1
|
Avena MV, Funes AK, Monclus MÁ, Boarelli PV, Barbisan LF, Bernal-López MR, Gómez-Huelgas R, Lancellotti TES, Fornés MW. Cholesterol and SREBP2 Dynamics During Spermatogenesis Stages in Rabbits: Effects of High-Fat Diet and Protective Role of Extra Virgin Olive Oil. Int J Mol Sci 2025; 26:4062. [PMID: 40362302 PMCID: PMC12071441 DOI: 10.3390/ijms26094062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
High-fat diets (HFDs) have been found to compromise male fertility, with cholesterol dysregulation being a key factor. Sterol regulatory element-binding protein 2 (SREBP2) is a crucial transcription factor that regulates cholesterol biosynthesis and uptake, playing an essential role in maintaining cholesterol homeostasis in the testes. This study investigated the dynamics of SREBP2 and cholesterol levels during rabbit spermatogenesis under HFD conditions. Our findings reveal that SREBP2 expression fluctuates throughout the seminiferous epithelium cycle. However, HFDs induce stage-specific disruptions in cholesterol balance, leading to sperm with increased membrane cholesterol, a reduced sperm count in semen analysis, impaired motility, abnormal morphology, and decreased functionality. In the control group, SREBP2 expression patterns underscored its critical role in normal spermatogenesis. Interestingly, supplementation with extra virgin olive oil (EVOO) reversed the negative effects of HFD, normalizing SREBP2 expression and cholesterol content, which improved sperm quality. These findings emphasize the importance of stage-specific analysis in understanding how dietary fat impacts male fertility and suggest that EVOO may serve as a potential nutritional intervention to protect reproductive health.
Collapse
Affiliation(s)
- María Virginia Avena
- Andrological Research Laboratory of Mendoza (LIAM), IHEM, National University of Cuyo, CONICET, Mendoza M5502JMA, Argentina; (M.V.A.); (A.K.F.); (M.Á.M.)
- Metabolic Diseases Laboratory (LEM), Juan Agustín Maza University, Mendoza M5519, Argentina;
| | - Abi Karenina Funes
- Andrological Research Laboratory of Mendoza (LIAM), IHEM, National University of Cuyo, CONICET, Mendoza M5502JMA, Argentina; (M.V.A.); (A.K.F.); (M.Á.M.)
- Metabolic Diseases Laboratory (LEM), Juan Agustín Maza University, Mendoza M5519, Argentina;
| | - María Ángeles Monclus
- Andrological Research Laboratory of Mendoza (LIAM), IHEM, National University of Cuyo, CONICET, Mendoza M5502JMA, Argentina; (M.V.A.); (A.K.F.); (M.Á.M.)
| | - Paola Vanina Boarelli
- Metabolic Diseases Laboratory (LEM), Juan Agustín Maza University, Mendoza M5519, Argentina;
| | - Luis Fernando Barbisan
- Biosciences Institute, Department of Structural and Functional Biology, São Paulo State University (UNESP), Botucatu 18610-034, SP, Brazil;
| | - M. Rosa Bernal-López
- Internal Medicine Department, Regional University Hospital of Málaga, Málaga Biomedical Research Institute (IBIMA), 29009 Málaga, Spain; (M.R.B.-L.); (R.G.-H.)
- Biomedical Research Networking Center on Obesity and Nutrition (CIBERobn), Carlos III Health Institute, 28029 Madrid, Spain
| | - Ricardo Gómez-Huelgas
- Internal Medicine Department, Regional University Hospital of Málaga, Málaga Biomedical Research Institute (IBIMA), 29009 Málaga, Spain; (M.R.B.-L.); (R.G.-H.)
- Biomedical Research Networking Center on Obesity and Nutrition (CIBERobn), Carlos III Health Institute, 28029 Madrid, Spain
| | - Tania Estefania Saez Lancellotti
- Andrological Research Laboratory of Mendoza (LIAM), IHEM, National University of Cuyo, CONICET, Mendoza M5502JMA, Argentina; (M.V.A.); (A.K.F.); (M.Á.M.)
- Internal Medicine Department, Regional University Hospital of Málaga, Málaga Biomedical Research Institute (IBIMA), 29009 Málaga, Spain; (M.R.B.-L.); (R.G.-H.)
- Biomedical Research Networking Center on Obesity and Nutrition (CIBERobn), Carlos III Health Institute, 28029 Madrid, Spain
| | - Miguel Walter Fornés
- Andrological Research Laboratory of Mendoza (LIAM), IHEM, National University of Cuyo, CONICET, Mendoza M5502JMA, Argentina; (M.V.A.); (A.K.F.); (M.Á.M.)
| |
Collapse
|
2
|
Zhu Y, Liu X, Liu X, Shi Y, Li H, Ru S, Tian H. Toxicokinetics and reproductive toxicity of maternal bisphenol AF exposure during gestation in offspring of Sprague Dawley rats. Chem Biol Interact 2025; 409:111424. [PMID: 39938710 DOI: 10.1016/j.cbi.2025.111424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/10/2025] [Accepted: 02/10/2025] [Indexed: 02/14/2025]
Abstract
Bisphenol AF (BPAF) has been widely used as a main alternative to bisphenol A (BPA), and previous in vitro studies have shown that BPAF has higher reproductive toxicity potentials than BPA. However, data on in vivo toxicity of BPAF is still limited. In this study, Sprague Dawley rats were exposed to BPAF (0, 50, and 100 mg/kg/day) during gestation to study toxicokinetics and reproductive toxicity in offspring. The results showed that plasma concentrations BPAF peaked within 6 h after birth, followed by a two-phase decay, with clearance rates of approximately 3.0 l/h and terminal half-life values ranging from 77 h to 114 h, suggesting fast absorption and high persistence of BPAF. At postnatal day 21 (PND21), BPAF was found to be bioaccumulated in reproductive organs (testes and ovaries) of the offspring, resulting in adverse effects on reproduction in both sexes. Lower anogenital distance, reduced relative testicular weight, dissolved interstitial cells, fewer primary spermatocytes, decreased testosterone levels, and increased luteinizing hormone levels were detected in male offspring. In female offspring, vacuolization in follicular antrum, fewer follicles, increased 17β-estradiol levels, and increased luteinizing hormone levels in female offspring were found. Gene expression of scavenger receptor class B type I (SR-B1), 3-hydroxy-3-methylglutaryl-CoA reductase (HMGR), sterol regulatory element-binding protein-1c (SREBP-1c), and several steroidogenic enzymes was significantly decreased in male offspring following maternal exposure to BPAF, suggesting that the decreases in testosterone levels is a result of inhibited cholesterol uptake, cholesterol de novo synthesis, and steroidogenesis. In addition, inhibition of pathways of phagosome and cell adhesion molecules might be the underlying molecular mechanism involved in BPAF-induced reproductive disorders in male offspring. This study provides the scientific basis for a comprehensive assessment of the safety of BPAF.
Collapse
Affiliation(s)
- Yaxuan Zhu
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Xiuxiang Liu
- Qingdao Women and Children's Hospital, Qingdao, 266034, China
| | - Xiuying Liu
- Wudi County Hospital of Traditional Chinese Medicine, Binzhou, 251900, China
| | - Yijiao Shi
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Huaxin Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Hua Tian
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
3
|
Understanding the Underlying Molecular Mechanisms of Meiotic Arrest during In Vitro Spermatogenesis in Rat Prepubertal Testicular Tissue. Int J Mol Sci 2022; 23:ijms23115893. [PMID: 35682573 PMCID: PMC9180380 DOI: 10.3390/ijms23115893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 12/10/2022] Open
Abstract
In vitro spermatogenesis appears to be a promising approach to restore the fertility of childhood cancer survivors. The rat model has proven to be challenging, since germ cell maturation is arrested in organotypic cultures. Here, we report that, despite a meiotic entry, abnormal synaptonemal complexes were found in spermatocytes, and in vitro matured rat prepubertal testicular tissues displayed an immature phenotype. RNA-sequencing analyses highlighted up to 600 differentially expressed genes between in vitro and in vivo conditions, including genes involved in blood-testis barrier (BTB) formation and steroidogenesis. BTB integrity, the expression of two steroidogenic enzymes, and androgen receptors were indeed altered in vitro. Moreover, most of the top 10 predicted upstream regulators of deregulated genes were involved in inflammatory processes or immune cell recruitment. However, none of the three anti-inflammatory molecules tested in this study promoted meiotic progression. By analysing for the first time in vitro matured rat prepubertal testicular tissues at the molecular level, we uncovered the deregulation of several genes and revealed that defective BTB function, altered steroidogenic pathway, and probably inflammation, could be at the origin of meiotic arrest.
Collapse
|
4
|
Funes AK, Simón L, Colombo R, Avena MV, Monclús M, Crescitelli J, Cabrillana ME, Conte MI, Cayado N, Boarelli P, Fornés MW, Saez Lancellotti TE. Impact of high fat diet on the sterol regulatory element-binding protein 2 cholesterol pathway in the testicle. Mol Hum Reprod 2021; 27:6206393. [PMID: 33787903 DOI: 10.1093/molehr/gaab023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/18/2021] [Indexed: 12/30/2022] Open
Abstract
Male fertility has been shown to be dependent on cholesterol homeostasis. This lipid is essential for testosterone synthesis and spermatogenesis, but its levels must be maintained in an optimal range for proper testicular function. In particular, sperm cells' development is very sensitive to high cholesterol levels, noticeably during acrosomal formation. The aim of this work was to study whether the molecular pathway that regulates intracellular cholesterol, the sterol regulatory element-binding protein (SREBP) pathway, is affected in the testicles of animals under a fat diet. To investigate this, we took advantage of the non-obese hypercholesterolemia (HC) model in New Zealand rabbits that displays poor sperm and seminal quality. The testicular expression of SREBP isoform 2 (SREBP2) and its target molecules 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR) and low-density lipoprotein receptor (LDLR) were studied under acute (6 months) and chronic (more than 12 months) fat intake by RT-PCR, western blot and immunofluorescence. Our findings showed that fat consumption promoted down-regulation of the SREBP2 pathway in the testicle at 6 months, but upregulation after a chronic period. This was consistent with load of testicular cholesterol, assessed by filipin staining. In conclusion, the intracellular pathway that regulates cholesterol levels in the testicle is sensitive to dietary fats, and behaves differently depending on the duration of consumption: it has a short-term protective effect, but became deregulated in the long term, ultimately leading to a detrimental situation. These results will contribute to the understanding of the basic mechanisms of the effect of fat consumption in humans with idiopathic infertility.
Collapse
Affiliation(s)
- Abi K Funes
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Layla Simón
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Regina Colombo
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - María Virginia Avena
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - María Monclús
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - Julieta Crescitelli
- Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| | - María E Cabrillana
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - María Inés Conte
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Niubys Cayado
- Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina.,Laboratorio de Oncología, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Paola Boarelli
- Laboratorio de Enfermedades Metabólicas (LEM), Universidad Juan Agustín Maza, Mendoza, Argentina
| | - Miguel W Fornés
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - Tania E Saez Lancellotti
- Laboratorio de Investigaciones Andrológicas de Mendoza (LIAM) †. Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Laboratorio de Biología Molecular del Metabolismo & Nutrición (bMeNu)†, Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Científico y Tecnológico (CCT), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Instituto de Investigaciones. Facultad de Ciencias Médicas. Universidad del Aconcagua. Mendoza, Argentina
| |
Collapse
|
5
|
Liver X Receptors and Male (In)fertility. Int J Mol Sci 2019; 20:ijms20215379. [PMID: 31671745 PMCID: PMC6862486 DOI: 10.3390/ijms20215379] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
Liver X receptors (LXRs) are ligand-dependent transcription factors acting as ‘cholesterol sensors’ to regulate lipid homeostasis in cells. The two isoforms, LXRα (NR1H3) and LXRβ (NR1H2), are differentially expressed, with the former expressed predominantly in metabolically active tissues and the latter more ubiquitously. Both are activated by oxidised cholesterol metabolites, endogenously produced oxysterols. LXRs have important roles in lipid metabolism and inflammation, plus a number of newly emerging roles. They are implicated in regulating lipid balance in normal male reproductive function and may provide a link between male infertility and lipid disorders and/or obesity. Studies from Lxr knockout mouse models provide compelling evidence to support this. More recently published data suggest distinct and overlapping roles of the LXR isoforms in the testis and recent evidence of a role for LXRs in human male fertility. This review summarises the current literature and explores the likely link between LXR, lipid metabolism and male fertility as part of a special issue on Liver X receptors in International Journal of Molecular Sciences.
Collapse
|
6
|
Ačimovič J, Goyal S, Košir R, Goličnik M, Perše M, Belič A, Urlep Ž, Guengerich FP, Rozman D. Cytochrome P450 metabolism of the post-lanosterol intermediates explains enigmas of cholesterol synthesis. Sci Rep 2016; 6:28462. [PMID: 27334049 PMCID: PMC4917857 DOI: 10.1038/srep28462] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 06/02/2016] [Indexed: 11/18/2022] Open
Abstract
Cholesterol synthesis is among the oldest metabolic pathways, consisting of the Bloch and Kandutch-Russell branches. Following lanosterol, sterols of both branches are proposed to be dedicated to cholesterol. We challenge this dogma by mathematical modeling and with experimental evidence. It was not possible to explain the sterol profile of testis in cAMP responsive element modulator tau (Crem τ) knockout mice with mathematical models based on textbook pathways of cholesterol synthesis. Our model differs in the inclusion of virtual sterol metabolizing enzymes branching from the pathway. We tested the hypothesis that enzymes from the cytochrome P450 (CYP) superfamily can participate in the catalysis of non-classical reactions. We show that CYP enzymes can metabolize multiple sterols in vitro, establishing novel branching points of cholesterol synthesis. In conclusion, sterols of cholesterol synthesis can be oxidized further to metabolites not dedicated to production of cholesterol. Additionally, CYP7A1, CYP11A1, CYP27A1, and CYP46A1 are parts of a broader cholesterol synthesis network.
Collapse
Affiliation(s)
- Jure Ačimovič
- Center for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Sandeep Goyal
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146, United States
| | - Rok Košir
- Center for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Marko Goličnik
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Martina Perše
- Medical Experimental Centre, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
| | - Ales Belič
- Faculty of Electrical Engineering, University of Ljubljana, Slovenia
| | - Žiga Urlep
- Center for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - F. Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232-0146, United States
| | - Damjana Rozman
- Center for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
7
|
Inoue K, Imai Y. Fatostatin, an SREBP inhibitor, prevented RANKL-induced bone loss by suppression of osteoclast differentiation. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2432-41. [DOI: 10.1016/j.bbadis.2015.08.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/31/2015] [Accepted: 08/24/2015] [Indexed: 11/27/2022]
|
8
|
Keber R, Ačimovič J, Majdič G, Motaln H, Rozman D, Horvat S. Male germ cell-specific knockout of cholesterogenic cytochrome P450 lanosterol 14α-demethylase (Cyp51). J Lipid Res 2013; 54:1653-1661. [PMID: 23509403 DOI: 10.1194/jlr.m035717] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cytochrome P450 lanosterol 14α-demethylase (CYP51) and its products, meiosis-activating sterols (MASs), were hypothesized by previous in vitro studies to have an important role in regulating meiosis and reproduction. To test this in vivo, we generated a conditional male germ cell-specific knockout of the gene Cyp51 in the mouse. High excision efficiency of Cyp51 allele in germ cells resulted in 85-89% downregulation of Cyp51 mRNA and protein levels in germ cells. Quantitative metabolic profiling revealed significantly higher levels of CYP51 substrates lanosterol and 24,25-dihydrolanosterol and substantially diminished levels of MAS, the immediate products of CYP51. However, germ cell-specific ablation of Cyp51, leading to lack of MAS, did not affect testicular morphology, daily sperm production, or reproductive performance in males. It is plausible that due to the similar structures of cholesterol intermediates, previously proposed biological function of MAS in meiosis progression can be replaced by some other yet-unidentified functionally redundant lipid molecule(s). Our results using the germ cell-specific knockout model provide first in vivo evidence that the de novo synthesis of MAS and cholesterol in male germ cells is most likely not essential for spermatogenesis and reproduction and that MASs, originating from germ cells, do not cell-autonomously regulate spermatogenesis and fertility.
Collapse
Affiliation(s)
- Rok Keber
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Domžale, Slovenia
| | - Jure Ačimovič
- Centre for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Majdič
- Center for Animal Genomics, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Helena Motaln
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Damjana Rozman
- Centre for Functional Genomics and Bio-Chips, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Simon Horvat
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Domžale, Slovenia; National Institute of Chemistry, Laboratory for Biotechnology, Ljubljana, Slovenia.
| |
Collapse
|
9
|
Fon Tacer K, Pompon D, Rozman D. Adaptation of cholesterol synthesis to fasting and TNF-alpha: profiling cholesterol intermediates in the liver, brain, and testis. J Steroid Biochem Mol Biol 2010; 121:619-25. [PMID: 20206258 DOI: 10.1016/j.jsbmb.2010.02.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 02/16/2010] [Accepted: 02/24/2010] [Indexed: 12/18/2022]
Abstract
Key players in pathogenesis of metabolic disorders are disturbed cholesterol balance and inflammation. In addition to cholesterol also sterol intermediates are biologically active, however, surprisingly little is known about their synthesis and roles. The aim of our study was to assess the interplay between the inflammatory cytokine TNF-alpha and cholesterol synthesis by measuring cholesterol and its intermediates in the liver, brain, and testis. Liquid chromatography-mass spectrometry has been applied to profile sterols of normally fed mice, during fasting and after TNF-alpha administration. In mice on normal chow diet, sterols other than cholesterol represent 0.5% in the liver, 1% in brain and 5% in testis. In the liver only 7-dehydrocholesterol, lanosterol and desmosterol were detected. Major sterol intermediates of the brain are desmosterol, testis meiosis activating sterol (T-MAS), and 7-dehydrocholesterol while in testis T-MAS predominates (4%), followed by desmosterol, lanosterol, 7-dehydrocholesterol and others. In 20h fasting there is no significant change in cholesterol of the three tissues, and no significant change in intermediates of the liver. In the brain sterol intermediates are lowered (significant for zymosterol) while in the testis the trend is opposite. TNF-alpha provokes a significant raise of some intermediates whereas the level of cholesterol is again unchanged. The proportion of sterols in the liver rises from 0.5% in controls to 1.2% in TNF-alpha-treated mice, which is in accordance with published expression profiling data. In conclusion, our data provide novel insights into the interaction between the inflammatory cytokine TNF-alpha and the tissue-specific cholesterol biosynthesis of the liver, brain and testis.
Collapse
Affiliation(s)
- Klementina Fon Tacer
- Center for Functional Genomic and Biochips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| | | | | |
Collapse
|
10
|
Hermo L, Pelletier RM, Cyr DG, Smith CE. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 4: intercellular bridges, mitochondria, nuclear envelope, apoptosis, ubiquitination, membrane/voltage-gated channels, methylation/acetylation, and transcription factors. Microsc Res Tech 2010; 73:364-408. [PMID: 19941288 DOI: 10.1002/jemt.20785] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
As germ cells divide and differentiate from spermatogonia to spermatozoa, they share a number of structural and functional features that are common to all generations of germ cells and these features are discussed herein. Germ cells are linked to one another by large intercellular bridges which serve to move molecules and even large organelles from the cytoplasm of one cell to another. Mitochondria take on different shapes and features and topographical arrangements to accommodate their specific needs during spermatogenesis. The nuclear envelope and pore complex also undergo extensive modifications concomitant with the development of germ cell generations. Apoptosis is an event that is normally triggered by germ cells and involves many proteins. It occurs to limit the germ cell pool and acts as a quality control mechanism. The ubiquitin pathway comprises enzymes that ubiquitinate as well as deubiquitinate target proteins and this pathway is present and functional in germ cells. Germ cells express many proteins involved in water balance and pH control as well as voltage-gated ion channel movement. In the nucleus, proteins undergo epigenetic modifications which include methylation, acetylation, and phosphorylation, with each of these modifications signaling changes in chromatin structure. Germ cells contain specialized transcription complexes that coordinate the differentiation program of spermatogenesis, and there are many male germ cell-specific differences in the components of this machinery. All of the above features of germ cells will be discussed along with the specific proteins/genes and abnormalities to fertility related to each topic.
Collapse
Affiliation(s)
- Louis Hermo
- Department of Anatomy and Cell Biology, Faculty of Medicine, McGill University, 3640 University Street, Montreal, QC Canada H3A 2B2.
| | | | | | | |
Collapse
|
11
|
Korošec T, Ačimovič J, Seliškar M, Kocjan D, Tacer KF, Rozman D, Urleb U. Novel cholesterol biosynthesis inhibitors targeting human lanosterol 14α-demethylase (CYP51). Bioorg Med Chem 2008; 16:209-21. [DOI: 10.1016/j.bmc.2007.10.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Revised: 09/15/2007] [Accepted: 10/01/2007] [Indexed: 11/29/2022]
|
12
|
Eacker SM, Agrawal N, Qian K, Dichek HL, Gong EY, Lee K, Braun RE. Hormonal regulation of testicular steroid and cholesterol homeostasis. Mol Endocrinol 2007; 22:623-35. [PMID: 18032697 DOI: 10.1210/me.2006-0534] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The male sex steroid, testosterone (T), is synthesized from cholesterol in the testicular Leydig cell under control of the pituitary gonadotropin LH. Unlike most cells that use cholesterol primarily for membrane synthesis, steroidogenic cells have additional requirements for cholesterol, because it is the essential precursor for all steroid hormones. Little is known about how Leydig cells satisfy their specialized cholesterol requirements for steroid synthesis. We show that in mice with a unique hypomorphic androgen mutation, which disrupts the feedback loop governing T synthesis, that genes involved in cholesterol biosynthesis/uptake and steroid biosynthesis are up-regulated. We identify LH as the central regulatory molecule that controls both steroidogenesis and Leydig cell cholesterol homeostasis in vivo. In addition to the primary defect caused by high levels of LH, absence of T signaling exacerbates the lipid homeostasis defect in Leydig cells by eliminating a short feedback loop. We show that T signaling can affect the synthesis of steroids and modulates the expression of genes involved in de novo cholesterol synthesis. Surprisingly, accumulation of active sterol response element-binding protein 2 is not required for up-regulation of genes involved in cholesterol biosynthesis and uptake in Leydig cells.
Collapse
Affiliation(s)
- Stephen M Eacker
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Fon Tacer K, Kuzman D, Seliskar M, Pompon D, Rozman D. TNF-alpha interferes with lipid homeostasis and activates acute and proatherogenic processes. Physiol Genomics 2007; 31:216-27. [PMID: 17566076 DOI: 10.1152/physiolgenomics.00264.2006] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The interaction between disrupted lipid homeostasis and immune response is implicated in the pathogenesis of several diseases, but the molecular bridges between the major players are still a matter of controversy. Our systemic study of the inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) in the livers of mice exposed to 20-h cytokine/fasting for the first time shows that TNF-alpha interferes with adaptation to fasting and activates harmful proatherogenic pathways, partially through interaction with the insulin-Insig-sterol regulatory element binding protein (Srebp) signaling pathway. In addition to the increased expression of acute-phase inflammatory genes, the most prominent alterations represent modified lipid homeostasis observed on the gene expression and metabolite levels. These include reduction of HDL-cholesterol, increase of LDL-cholesterol, and elevated expression of cholesterogenic genes, accompanied by increase of potentially harmful precholesterol metabolites and suppression of cholesterol elimination through bile acids, likely by farnesoid X receptor-independent mechanisms. On the transcriptional level, a shift from fatty oxidation toward fatty acid synthesis is observed. The concept of the influence of TNF-alpha on the Srebp regulatory network, followed by downstream effects on sterol metabolism, is novel. Observed acute alterations in lipid metabolism are in agreement with chronic disturbances found in patients.
Collapse
Affiliation(s)
- Klementina Fon Tacer
- Center for Functional Genomics and Biochips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Slovenia
| | | | | | | | | |
Collapse
|
14
|
Felder TK, Klein K, Patsch W, Oberkofler H. A novel SREBP-1 splice variant: tissue abundance and transactivation potency. ACTA ACUST UNITED AC 2005; 1731:41-7. [PMID: 16153721 DOI: 10.1016/j.bbaexp.2005.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2005] [Revised: 07/25/2005] [Accepted: 08/12/2005] [Indexed: 11/30/2022]
Abstract
Sterol regulatory element binding proteins (SREBPs) belong to the family of basic helix-loop-helix-leucine zipper transcription factors. The SREBP-1 gene encodes two different isoforms, SREBP-1a and -1c, that are expressed at varying levels in different tissues and cultured cells and exhibit common and distinct functions. We identified an additional SREBP-1 isoform, termed SREBP-1ac, and determined its mRNA abundance in different human tissues and cell lines. SREBP-1ac mRNA was detectable in all tissues studied, although at lower levels than the major SREBP-1a and -1c isoforms. Transcription of the novel SREBP isoform was not induced by insulin or cholesterol depletion. SREBP-1ac did not transactivate the human LDLR and UCP2 promoters but robustly attenuated the transactivation capacity of SREBP-1a, -1c and -2 in cotransfection experiments.
Collapse
Affiliation(s)
- Thomas Klaus Felder
- Department of Laboratory Medicine, Salzburger Landeskliniken and Paracelsus Private Medical University, Muellner Hauptstrasse 48, A-5020 Salzburg, Austria
| | | | | | | |
Collapse
|
15
|
Tully DB, Luft JC, Rockett JC, Ren H, Schmid JE, Wood CR, Dix DJ. Reproductive and genomic effects in testes from mice exposed to the water disinfectant byproduct bromochloroacetic acid. Reprod Toxicol 2005; 19:353-66. [PMID: 15686870 DOI: 10.1016/j.reprotox.2004.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2004] [Revised: 05/14/2004] [Accepted: 06/14/2004] [Indexed: 11/25/2022]
Abstract
A byproduct of drinking water disinfection, bromochloroacetic acid (BCA), acts as a reproductive toxicant in rats. To determine if BCA produces similar reproductive toxicity in mice, juvenile and adult C57BL/6 males were exposed to 0, 8, 24, 72 or 216 mg/kg of BCA once daily for 14 days. Five of 12 animals from each dose-group were sacrificed at the end of dosing, and testes, epididymes, and seminal vesicles harvested and weighed. Seven mice from each dose-group (including juvenile-exposed mice, following a 14-week maturation period) were used in a 40-day sequential breeding assay to determine if BCA targets a particular phase of spermatogenesis. No significant effects were observed in mice exposed to BCA as juveniles, and there were no effects on fertility by 14 weeks after dosing. However, effects were observed in adult-exposed mice over the first 10 days after BCA exposure: mean number of litters/male, percentage of litters/female bred, and total number of fetuses/male were all reduced by 72 and 216 mg/kg BCA. These results in adult mice indicate BCA disrupted differentiation of spermatids during dosing and the first 10 days of mating, and are consistent with the spermatid retention and atypical residual bodies observed in animals exposed to 72 and 216 mg/kg BCA. To investigate mechanisms involved, we utilized cDNA microarrays containing 950 testis-expressed genes to profile gene expression from Control and BCA-treated mice. Statistical analyses of microarray results identified 40 well-characterized genes differentially expressed in a dose responsive manner as a result of BCA exposure. Microarray results were supplemented with quantitative real-time PCR and Westerns for several genes and proteins. The 40 genes whose expression was altered by BCA are involved in numerous biological processes including: cell communication and adhesion, cell cycle and cell proliferation, metabolism, signal transduction, stress response, and spermatogenesis and male fertility. Modulated expression of these genes, particularly the 15 expressed in Sertoli cells and spermatids, offers new insights into potential mechanisms of BCA toxicity in the mouse testis.
Collapse
Affiliation(s)
- Douglas B Tully
- National Health and Environmental Effects Research Laboratory, Reproductive Toxicology Division, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, 27711, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Rozman D, Seliskar M, Cotman M, Fink M. Pre-cholesterol precursors in gametogenesis. Mol Cell Endocrinol 2005; 234:47-56. [PMID: 15836952 DOI: 10.1016/j.mce.2004.11.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Accepted: 11/09/2004] [Indexed: 10/25/2022]
Abstract
Meiosis activating sterols (MAS) are biologically active post-lanosterol intermediates of cholesterol biosynthesis that are synthetized primarily in the gonads, including the sperm. MAS reinitiate the meiosis of oocytes in vitro while in vivo they seem to contribute to the oocyte quality and the progression of meiosis. The mRNAs for the MAS-producing enzyme lanosterol 14alpha-demethylase (CYP51) arise by alternative poly (A) signal selection. Only signals with low cleavage activity are used in the testis. Translation of mammalian CYP51s starts at one of the tandem in-frame ATGs. CYP51 protein of the bull is shorter compared to the human due to the usage of a more downstream translation start site. CYP51 proteins are post-translationally modified by glycosylations in the Golgi and on acrosomal membranes of the sperm. Green fluorescence protein-based ex vivo system has been developed to aid studying the intracellular transport of the MAS-producing CYP51. The influence of the post-translational modifications on MAS-synthesizing capacity is under investigation.
Collapse
Affiliation(s)
- Damjana Rozman
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia.
| | | | | | | |
Collapse
|
17
|
Smith LH, Petrie MS, Morrow JD, Oates JA, Vaughan DE. The sterol response element binding protein regulates cyclooxygenase-2 gene expression in endothelial cells. J Lipid Res 2005; 46:862-71. [PMID: 15716578 DOI: 10.1194/jlr.m500021-jlr200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We previously demonstrated that cholesterol deprivation increases endothelial cyclooxygenase-2 (COX-2)-dependent prostacyclin [prostaglandin I2 (PGI2)] production in vitro. Cholesterol directly regulates gene transcription through the sterol response element binding protein (SREBP). In this work, we demonstrate that SREBP directly regulates COX-2 expression. Cholesterol reduces human COX-2 promoter-luciferase reporter construct activity in transiently transfected endothelial cells. Conversely, cotransfection with a constitutively active mutant SREBP increases COX-2 promoter activity. SREBP-1a and -2 specifically bind a putative sterol response element (SRE) sequence in the COX-2 promoter. This sequence competes for SREBP binding to a low density lipoprotein receptor consensus sequence in an electromobility-shift assay. These data indicate that endothelial COX-2 is regulated by cholesterol via the SREBP pathway. The present study identifies COX-2 as the first vascular gene without a clear role in lipid metabolism transactivated by SREBP, and suggests that enhanced production of PGI2 through this pathway may be an additional benefit of cholesterol-lowering therapies.
Collapse
Affiliation(s)
- Layton Harris Smith
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | |
Collapse
|
18
|
Cao X, Pomerantz SH, Popliker M, Tsafriri A. Meiosis-Activating Sterol Synthesis in Rat Preovulatory Follicle: Is It Involved in Resumption of Meiosis?1. Biol Reprod 2004; 71:1807-12. [PMID: 15269103 DOI: 10.1095/biolreprod.104.031773] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Meiosis-activating sterol (MAS) was shown to overcome the inhibitory effect of hypoxanthine on spontaneous maturation of mouse oocytes and was suggested to mediate the stimulation of meiosis by gonadotropins. Follicular fluid (FF)-MAS is synthesized by cytochrome P450 lanosterol 14alpha-demethylase (LDM). Follicular LDM was preferentially localized in oocytes by immunohistochemistry. Using [3H]acetate or R-[5-3H]mevalonate as precursors as well as high-performance liquid chromatographic and thin-layer chromatographic separation, we have measured the concentrations of de novo-synthesized lanosterol, FF-MAS, and cholesterol in rat graafian follicles, cumulus-oocyte complexes (COCs), and denuded oocytes (DOs) treated with LH, AY-9944 (an inhibitor of Delta14-reductase, which was anticipated to increase FF-MAS levels by inhibiting its metabolism), or both after 8 h of culture. In follicles, both LH and AY-9944 increased the accumulation of FF-MAS as compared to controls. In COCs, AY-9944 caused a marked increase in FF-MAS, but we were unable to detect accumulation of FF-MAS in DOs. Neither the endogenous increases in FF-MAS accumulation nor the addition of FF-MAS to the culture medium could overcome the inhibition on resumption of meiosis by phosphodiesterase inhibitors. Compared to LH-induced resumption of meiosis in follicles, that induced by AY-9944 was much delayed. These results call into question any role of FF-MAS as an obligatory mediator of LH activity on germinal vesicle breakdown. The discrepancy between the positive staining for LDM in oocytes and our inability to detect de novo synthesized FF-MAS in DOs may relate to the sensitivity of the methodology employed and either the number of oocytes used or a deficiency in LDM synthetic activity in such oocytes. Further studies are required to confirm any of these alternatives.
Collapse
Affiliation(s)
- Xiumei Cao
- Bernhard Zondek Hormone Research Laboratory, Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | |
Collapse
|