UMIB), University of Porto, Portugal
| | - Mariana Cunha
- Centre for Reproductive Genetics Prof. Alberto Barros, Porto, Portugal
| | - Alberto Barros
- Centre for Reproductive Genetics Prof. Alberto Barros, Porto, Portugal.,Department of Genetics, Faculty of Medicine, University of Porto, Portugal.,i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Elsa Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences, Abel Salazar (ICBAS) and Multidisciplinary Unit for Biomedical Research (UMIB), University of Porto, Portugal
| | - Mário Sousa
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences, Abel Salazar (ICBAS) and Multidisciplinary Unit for Biomedical Research (UMIB), University of Porto, Portugal.,Centre for Reproductive Genetics Prof. Alberto Barros, Porto, Portugal
| | - Marco G Alves
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences, Abel Salazar (ICBAS) and Multidisciplinary Unit for Biomedical Research (UMIB), University of Porto, Portugal
| | - Pedro F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences, Abel Salazar (ICBAS) and Multidisciplinary Unit for Biomedical Research (UMIB), University of Porto, Portugal.,Department of Genetics, Faculty of Medicine, University of Porto, Portugal.,i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| |
Collapse
|
8
|
Martins AD, Monteiro MP, Silva BM, Barros A, Sousa M, Carvalho RA, Oliveira PF, Alves MG. Metabolic dynamics of human Sertoli cells are differentially modulated by physiological and pharmacological concentrations of GLP-1. Toxicol Appl Pharmacol 2019; 362:1-8. [DOI: 10.1016/j.taap.2018.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 10/01/2018] [Accepted: 10/05/2018] [Indexed: 01/30/2023]
|
9
|
Yu CL, Guan JY, Ding J, Huang S, Lian Y, Luo HY, Wang XZ. AMP-activated protein kinase negatively regulates heat treatment-induced lactate secretion in cultured boar sertoli cells. Theriogenology 2018; 121:35-41. [DOI: 10.1016/j.theriogenology.2018.07.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 07/30/2018] [Accepted: 07/30/2018] [Indexed: 12/16/2022]
|
10
|
Martin-Hidalgo D, Hurtado de Llera A, Calle-Guisado V, Gonzalez-Fernandez L, Garcia-Marin L, Bragado MJ. AMPK Function in Mammalian Spermatozoa. Int J Mol Sci 2018; 19:ijms19113293. [PMID: 30360525 PMCID: PMC6275045 DOI: 10.3390/ijms19113293] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 01/03/2023] Open
Abstract
AMP-activated protein kinase AMPK regulates cellular energy by controlling metabolism through the inhibition of anabolic pathways and the simultaneous stimulation of catabolic pathways. Given its central regulator role in cell metabolism, AMPK activity and its regulation have been the focus of relevant investigations, although only a few studies have focused on the AMPK function in the control of spermatozoa's ability to fertilize. This review summarizes the known cellular roles of AMPK that have been identified in mammalian spermatozoa. The involvement of AMPK activity is described in terms of the main physiological functions of mature spermatozoa, particularly in the regulation of suitable sperm motility adapted to the fluctuating extracellular medium, maintenance of the integrity of sperm membranes, and the mitochondrial membrane potential. In addition, the intracellular signaling pathways leading to AMPK activation in mammalian spermatozoa are reviewed. We also discuss the role of AMPK in assisted reproduction techniques, particularly during semen cryopreservation and preservation (at 17 °C). Finally, we reinforce the idea of AMPK as a key signaling kinase in spermatozoa that acts as an essential linker/bridge between metabolism energy and sperm's ability to fertilize.
Collapse
Affiliation(s)
- David Martin-Hidalgo
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory of Cell Biology, Department of Microscopy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 40050-313 Porto, Portugal.
| | - Ana Hurtado de Llera
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
- Hormones and Metabolism Research Group, Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Violeta Calle-Guisado
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
| | - Lauro Gonzalez-Fernandez
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
| | - Luis Garcia-Marin
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
| | - M Julia Bragado
- Research Group of Intracellular Signaling and Technology of Reproduction (SINTREP), Institute of Biotechnology in Agriculture and Livestock (INBIO G+C), University of Extremadura, 10003 Cáceres, Spain.
| |
Collapse
|
11
|
Rajamanickam GD, Kastelic JP, Thundathil JC. The ubiquitous isoform of Na/K-ATPase (ATP1A1) regulates junctional proteins, connexin 43 and claudin 11 via Src-EGFR-ERK1/2-CREB pathway in rat Sertoli cells. Biol Reprod 2018; 96:456-468. [PMID: 28203706 DOI: 10.1095/biolreprod.116.141267] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 11/10/2016] [Accepted: 12/13/2016] [Indexed: 01/03/2023] Open
Abstract
Interaction of Na/K-ATPase with its ligand ouabain has been implicated in the regulation of various biological processes. The objective was to investigate roles of Na/K-ATPase isoforms in formation and function of junctional complexes in Sertoli cells. Primary cultures of Sertoli cells were obtained by enzymatic digestion of 20-day-old rat testes and grown on Matrigel-coated dishes for 7 days. Sertoli cells predominantly expressed the ubiquitous isoform of Na/K-ATPase (ATP1A1), confirmed by immunoblotting, PCR, immunofluorescence, and mass spectrometry. Treatment of Sertoli cells with 50 nM ouabain increased transepithelial electrical resistance (TER) and expression of claudin 11 (tight junctions) and connexin 43 (gap junctions), whereas 1 mM ouabain had opposite effects. Involvement of Src-EGFR-ERK1/2-CREB pathway in ouabain-mediated expression of claudin 11 and connexin 43 was evaluated. Incubation of Sertoli cells with 50 nM ouabain increased content of p-Src, p-EGFR, p-ERK1/2, and p-CREB; in contrast, 1 mM ouabain decreased phosphorylation of these signaling molecules. Preincubation of Sertoli cells with inhibitors of Src and MAPK pathways inhibited ouabain-induced effects on these signaling molecules, TER, and expression of claudin 11 and connexin 43. In conclusion, we inferred that ATP1A1 regulated Sertoli cell tight junctions and gap junctions through the Src-EGFR-ERK1/2-CREB pathway. Ouabain is an endogenous steroid; therefore, its interaction with ATP1A1 may be a critical signaling mechanism for the regulation of Sertoli cell function and male fertility.
Collapse
Affiliation(s)
- Gayathri D Rajamanickam
- Department of Production Animal Health, Faculty of Veterinary Medicine, Heritage Medical Research Building RM 400, 3330 Hospital Drive NW, University of Calgary, Calgary, AB, Canada
| | - John P Kastelic
- Department of Production Animal Health, Faculty of Veterinary Medicine, Heritage Medical Research Building RM 400, 3330 Hospital Drive NW, University of Calgary, Calgary, AB, Canada
| | - Jacob C Thundathil
- Department of Production Animal Health, Faculty of Veterinary Medicine, Heritage Medical Research Building RM 400, 3330 Hospital Drive NW, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
12
|
Vogl W, Lyon K, Adams A, Piva M, Nassour V. The endoplasmic reticulum, calcium signaling and junction turnover in Sertoli cells. Reproduction 2017; 155:R93-R104. [PMID: 29066527 DOI: 10.1530/rep-17-0281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/12/2017] [Accepted: 10/23/2017] [Indexed: 12/19/2022]
Abstract
The endoplasmic reticulum (ER) forms a continuous network throughout morphologically differentiated Sertoli cells. It is an integral component of intercellular adhesion junctions in this cell type, as well as forming membrane contact sites with the plasma membrane and intracellular organelles. One of the major functions of the ER in cells generally is maintaining calcium homeostasis and generating calcium signals. In this review, we discuss what is currently known about the overall pattern of distribution of the ER in Sertoli cells and the location of calcium regulatory machinery in the various subdomains of the organelle. Current data are consistent with the hypothesis that calcium signaling by the ER of Sertoli cells may play a significant role in events related to junction remodeling that occur in the seminiferous epithelium during spermatogenesis.
Collapse
Affiliation(s)
- Wayne Vogl
- Department of Cellular and Physiological SciencesUniversity of British Columbia, British Columbia, Canada .,Department of Obstetrics and GynaecologyUniversity of British Columbia, British Columbia, Canada
| | - Kevin Lyon
- Department of Obstetrics and GynaecologyUniversity of British Columbia, British Columbia, Canada
| | - Arlo Adams
- Department of Cellular and Physiological SciencesUniversity of British Columbia, British Columbia, Canada
| | - Matthew Piva
- Department of Cellular and Physiological SciencesUniversity of British Columbia, British Columbia, Canada
| | - Vanessa Nassour
- Department of Cellular and Physiological SciencesUniversity of British Columbia, British Columbia, Canada
| |
Collapse
|
13
|
Dias TR, Alves MG, Silva J, Barros A, Sousa M, Casal S, Silva BM, Oliveira PF. Implications of epigallocatechin-3-gallate in cultured human Sertoli cells glycolytic and oxidative profile. Toxicol In Vitro 2017; 41:214-222. [DOI: 10.1016/j.tiv.2017.03.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/13/2017] [Accepted: 03/16/2017] [Indexed: 11/26/2022]
|
14
|
Cardoso AM, Alves MG, Mathur PP, Oliveira PF, Cavaco JE, Rato L. Obesogens and male fertility. Obes Rev 2017; 18:109-125. [PMID: 27776203 DOI: 10.1111/obr.12469] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/04/2016] [Accepted: 08/15/2016] [Indexed: 12/28/2022]
Abstract
In the last decades, several studies evidenced a decrease in male fertility in developed countries. Although the aetiology of this trend in male reproductive health remains a matter of debate, environmental compounds that predispose to weight gain, namely obesogens, are appointed as contributors because of their action as endocrine disruptors. Obesogens favour adipogenesis by an imbalance of metabolic processes and can be found virtually everywhere. These compounds easily accumulate in tissues with high lipid content. Obesogens change the functioning of male reproductive axis, and, consequently, the testicular physiology and metabolism that are pivotal for spermatogenesis. The disruption of these tightly regulated metabolic pathways leads to adverse reproductive outcomes. Notably, adverse effects of obesogens may also promote disturbances in the metabolic performance of the following generations, through epigenetic modifications passed by male gametes. Thus, unveiling the molecular pathways by which obesogens induce toxicity that may end up in epigenetic modifications is imperative. Otherwise, a transgenerational susceptibility to metabolic diseases may be favoured. We present an up-to-date overview of the impact of obesogens on testicular physiology, with a particular focus on testicular metabolism. We also address the effects of obesogens on male reproductive parameters and the subsequent consequences for male fertility.
Collapse
Affiliation(s)
- A M Cardoso
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| | - M G Alves
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| | - P P Mathur
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India.,KIIT University, Bhubaneswar, India
| | - P F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.,i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - J E Cavaco
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| | - L Rato
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
15
|
Martins AD, Sá R, Monteiro MP, Barros A, Sousa M, Carvalho RA, Silva BM, Oliveira PF, Alves MG. Ghrelin acts as energy status sensor of male reproduction by modulating Sertoli cells glycolytic metabolism and mitochondrial bioenergetics. Mol Cell Endocrinol 2016; 434:199-209. [PMID: 27392494 DOI: 10.1016/j.mce.2016.07.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 07/04/2016] [Accepted: 07/04/2016] [Indexed: 02/06/2023]
Abstract
Ghrelin is a growth hormone-releasing peptide that has been suggested to interfere with spermatogenesis, though the underling mechanisms remain unknown. We studied the effect of ghrelin in human Sertoli cells (hSCs) metabolic phenotype. For that, hSCs were exposed to increasing concentrations of ghrelin (20, 100 and 500 pM) mimicking the levels reported in obese, normal weight, and severely undernourished individuals. The metabolite production/consumption was determined. The protein levels of key glycolysis-related transporters and enzymes were assessed. The lactate dehydrogenase (LDH) activity was measured. Mitochondrial complexes protein levels and mitochondria membrane potential were also measured. We showed that hSCs express the growth hormone secretagogue receptor. At the concentration present in the plasma of normal weight men, ghrelin caused a decrease of glucose consumption and mitochondrial membrane potential in hSCs, though LDH activity and lactate production remained unchanged, illustrating an alteration of glycolytic flux efficiency. Exposure of hSCs to levels of ghrelin found in the plasma of severely undernourished individuals decreased pyruvate consumption and mitochondrial complex III protein expression. All concentrations of ghrelin decreased alanine and acetate production by hSCs. Notably, the effects of ghrelin levels found in severely undernourished individuals were more pronounced in hSCs metabolic phenotype highlighting the importance of a proper eating behavior to maintain male reproductive potential. In conclusion, ghrelin acts as an energy status sensor for hSCs in a dose-dependent manner, showing an inverse association with the production of lactate, thus controlling the nutritional support of spermatogenesis.
Collapse
Affiliation(s)
- A D Martins
- Department of Microscopy, Laboratory of Cell Biology, Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, 4050-313, Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine, Abel Salazar Institute of Biomedical Sciences (UMIB-ICBAS), University of Porto, 4050-313, Porto, Portugal
| | - R Sá
- Department of Microscopy, Laboratory of Cell Biology, Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, 4050-313, Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine, Abel Salazar Institute of Biomedical Sciences (UMIB-ICBAS), University of Porto, 4050-313, Porto, Portugal
| | - M P Monteiro
- Unit for Multidisciplinary Research in Biomedicine, Abel Salazar Institute of Biomedical Sciences (UMIB-ICBAS), University of Porto, 4050-313, Porto, Portugal; Department of Anatomy, Abel Salazar Institute of Biomedical Sciences, ICBAS, University of Porto, 4050-313, Porto, Portugal
| | - A Barros
- Centre for Reproductive Genetics Professor Alberto Barros, 4100-009, Porto, Portugal; Department of Genetics, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - M Sousa
- Department of Microscopy, Laboratory of Cell Biology, Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, 4050-313, Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine, Abel Salazar Institute of Biomedical Sciences (UMIB-ICBAS), University of Porto, 4050-313, Porto, Portugal; Centre for Reproductive Genetics Professor Alberto Barros, 4100-009, Porto, Portugal
| | - R A Carvalho
- Department of Life Sciences, Faculty of Sciences and Technology and Center for Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-504, Coimbra, Portugal
| | - B M Silva
- Health Sciences Research Centre (CICS), University of Beira Interior, 6201-506, Covilhã, Portugal
| | - P F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, 4050-313, Porto, Portugal; Unit for Multidisciplinary Research in Biomedicine, Abel Salazar Institute of Biomedical Sciences (UMIB-ICBAS), University of Porto, 4050-313, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal
| | - M G Alves
- Department of Life Sciences, Faculty of Sciences and Technology and Center for Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-504, Coimbra, Portugal; Health Sciences Research Centre (CICS), University of Beira Interior, 6201-506, Covilhã, Portugal.
| |
Collapse
|
16
|
Bernardino RL, Marinelli RA, Maggio A, Gena P, Cataldo I, Alves MG, Svelto M, Oliveira PF, Calamita G. Hepatocyte and Sertoli Cell Aquaporins, Recent Advances and Research Trends. Int J Mol Sci 2016; 17:ijms17071096. [PMID: 27409609 PMCID: PMC4964472 DOI: 10.3390/ijms17071096] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 06/22/2016] [Accepted: 07/04/2016] [Indexed: 12/30/2022] Open
Abstract
Aquaporins (AQPs) are proteinaceous channels widespread in nature where they allow facilitated permeation of water and uncharged through cellular membranes. AQPs play a number of important roles in both health and disease. This review focuses on the most recent advances and research trends regarding the expression and modulation, as well as physiological and pathophysiological functions of AQPs in hepatocytes and Sertoli cells (SCs). Besides their involvement in bile formation, hepatocyte AQPs are involved in maintaining energy balance acting in hepatic gluconeogenesis and lipid metabolism, and in critical processes such as ammonia detoxification and mitochondrial output of hydrogen peroxide. Roles are played in clinical disorders including fatty liver disease, diabetes, obesity, cholestasis, hepatic cirrhosis and hepatocarcinoma. In the seminiferous tubules, particularly in SCs, AQPs are also widely expressed and seem to be implicated in the various stages of spermatogenesis. Like in hepatocytes, AQPs may be involved in maintaining energy homeostasis in these cells and have a major role in the metabolic cooperation established in the testicular tissue. Altogether, this information represents the mainstay of current and future investigation in an expanding field.
Collapse
Affiliation(s)
- Raquel L Bernardino
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal.
| | - Raul A Marinelli
- Instituto de Fisiología Experimental-CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario, 531 S2002LRK Rosario, Santa Fe, Argentina.
| | - Anna Maggio
- Department of Biosciences, Biotechnologies and Biopharnaceutics, University of Bari "Aldo Moro", 70125 Bari, Italy.
| | - Patrizia Gena
- Department of Biosciences, Biotechnologies and Biopharnaceutics, University of Bari "Aldo Moro", 70125 Bari, Italy.
| | - Ilaria Cataldo
- Department of Biosciences, Biotechnologies and Biopharnaceutics, University of Bari "Aldo Moro", 70125 Bari, Italy.
| | - Marco G Alves
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal.
| | - Maria Svelto
- Department of Biosciences, Biotechnologies and Biopharnaceutics, University of Bari "Aldo Moro", 70125 Bari, Italy.
| | - Pedro F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Giuseppe Calamita
- Department of Biosciences, Biotechnologies and Biopharnaceutics, University of Bari "Aldo Moro", 70125 Bari, Italy.
| |
Collapse
|
17
|
Bernardino RL, Costa AR, Martins AD, Silva J, Barros A, Sousa M, Sá R, Alves MG, Oliveira PF. Estradiol modulates Na(+) -dependent HCO3 (-) transporters altering intracellular pH and ion transport in human Sertoli cells: A role on male fertility? Biol Cell 2016; 108:179-88. [PMID: 26888167 DOI: 10.1111/boc.201500094] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/10/2016] [Accepted: 02/11/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND INFORMATION Infertile men often present deregulation of serum estrogen levels. Notably, high levels of estradiol (E2) are associated with low sperm production and quality. Sertoli cells (SCs) are responsible for spermatogenesis maintenance and are major targets for the hormonal signalling that regulates this complex process. RESULTS In this study, we used primary cultures of human SCs and studied the localisation, expression and functionality of the Na(+) -dependent HCO3 (-) transporters by confocal microscopy, immunoblot, epifluorescence and voltage clamp after 24 h of exposure to E2 (100 nM). All studied transporters were identified in human SCs. In E2-treated human SCs, there was an increase in NBCn1, NBCe1 and NDCBE protein levels, as well as an increase in intracellular pH and a decrease in transcellular transport. CONCLUSIONS We report an association between increased levels of E2 and the expression/function of Na(+) -dependent HCO3 (-) transporters in human SCs. Our results provide new evidence on the mechanisms by which E2 can regulate SCs physiology and consequently spermatogenesis. These mechanisms may have an influence on male reproductive potential and help to explain male infertility conditions associated with estrogen deregulation. SIGNIFICANCE Exposure to E2 increased human SCs intracellular pH. E2 is a modulator of ionic transcellular transport in human SCs.
Collapse
Affiliation(s)
- Raquel L Bernardino
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
| | - Ana R Costa
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Ana D Martins
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.,CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Joaquina Silva
- Centre for Reproductive Genetics Prof. Alberto Barros, Porto, Portugal
| | - Alberto Barros
- Centre for Reproductive Genetics Prof. Alberto Barros, Porto, Portugal.,Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal.,I3S, Institute for Innovation and Health Research, University of Porto, Porto, Portugal
| | - Mário Sousa
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.,Centre for Reproductive Genetics Prof. Alberto Barros, Porto, Portugal
| | - Rosália Sá
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal
| | - Marco G Alves
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Pedro F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.,I3S, Institute for Innovation and Health Research, University of Porto, Porto, Portugal
| |
Collapse
|
18
|
Jesus TT, Oliveira PF, Silva J, Barros A, Ferreira R, Sousa M, Cheng CY, Silva BM, Alves MG. Mammalian target of rapamycin controls glucose consumption and redox balance in human Sertoli cells. Fertil Steril 2015; 105:825-833.e3. [PMID: 26698679 DOI: 10.1016/j.fertnstert.2015.11.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/16/2015] [Accepted: 11/18/2015] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To study the role of mammalian target of rapamycin (mTOR) in the regulation of human Sertoli cell (hSC) metabolism, mitochondrial activity, and oxidative stress. DESIGN Experimental study. SETTING University research center and private assisted reproductive technology centers. PATIENT(S) Six men with anejaculation (psychological, vascular, neurologic) and conserved spermatogenesis. INTERVENTION(S) Testicular biopsies were used from patients under treatment for recovery of male gametes. Primary hSCs cultures were established from each biopsy and divided into a control group and one treated with rapamycin, the inhibitor of mTOR, for 24 hours. MAIN OUTCOME MEASURE(S) Cytotoxicity of hSCs to rapamycin was evaluated by sulforhodamine B assay. The glycolytic profile of hSCs was assessed by proton nuclear magnetic resonance and by studying protein expression of key glycolysis-related transporters and enzymes. Expression of mitochondrial complexes and citrate synthase activity were determined. Protein carbonylation, nitration, lipid peroxidation, and sulfhydryl protein group contents were quantified. The mTOR signaling pathway was studied. RESULT(S) Rapamycin increased glucose consumption by hSCs, maintaining lactate production. Alanine production by rapamycin-exposed hSCs was affected, resulting in an unbalanced intracellular redox state. Rapamycin-exposed hSCs had decreased expression of mitochondrial complex III and increased lipid peroxidation, whereas other oxidative stress markers were unaltered. Treatment of hSCs with rapamycin down-regulated phospho-mTOR (Ser-2448) levels, illustrating an effective partial inhibition of mTORC1. Protein levels of downstream signaling molecule p-4E-BP1 were not altered, suggesting that during treatment it became rephosphorylated. CONCLUSION(S) We show that mTOR regulates the nutritional support of spermatogenesis by hSCs and redox balance in these cells.
Collapse
Affiliation(s)
- Tito T Jesus
- Department of Microscopy, Laboratory of Cell Biology and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã
| | - Pedro F Oliveira
- Department of Microscopy, Laboratory of Cell Biology and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Institute of Health Research an Innovation, Portugal
| | - Joaquina Silva
- Centre for Reproductive Genetics Prof. Alberto Barros, Porto, Portugal
| | - Alberto Barros
- Institute of Health Research an Innovation, Portugal; Centre for Reproductive Genetics Prof. Alberto Barros, Porto, Portugal; Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rita Ferreira
- Organic Chemistry, Natural and Agrofood Products Centre, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Mário Sousa
- Department of Microscopy, Laboratory of Cell Biology and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Centre for Reproductive Genetics Prof. Alberto Barros, Porto, Portugal
| | - C Yan Cheng
- Center for Biomedical Research, Population Council, New York, New York
| | - Branca M Silva
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã
| | - Marco G Alves
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã.
| |
Collapse
|
19
|
Jiao ZJ, Yi W, Rong YW, Kee JD, Zhong WX. MicroRNA-1285 Regulates 17β-Estradiol-Inhibited Immature Boar Sertoli Cell Proliferation via Adenosine Monophosphate-Activated Protein Kinase Activation. Endocrinology 2015; 156:4059-70. [PMID: 26287402 DOI: 10.1210/en.2014-1982] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This study investigated the capacity of 10 μM 17β-estradiol to inhibit immature boar Sertoli cell (SC) proliferation and the involvement of microRNA (miR)-1285 in this process. SC viability and cell cycle progression were investigated using a cell counting kit-8 and flow cytometry, respectively. Expression of AMP-activated protein kinase (AMPK), S phase kinase-associated protein 2 (Skp2), and miR-1285 was analyzed by real-time RT-PCR and Western blotting. 17β-Estradiol (10 μM) reduced SC viability and miR-1285 expression and promoted AMPK phosphorylation. A double-stranded synthetic miR-1285 mimic promoted SC viability, increased levels of ATP, and phosphorylated mammalian target of rapamycin (mTOR) and Skp2 mRNA and protein, whereas p53 and p27 expression decreased, and 17β-estradiol-mediated effects on SCs were significantly attenuated. A single-stranded synthetic miR-1285 inhibitor produced the opposite effects on these measures. Activation of AMPK inhibited SC viability, reduced levels of ATP, phosphorylated mTOR and Skp2 mRNA and protein, and increased p53 and p27 expression. An AMPK inhibitor (compound C) attenuated the effects of 17β-estradiol on SCs. This indicated that 17β-estradiol (10 μM) reduced SC proliferation by inhibiting miR-1285 and thus activating AMPK. Phosphorylated AMPK is involved in the regulation of 17β-estradiol-mediated inhibition of SC viability through increasing p53 and p27 expression and inhibiting mTOR and Skp2 expression. Our findings also implicated Skp2 as the downstream integration point of p53 and mTOR. These findings indicated that miR-1285 may represent a target for the manipulation of boar sperm production.
Collapse
Affiliation(s)
- Zhang Jiao Jiao
- Chongqing Key Laboratory of Forage and Herbivore (Z.J.J., W.Y., Y.W.R., W.X.Z.), College of Animal Science and Technology, Southwest University, Chongqing 400715, China; and Genetic Engineering and Stem Cell Biology Laboratory (Z.J.J., J.D.K.), Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju 690756, South Korea
| | - Wang Yi
- Chongqing Key Laboratory of Forage and Herbivore (Z.J.J., W.Y., Y.W.R., W.X.Z.), College of Animal Science and Technology, Southwest University, Chongqing 400715, China; and Genetic Engineering and Stem Cell Biology Laboratory (Z.J.J., J.D.K.), Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju 690756, South Korea
| | - Yang Wei Rong
- Chongqing Key Laboratory of Forage and Herbivore (Z.J.J., W.Y., Y.W.R., W.X.Z.), College of Animal Science and Technology, Southwest University, Chongqing 400715, China; and Genetic Engineering and Stem Cell Biology Laboratory (Z.J.J., J.D.K.), Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju 690756, South Korea
| | - Jeong Dong Kee
- Chongqing Key Laboratory of Forage and Herbivore (Z.J.J., W.Y., Y.W.R., W.X.Z.), College of Animal Science and Technology, Southwest University, Chongqing 400715, China; and Genetic Engineering and Stem Cell Biology Laboratory (Z.J.J., J.D.K.), Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju 690756, South Korea
| | - Wang Xian Zhong
- Chongqing Key Laboratory of Forage and Herbivore (Z.J.J., W.Y., Y.W.R., W.X.Z.), College of Animal Science and Technology, Southwest University, Chongqing 400715, China; and Genetic Engineering and Stem Cell Biology Laboratory (Z.J.J., J.D.K.), Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju 690756, South Korea
| |
Collapse
|
20
|
Dias TR, Alves MG, Almeida SP, Silva J, Barros A, Sousa M, Silva BM, Silvestre SM, Oliveira PF. Dehydroepiandrosterone and 7-oxo-dehydroepiandrosterone in male reproductive health: Implications of differential regulation of human Sertoli cells metabolic profile. J Steroid Biochem Mol Biol 2015; 154:1-11. [PMID: 26134425 DOI: 10.1016/j.jsbmb.2015.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 05/28/2015] [Accepted: 06/22/2015] [Indexed: 11/25/2022]
Abstract
Dehydroepiandrosterone (DHEA) is a precursor of androgen synthesis whose action is partially exerted through its metabolites. 7-Oxo-dehydroepiandrosterone (7-oxo-DHEA) is a common DHEA metabolite, non-convertible to androgens, which constitutes a promising therapeutic strategy for multiple conditions. Sertoli cells (SCs) are responsible for the support of spermatogenesis, having unique metabolic characteristics strongly modulated by androgens. Consequently, disruptions in androgen synthesis compromise SCs function and hence male fertility. We aimed to evaluate the effects of DHEA and 7-oxo-DHEA in human SCs (hSCs) metabolism and oxidative profile. To do so, hSCs were exposed to increasing concentrations of DHEA and 7-oxo-DHEA (0.025, 1 and 50 μM) that revealed to be non-cytotoxic in these experimental conditions. We measured hSCs metabolites consumption/production by (1)H NMR, the protein expression levels of key players of the glycolytic pathway by Western blot as well as the levels of carbonyl groups, nitration and lipid peroxidation by Slot blot. The obtained data demonstrated that 7-oxo-DHEA is a more potent metabolic modulator than DHEA since it increased hSCs glycolytic flux. DHEA seem to redirect hSCs metabolism to the Krebs cycle, while 7-oxo-DHEA has some inhibitory effect in this path. The highest 7-oxo-DHEA concentrations (1 and 50 μM) also increased lactate production, which is of extreme relevance for the successful progression of spermatogenesis in vivo. None of these steroids altered the intracellular oxidative profile of hSCs, illustrating that, at the concentrations used they do not have pro- nor antioxidant actions in hSCs. Our study represents a further step in the establishment of safe doses of DHEA and 7-oxo-DHEA to hSCs, supporting its possible use in hormonal and non-hormonal therapies against male reproductive problems.
Collapse
Affiliation(s)
- Tânia R Dias
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Marco G Alves
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Susana P Almeida
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal
| | - Joaquina Silva
- Centre for Reproductive Genetics Prof. Alberto Barros, 4100-009 Porto, Portugal
| | - Alberto Barros
- Centre for Reproductive Genetics Prof. Alberto Barros, 4100-009 Porto, Portugal; Department of Genetics, Faculty of Medicine, University of Porto, 4100-009 Porto, Portugal; Institute of Health Research and Innovation, University of Porto, 4100-009 Porto, Portugal
| | - Mário Sousa
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal; Centre for Reproductive Genetics Prof. Alberto Barros, 4100-009 Porto, Portugal
| | - Branca M Silva
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Samuel M Silvestre
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal; CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Pedro F Oliveira
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal; Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
21
|
Bernardino RL, Martins AD, Jesus TT, Sá R, Sousa M, Alves MG, Oliveira PF. Estrogenic regulation of bicarbonate transporters from SLC4 family in rat Sertoli cells. Mol Cell Biochem 2015; 408:47-54. [DOI: 10.1007/s11010-015-2481-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 06/13/2015] [Indexed: 11/30/2022]
|
22
|
Dose-dependent effects of caffeine in human Sertoli cells metabolism and oxidative profile: Relevance for male fertility. Toxicology 2015; 328:12-20. [DOI: 10.1016/j.tox.2014.12.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 12/03/2014] [Accepted: 12/04/2014] [Indexed: 02/08/2023]
|
23
|
Rato L, Alves MG, Cavaco JE, Oliveira PF. High-energy diets: a threat for male fertility? Obes Rev 2014; 15:996-1007. [PMID: 25346452 DOI: 10.1111/obr.12226] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/26/2014] [Accepted: 08/26/2014] [Indexed: 12/18/2022]
Abstract
Male fertility is declining in developed countries, as well as in developing countries. External factors linked to lifestyle, such as eating disorders, negatively affect spermatogenesis, both at central and gonadal levels. The overconsumption of high-energy diets (HED) alters the functioning of the male reproductive axis and consequently affects the testicular physiology, disrupting its metabolism and bioenergetic capacity. Testicular metabolism presents unique characteristics, partly because of its cellular heterogeneity and to the specific functions that each cell type plays within the testicular environment. Disruption of the tightly regulated metabolic pathways leads to adverse reproductive outcomes, such as inefficient energy supply to germ cells, sperm defects or spermatogenesis arrest. Testicular metabolic alterations induced by HED intake may also lead to mitochondrial dysfunction, which is closely associated to reactive oxygen species (ROS) overproduction and oxidative stress. ROS easily target spermatozoa DNA and lipids, contributing to decreased sperm quality. Thus, understanding the detrimental effects of HED overconsumption on the pathways underlying testicular metabolism and sperm production is imperative; otherwise, one may favour a transgenerational amplification of subfertility. Herein, we present an up-to-date overview of the effects of HED on testicular metabolism, sperm parameters and the subsequent consequences for male fertility.
Collapse
Affiliation(s)
- L Rato
- CICS - UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | | | | |
Collapse
|
24
|
Alves MG, Martins AD, Vaz CV, Correia S, Moreira PI, Oliveira PF, Socorro S. Metformin and male reproduction: effects on Sertoli cell metabolism. Br J Pharmacol 2014; 171:1033-42. [PMID: 24261663 DOI: 10.1111/bph.12522] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 11/04/2013] [Accepted: 11/18/2013] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Metformin is commonly used to treat type 2 diabetes (T2D). While new clinical applications have been ascribed to metformin, including treatment of anovulatory infertility, its effects on male reproduction have not been investigated. The Sertoli cell (SC) is crucial for germ cell development, exerting metabolic control of spermatogenesis, therefore, we investigated the effects of metformin on SC metabolism. EXPERIMENTAL APPROACH Rat SCs were cultured in the absence and presence of metformin (5, 50 and 500 μM). mRNA and protein levels of glucose transporters (GLUT1 and GLUT3), phosphofructokinase 1 (PFK 1), lactate dehydrogenase (LDH) and monocarboxylate transporter 4 (MCT4) were determined by quantitative PCR and Western blot respectively. LDH activity was assessed and metabolite production/consumption determined by (1) H-NMR. KEY RESULTS Metformin (50 μM) decreased mRNA and protein levels of GLUT1, GLUT3, MCT4 and PFK 1 but did not affect LDH mRNA or protein levels. However, although glucose consumption was maintained in metformin-treated cells, LDH activity, lactate and alanine production were increased, indicating an enhanced glycolytic flux. No metabolic cytotoxicity was detected in SCs exposed to supra-pharmacological concentration of metformin. CONCLUSIONS AND IMPLICATIONS Our results indicate that metformin: (i) decreases mRNA and protein levels of glycolysis-related transporters in SCs but increases their activity; and (ii) stimulates alanine production, which induces antioxidant activity and maintains the NADH/NAD(+) equilibrium. The increased lactate in metformin-treated SCs provides nutritional support and has an anti-apoptotic effect in developing germ cells. Thus, metformin can be considered as a suitable antidiabetic drug for male patients of reproductive age with T2D.
Collapse
Affiliation(s)
- M G Alves
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | | | | | | | | | | |
Collapse
|
25
|
Rocha CS, Martins AD, Rato L, Silva BM, Oliveira PF, Alves MG. Melatonin alters the glycolytic profile of Sertoli cells: implications for male fertility. Mol Hum Reprod 2014; 20:1067-76. [PMID: 25205674 DOI: 10.1093/molehr/gau080] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Melatonin co-operates with insulin in the regulation of glucose homeostasis. Within the testis, glucose metabolism in the somatic Sertoli cells (SCs) is pivotal for spermatogenesis. Since the effects of melatonin on male reproductive physiology remain largely unknown, we hypothesized that melatonin may affect spermatogenesis by modulating SC metabolism, interacting with insulin. To test our hypothesis, rat SCs were maintained in culture for 24 h in the presence of insulin, melatonin or both and metabolite production/consumption was determined by proton nuclear magnetic resonance ((1)H-NMR). Protein levels of glucose transporters (GLUT1 and GLUT3), phosphofructokinase 1, lactate dehydrogenase (LDH) and monocarboxylate transporter 4 were determined by western blot. LDH activity was also assessed. SCs treated with melatonin showed an increase in glucose consumption via modulation of GLUT1 levels, but decreased LDH protein expression and activity, which resulted in lower lactate production. Moreover, SCs exposed to melatonin produced and accumulated less acetate than insulin-exposed cells. The combined treatment (insulin plus melatonin) increased acetate production by SCs, but intracellular acetate content remained lower than in insulin exposed cells. Finally, the intracellular redox state, as reflected by intracellular lactate/alanine ratio, was maintained at control levels in SCs by melatonin exposure (i.e. melatonin, alone or with insulin, increased the lactate/alanine ratio versus cells treated with insulin). Furthermore, SCs exposed to insulin plus melatonin produced more lactate and maintained the protein levels of some glycolysis-related enzymes and transporters at control levels. These findings illustrate that melatonin regulates SCs metabolism, and thus may affect spermatogenesis. Since lactate produced by SCs provides nutritional support and has an anti-apoptotic effect in developing germ cells, melatonin supplementation may be an effective therapy for diabetic male individuals facing subfertility/infertility.
Collapse
Affiliation(s)
- Cátia S Rocha
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana D Martins
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Luís Rato
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Branca M Silva
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Pedro F Oliveira
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Marco G Alves
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
26
|
Alves MG, Oliveira PF. 2,4-Dichlorophenoxyacetic acid alters intracellular pH and ion transport in the outer mantle epithelium of the bivalve Anodonta cygnea. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 154:12-18. [PMID: 24854203 DOI: 10.1016/j.aquatox.2014.04.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 04/15/2014] [Accepted: 04/28/2014] [Indexed: 06/03/2023]
Abstract
Bivalve molluscs, due to their sedentary mode of life and filter-feeding behavior, are very susceptible to pollutant bioaccumulation and used as sentinel organisms in the assessment of environment pollution. Herein we aimed to determine the in vivo, ex vivo and in vitro effects of 2,4-dichlorophenoxyacetic acid (2,4-D), a widely used herbicide, in Anodonta cygnea shell growth mechanisms. For that, we evaluated the effect of 2,4-D (100 μM) exposure on the transepithelial short-circuit current (Isc), potential (Vt) and conductance (Gt), as well as on OME ion transport systems and intracellular pH (pHi). In vivo exposure to 2,4-D caused an increase of 50% on the Isc generated by OME and ex vivo addition of that compound to the apical side of OME also induced an Isc increase. Furthermore, 2,4-D was able to cause a pHi increase in isolated cells of OME. Noteworthy, when 2,4-D was added following the exposure to specific inhibitors of several membrane transporters identified as responsible for pHi maintenance in these cells, no significant effect was observed on pHi except when the V-type ATPase inhibitor was used, indicating an overlap with the effect of 2,4-D. Thus, we concluded that 2,4-D is able of enhancing the activity of the V-ATPases present on the OME of A. cygnea and that this effect seems to be due to a direct stimulation of those H(+) transporters present on the apical portion of the membrane of OME cells, which are vital for shell maintenance and growth. This study allows us to better understand the molecular mechanisms behind 2,4-D toxicity and its deleterious effect in aquatic ecosystems, with particular emphasis on those involved in shell formation of bivalves.
Collapse
Affiliation(s)
- Marco G Alves
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal
| | - Pedro F Oliveira
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal.
| |
Collapse
|
27
|
Oliveira PF, Martins AD, Moreira AC, Cheng CY, Alves MG. The Warburg effect revisited--lesson from the Sertoli cell. Med Res Rev 2014; 35:126-51. [PMID: 25043918 DOI: 10.1002/med.21325] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Otto Warburg observed that cancerous cells prefer fermentative instead of oxidative metabolism of glucose, although the former is in theory less efficient. Since Warburg's pioneering works, special attention has been given to this difference in cell metabolism. The Warburg effect has been implicated in cell transformation, immortalization, and proliferation during tumorigenesis. Cancer cells display enhanced glycolytic activity, which is correlated with high proliferation, and thus, glycolysis appears to be an excellent candidate to target cancer cells. Nevertheless, little attention has been given to noncancerous cells that exhibit a "Warburg-like" metabolism with slight, but perhaps crucial, alterations that may provide new directions to develop new and effective anticancer therapies. Within the testis, the somatic Sertoli cell (SC) presents several common metabolic features analogous to cancer cells, and a clear "Warburg-like" metabolism. Nevertheless, SCs actively proliferate only during a specific time period, ceasing to divide in most species after puberty, when they become terminally differentiated. The special metabolic features of SC, as well as progression from the immature but proliferative state, to the mature nonproliferative state, where a high glycolytic activity is maintained, make these cells unique and a good model to discuss new perspectives on the Warburg effect. Herein we provide new insight on how the somatic SC may be a source of new and exciting information concerning the Warburg effect and cell proliferation.
Collapse
Affiliation(s)
- Pedro F Oliveira
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | | | | | | |
Collapse
|
28
|
Oliveira PF, Alves MG, Martins AD, Correia S, Bernardino RL, Silva J, Barros A, Sousa M, Cavaco JE, Socorro S. Expression pattern of G protein-coupled receptor 30 in human seminiferous tubular cells. Gen Comp Endocrinol 2014; 201:16-20. [PMID: 24681226 DOI: 10.1016/j.ygcen.2014.02.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/23/2014] [Accepted: 02/26/2014] [Indexed: 12/30/2022]
Abstract
The role of estrogens in male reproductive physiology has been intensively studied over the last few years. Yet, the involvement of their specific receptors has long been a matter of debate. The selective testicular expression of the classic nuclear estrogen receptors (ERα and ERβ) argues in favor of ER-specific functions in the spermatogenic event. Recently, the existence of a G protein-coupled estrogen receptor (GPR30) mediating non-genomic effects of estrogens has also been described. However, little is known about the specific testicular expression pattern of GPR30, as well as on its participation in the control of male reproductive function. Herein, by means of immunohistochemical and molecular biology techniques (RT-PCR and Western blot), we aimed to present the first exhaustive evaluation of GPR30 expression in non-neoplastic human testicular cells. Indeed, we were able to demonstrate that GPR30 was expressed in human testicular tissue and that the staining pattern was consistent with its cytoplasmic localization. Additionally, by using cultured human Sertoli cells (SCs) and isolated haploid and diploid germ cells fractions, we confirmed that GPR30 is expressed in SCs and diploid germ cells but not in haploid germ cells. This specific expression pattern suggests a role for GPR30 in spermatogenesis.
Collapse
Affiliation(s)
- Pedro F Oliveira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - Marco G Alves
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana D Martins
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Sara Correia
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Raquel L Bernardino
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Joaquina Silva
- Centre for Reproductive Genetics Alberto Barros, 4100-009 Porto, Portugal
| | - Alberto Barros
- Centre for Reproductive Genetics Alberto Barros, 4100-009 Porto, Portugal; Department of Genetics, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Mário Sousa
- Centre for Reproductive Genetics Alberto Barros, 4100-009 Porto, Portugal; Department of Microscopy, Laboratory of Cell Biology and Biomedical Research Multidisciplinary Unit (UMIB-FCT), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4099-003 Porto, Portugal
| | - José E Cavaco
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| |
Collapse
|
29
|
Alves MG, Oliveira PF. Effects of non-steroidal estrogen diethylstilbestrol on pH and ion transport in the mantle epithelium of a bivalve Anodonta cygnea. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2013; 97:230-235. [PMID: 23953926 DOI: 10.1016/j.ecoenv.2013.07.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/20/2013] [Accepted: 07/25/2013] [Indexed: 06/02/2023]
Abstract
Freshwater bivalves are used as sentinel organisms to detect pollutants effects in the aquatic environment due to their sedentary nature, filter-feeding behaviour. We aimed to determine the in vivo, ex vivo and in vitro influence of Diethylstilbestrol (DES), a widely used synthetic non-steroidal estrogen and endocrine disruptor, in Anodonta cygnea shell growth mechanisms. For that, in vivo exposure to DES (0.75μM) during 15 days, in vitro and ex vivo exposure of outer mantle epithelium (OME) cells to DES (0.75μM), were performed followed by study of short-circuit current (Isc), transepithelial potential (Vt) and transepithelial conductance (Gt) as well as identification of membrane transport systems and intracellular pH (pHi). Our results show that in vivo exposure to DES decreases in 30% the OME Isc and ex vivo addition of DES to the basolateral side of OME also induced Isc decrease. Several membrane transporters such as V-type ATPases, Na(+)/H(+) exchangers, Na(+)-K(+) pump, Na(+)-driven and Na(+)-independent HCO3(-)/Cl(-) transporters and Na(+)/HCO3(-) co-transporter were identified as responsible for pHi maintenance in OME and noteworthy, DES caused a pHi decrease in OME cells similar to the effect observed when OME cells were exposed to 4,4'-diisothiocyanostilbene disulfonic acid (DIDS), an inhibitor of several bicarbonate membrane transporters. The addition of DIDS after OME cells exposure to DES did not cause any alteration. We concluded that DES is able to modulate membrane ion transport and pHi in the OME of A. cygnea and that this effect seems to be due to inhibition of HCO3(-)/Cl(-) co-transporters present on the basolateral membrane.
Collapse
Affiliation(s)
- Marco G Alves
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6201-506 Covilhã, Portugal
| | | |
Collapse
|
30
|
Insulin deprivation decreases caspase-dependent apoptotic signaling in cultured rat sertoli cells. ISRN UROLOGY 2013; 2013:970370. [PMID: 24228182 PMCID: PMC3817687 DOI: 10.1155/2013/970370] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/12/2013] [Indexed: 11/30/2022]
Abstract
Insulin is essential for the regulation of glucose homeostasis. Insulin dysfunction occurs in several pathologies, such as diabetes mellitus, which is associated with fertility problems. Somatic Sertoli cells (SCs) not only metabolize glucose to lactate, which is the central energy source used by developing germ cells, but also determine the germ cell population size. If a deregulation in SCs apoptosis occurs, it will affect germ cells, compromising spermatogenesis. As SCs apoptotic signaling is a hormonally regulated process, we hypothesized that the lack of insulin could lead to alterations in apoptotic signaling. Therefore, we examined the effect of insulin deprivation on several markers of apoptotic signaling in cultured rat SCs. We determined mRNA and protein expression of apoptotic markers as well as caspase-3 activity. SCs cultured in insulin deprivation demonstrated a significant decrease on mRNA levels of p53, Bax, caspase-9, and caspase-3 followed by a significant increase of Bax and decrease of caspase-9 protein levels relatively to the control. Caspase-3 activity was also decreased in SCs cultured in insulin deprivation conditions. Our results show that insulin deprivation decreases caspase-dependent apoptotic signaling in cultured rat SCs evidencing a possible mechanism by which lack of insulin can affect spermatogenesis and fertility.
Collapse
|
31
|
Bernardino RL, Martins AD, Socorro S, Alves MG, Oliveira PF. Effect of Prediabetes on Membrane Bicarbonate Transporters in Testis and Epididymis. J Membr Biol 2013; 246:877-83. [DOI: 10.1007/s00232-013-9601-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/24/2013] [Indexed: 11/29/2022]
|
32
|
Martins AD, Alves MG, Simões VL, Dias TR, Rato L, Moreira PI, Socorro S, Cavaco JE, Oliveira PF. Control of Sertoli cell metabolism by sex steroid hormones is mediated through modulation in glycolysis-related transporters and enzymes. Cell Tissue Res 2013; 354:861-8. [DOI: 10.1007/s00441-013-1722-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 08/16/2013] [Indexed: 11/29/2022]
|
33
|
Exposure to 2,4-dichlorophenoxyacetic acid alters glucose metabolism in immature rat Sertoli cells. Reprod Toxicol 2013; 38:81-8. [DOI: 10.1016/j.reprotox.2013.03.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 02/12/2013] [Accepted: 03/16/2013] [Indexed: 11/22/2022]
|
34
|
Alves MG, Martins AD, Cavaco JE, Socorro S, Oliveira PF. Diabetes, insulin-mediated glucose metabolism and Sertoli/blood-testis barrier function. Tissue Barriers 2013; 1:e23992. [PMID: 24665384 PMCID: PMC3875609 DOI: 10.4161/tisb.23992] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 02/10/2013] [Accepted: 02/12/2013] [Indexed: 02/06/2023] Open
Abstract
Blood testis barrier (BTB) is one of the tightest blood-barriers controlling the entry of substances into the intratubular fluid. Diabetes Mellitus (DM) is an epidemic metabolic disease concurrent with falling fertility rates, which provokes severe detrimental BTB alterations. It induces testicular alterations, disrupting the metabolic cooperation between the cellular constituents of BTB, with dramatic consequences on sperm quality and fertility. As Sertoli cells are involved in the regulation of spermatogenesis, providing nutritional support for germ cells, any metabolic alteration in these cells derived from DM may be responsible for spermatogenesis disruption, playing a crucial role in fertility/subfertility associated with this pathology. These cells have a glucose sensing machinery that reacts to hormonal fluctuations and several mechanisms to counteract hyper/hypoglycemic events. The role of DM on Sertoli/BTB glucose metabolism dynamics and the metabolic molecular mechanisms through which DM and insulin deregulation alter its functioning, affecting male reproductive potential will be discussed.
Collapse
Affiliation(s)
- Marco G. Alves
- CICS-UBI; Health Sciences Research Centre; University of Beira Interior; Covilhã, Portugal
| | - Ana D. Martins
- CICS-UBI; Health Sciences Research Centre; University of Beira Interior; Covilhã, Portugal
| | - José E. Cavaco
- CICS-UBI; Health Sciences Research Centre; University of Beira Interior; Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI; Health Sciences Research Centre; University of Beira Interior; Covilhã, Portugal
| | - Pedro F. Oliveira
- CICS-UBI; Health Sciences Research Centre; University of Beira Interior; Covilhã, Portugal
| |
Collapse
|
35
|
Gene expression networks underlying ovarian development in wild largemouth bass (Micropterus salmoides). PLoS One 2013; 8:e59093. [PMID: 23527095 PMCID: PMC3604104 DOI: 10.1371/journal.pone.0059093] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 02/12/2013] [Indexed: 01/12/2023] Open
Abstract
Background Oocyte maturation in fish involves numerous cell signaling cascades that are activated or inhibited during specific stages of oocyte development. The objectives of this study were to characterize molecular pathways and temporal gene expression patterns throughout a complete breeding cycle in wild female largemouth bass to improve understanding of the molecular sequence of events underlying oocyte maturation. Methods Transcriptomic analysis was performed on eight morphologically diverse stages of the ovary, including primary and secondary stages of oocyte growth, ovulation, and atresia. Ovary histology, plasma vitellogenin, 17β-estradiol, and testosterone were also measured to correlate with gene networks. Results Global expression patterns revealed dramatic differences across ovarian development, with 552 and 2070 genes being differentially expressed during both ovulation and atresia respectively. Gene set enrichment analysis (GSEA) revealed that early primary stages of oocyte growth involved increases in expression of genes involved in pathways of B-cell and T-cell receptor-mediated signaling cascades and fibronectin regulation. These pathways as well as pathways that included adrenergic receptor signaling, sphingolipid metabolism and natural killer cell activation were down-regulated at ovulation. At atresia, down-regulated pathways included gap junction and actin cytoskeleton regulation, gonadotrope and mast cell activation, and vasopressin receptor signaling and up-regulated pathways included oxidative phosphorylation and reactive oxygen species metabolism. Expression targets for luteinizing hormone signaling were low during vitellogenesis but increased 150% at ovulation. Other networks found to play a significant role in oocyte maturation included those with genes regulated by members of the TGF-beta superfamily (activins, inhibins, bone morphogenic protein 7 and growth differentiation factor 9), neuregulin 1, retinoid X receptor, and nerve growth factor family. Conclusions This study offers novel insight into the gene networks underlying vitellogenesis, ovulation and atresia and generates new hypotheses about the cellular pathways regulating oocyte maturation.
Collapse
|
36
|
Alves MG, Rato L, Carvalho RA, Moreira PI, Socorro S, Oliveira PF. Hormonal control of Sertoli cell metabolism regulates spermatogenesis. Cell Mol Life Sci 2013; 70:777-93. [PMID: 23011766 PMCID: PMC11113727 DOI: 10.1007/s00018-012-1079-1] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 06/11/2012] [Accepted: 06/26/2012] [Indexed: 11/30/2022]
Abstract
Hormonal regulation is essential to spermatogenesis. Sertoli cells (SCs) have functions that reach far beyond the physical support of germ cells, as they are responsible for creating the adequate ionic and metabolic environment for germ cell development. Thus, much attention has been given to the metabolic functioning of SCs. During spermatogenesis, germ cells are provided with suitable metabolic substrates, in a set of events mediated by SCs. Multiple signaling cascades regulate SC function and several of these signaling pathways are hormone-dependent and cell-specific. Within the seminiferous tubules, only SCs possess receptors for some hormones rendering them major targets for the hormonal signaling that regulates spermatogenesis. Although the mechanisms by which SCs fulfill their own and germ cells metabolic needs are mostly studied in vitro, SC metabolism is unquestionably a regulation point for germ cell development and the hormonal control of these processes is required for a normal spermatogenesis.
Collapse
Affiliation(s)
- Marco G. Alves
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Luís Rato
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Rui A. Carvalho
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology (FCTUC), University of Coimbra, 3004-517 Coimbra, Portugal
| | - Paula I. Moreira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Sílvia Socorro
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Pedro F. Oliveira
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
37
|
Alves MG, Martins AD, Rato L, Moreira PI, Socorro S, Oliveira PF. Molecular mechanisms beyond glucose transport in diabetes-related male infertility. Biochim Biophys Acta Mol Basis Dis 2013; 1832:626-35. [PMID: 23348098 DOI: 10.1016/j.bbadis.2013.01.011] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 01/15/2013] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus (DM) is one of the greatest public health threats in modern societies. Although during a few years it was suggested that DM had no significant effect in male reproductive function, this view has been challenged in recent years. The increasing incidence of DM worldwide will inevitably result in a higher prevalence of this pathology in men of reproductive age and subfertility or infertility associated with DM is expected to dramatically rise in upcoming years. From a clinical perspective, the evaluation of semen parameters, as well as spermatozoa deoxyribonucleic acid (DNA) integrity, are often studied due to their direct implications in natural and assisted conception. Nevertheless, recent studies based on the molecular mechanisms beyond glucose transport in testicular cells provide new insights in DM-induced alterations in male reproductive health. Testicular cells have their own glucose sensing machinery that react to hormonal fluctuations and have several mechanisms to counteract hyper- and hypoglycemic events. Moreover, the metabolic cooperation between testicular cells is crucial for normal spermatogenesis. Sertoli cells (SCs), which are the main components of blood-testis barrier, are not only responsible for the physical support of germ cells but also for lactate production that is then metabolized by the developing germ cells. Any alteration in this tied metabolic cooperation may have a dramatic consequence in male fertility potential. Therefore, we present an overview of the clinical significance of DM in the male reproductive health with emphasis on the molecular mechanisms beyond glucose fluctuation and transport in testicular cells.
Collapse
Affiliation(s)
- M G Alves
- University of Beira Interior, Covilhã, Portugal.
| | | | | | | | | | | |
Collapse
|
38
|
Lucas TFG, Amaral LS, Porto CS, Quintas LEM. Na+/K+-ATPase α1 isoform mediates ouabain-induced expression of cyclin D1 and proliferation of rat sertoli cells. Reproduction 2012; 144:737-45. [PMID: 23028124 DOI: 10.1530/rep-12-0232] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Novel roles for the interaction of cardiotonic steroids to Na(+)/K(+)-ATPase have been established in recent years. The aim of this study was to investigate the intracellular signaling events downstream the action of ouabain on Na(+)/K(+)-ATPase in Sertoli cell obtained from immature rats. Treatment of Sertoli cells with ouabain (1 μM) induced a rapid and transient increase in the extracellular signal-regulated kinase (ERK1/2 or MAPK3/1) and phosphatidylinositol 3-kinase (PI3K)/serine-threonine protein kinase (AKT) phosphorylation. Also, ouabain upregulated the expression of cyclin D1 and incorporation of [methyl-(3)H]thymidine, both of which were dependent on MAPK3/1 but not AKT intracellular cascade, as shown by pretreatment with MEK (MAP2K1/2) inhibitor U0126 and PI3K inhibitor wortmannin respectively. Moreover, the effect of ouabain on these proliferation parameters was completely prevented by phospho-cAMP response element-binding protein (CREB)/CREB-binding protein complex inhibitor KG501 and only partially by nuclear factor κB nuclear translocation inhibitor SN50. Pretreatment with estrogen receptor antagonist ICI 182780 showed that MAPK3/1 activation by ouabain does not involve this receptor. The Na(+)/K(+)-ATPase α1 isoform, but not α4, was detected in Sertoli cells, suggesting that ouabain effects in Sertoli cells are mediated via α1. Taken together, these results show a rapid ouabain action in the Sertoli cells, which in turn can modulate nuclear transcriptional events essential for Sertoli cell proliferation in a critical period of testicular development. Our findings are important to understand the role of ouabain in the testis and its possible implications in male infertility.
Collapse
Affiliation(s)
- Thaís F G Lucas
- Setor de Endocrinologia Experimental, Departamento de Farmacologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
39
|
Alves MG, Socorro S, Silva J, Barros A, Sousa M, Cavaco JE, Oliveira PF. In vitro cultured human Sertoli cells secrete high amounts of acetate that is stimulated by 17β-estradiol and suppressed by insulin deprivation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1389-94. [DOI: 10.1016/j.bbamcr.2012.06.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 05/22/2012] [Accepted: 06/05/2012] [Indexed: 10/28/2022]
|
40
|
Metabolic modulation induced by oestradiol and DHT in immature rat Sertoli cells cultured in vitro. Biosci Rep 2012; 32:61-9. [PMID: 21671886 DOI: 10.1042/bsr20110030] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Sertoli cells actively metabolize glucose that is converted into lactate, which is used by developing germ cells for their energy metabolism. Androgens and oestrogens have general metabolic roles that reach far beyond reproductive processes. Hence, the main purpose of this study was to examine the effect of sex hormones on metabolite secretion/consumption in primary cultures of rat Sertoli cells. Sertoli cell-enriched cultures were maintained in a defined medium for 50 h. Glucose and pyruvate consumption, and lactate and alanine secretion were determined, by 1H-NMR (proton NMR) spectra analysis, in the presence or absence of 100 nM E2 (17β-oestradiol) or 100 nM 5α-DHT (dihydrotestosterone). Cells cultured in the absence (control) or presence of E2 consumed the same amount of glucose (29±2 pmol/cell) at similar rates during the 50 h. After 25 h of treatment with DHT, glucose consumption and glucose consumption rate significantly increased. Control and E2-treated cells secreted similar amounts of lactate during the 50 h, while the amount of lactate secreted by DHT-treated cells was significantly lower. Such a decrease was concomitant with a significant decrease in LDH A [LDH (lactate dehydrogenase) chain A] and MCT4 [MCT (monocarboxylate transporter) isoform 4] mRNA levels after 50 h treatment in hormonally treated groups, being more pronounced in DHT-treated groups. Finally, alanine production was significantly increased in E2-treated cells after 25 h treatment, which indicated a lower redox/higher oxidative state for the cells in those conditions. Together, these results support the existence of a relation between sex hormones action and energy metabolism, providing an important assessment of androgens and oestrogens as metabolic modulators in rat Sertoli cells.
Collapse
|
41
|
Oliveira P, Alves M, Rato L, Laurentino S, Silva J, Sá R, Barros A, Sousa M, Carvalho R, Cavaco J, Socorro S. Effect of insulin deprivation on metabolism and metabolism-associated gene transcript levels of in vitro cultured human Sertoli cells. Biochim Biophys Acta Gen Subj 2012; 1820:84-9. [DOI: 10.1016/j.bbagen.2011.11.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 10/14/2011] [Accepted: 11/10/2011] [Indexed: 10/15/2022]
|
42
|
Laurentino SS, Correia S, Cavaco JE, Oliveira PF, Rato L, Sousa M, Barros A, Socorro S. Regucalcin is broadly expressed in male reproductive tissues and is a new androgen-target gene in mammalian testis. Reproduction 2011; 142:447-56. [DOI: 10.1530/rep-11-0085] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Regucalcin (RGN) is a calcium (Ca2+)-binding protein which regulates intracellular Ca2+homeostasis by modulating the activity of enzymes regulating Ca2+concentration and enhancing Ca2+-pumping activity. Several studies have described the pivotal role of proper Ca2+homeostasis regulation to spermatogenesis and male fertility. Recently,RGNwas identified as a sex steroid-regulated gene in prostate and breast; however, a possible role of RGN in spermatogenesis has not been examined. In this study, the expression and localization of RGN in rat and human testis, and other rat reproductive tissues was analyzed. Moreover, we studied whether RGN protein was present in seminiferous tubule fluid (STF). Finally, we examined the effect of 5α-dihydrotestosterone (DHT) on the expression ofRgnmRNA in rat seminiferous tubules (SeT) culturedex vivo. The results presented in this study show that RGN is expressed in Leydig and Sertoli cells, as well as in all types of germ cells of both rat and human testis. RGN is also expressed in rat prostate, epididymis, and seminal vesicles. Moreover, RGN protein is present in rat STF. The results also demonstrate thatRgnexpression is age dependent in rat testis, and is upregulated by the non-aromatizable androgen DHT in rat SeT culturedex vivo. Taken together, these findings indicate thatRgnis a novel androgen-target gene in rat testis and that it may have a role in male reproductive function, particularly in the control of spermatogenesis.
Collapse
|
43
|
Oliveira PF, Alves MG, Rato L, Silva J, Sá R, Barros A, Sousa M, Carvalho RA, Cavaco JE, Socorro S. Influence of 5α-dihydrotestosterone and 17β-estradiol on human Sertoli cells metabolism. ACTA ACUST UNITED AC 2011; 34:e612-20. [PMID: 21812787 DOI: 10.1111/j.1365-2605.2011.01205.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Sertoli cells metabolize glucose, converting it to lactate that is used by developing germ cells for their energy metabolism. Androgens and oestrogens have metabolic roles that reach far beyond reproductive processes. So, the main purpose of this study was to examine the effect of sex steroid hormones on metabolite secretion/consumption in human Sertoli cells. Human Sertoli cell-enriched primary cultures were maintained in a defined medium for 50 h and glucose, pyruvate, lactate and alanine variations were determined using (1) H-NMR spectra analysis, in the absence or presence of 100 nm 17β-estradiol (E(2) ) or 100 nm 5α-dihydrotestosterone (DHT). The mRNA expression levels of glucose transporters, lactate dehydrogenase and monocarboxylate transporters were also determined using semi-quantitative RT-PCR. Cells cultured in the absence (control) or presence of E(2) consumed the same amounts of glucose at similar rates during the 50 h. During the first 15 h of treatment with DHT, glucose consumption and glucose consumption rate were significantly higher. Nevertheless, DHT-treated cells secreted a significantly lower amount of lactate than control and E(2) -treated cells. Such a decrease was concomitant with a significant decrease in lactate dehydrogenase A mRNA levels after 50 h treatment in DHT-treated groups. Finally, alanine production was significantly increased in E(2) -treated cells after 25 h treatment, which indicated a lower redox/higher oxidative state for the cells on those conditions. These results support the existence of a relationship between sex steroid hormones action and energy metabolism, providing the first assessment of androgens and oestrogens as metabolic modulators of human Sertoli cells.
Collapse
Affiliation(s)
- P F Oliveira
- CICS - UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Rato L, Socorro S, Cavaco JEB, Oliveira PF. Tubular Fluid Secretion in the Seminiferous Epithelium: Ion Transporters and Aquaporins in Sertoli Cells. J Membr Biol 2010; 236:215-24. [DOI: 10.1007/s00232-010-9294-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2010] [Accepted: 07/20/2010] [Indexed: 01/01/2023]
|