1,612
Views
18
CrossRef citations to date
0
Altmetric
Research Article

BMI1 promotes steroidogenesis through maintaining redox homeostasis in mouse MLTC-1 and primary Leydig cells

, , , , , , , , ORCID Icon, , , , ORCID Icon, , , & show all
Pages 1884-1898 | Received 31 Mar 2020, Accepted 02 Jun 2020, Published online: 28 Jun 2020

References

  • Harman SM, Metter EJ, Tobin JD, et al. Longitudinal effects of aging on serum total and free testosterone levels in healthy men. Baltimore longitudinal study of aging. J Clin Endocrinol Metab. 2001;86(2):724–731.
  • Mohr BA, Guay AT, O’Donnell AB, et al. Normal, bound and nonbound testosterone levels in normally ageing men: results from the massachusetts male ageing study. Clin Endocrinol (Oxf). 2005;62(1):64–73.
  • Araujo AB, Esche GR, Kupelian V, et al. Prevalence of symptomatic androgen deficiency in men. J Clin Endocrinol Metab. 2007;92(11):4241–4247.
  • Bhasin S, Basaria S. Diagnosis and treatment of hypogonadism in men. Best Pract Res Clin Endocrinol Metab. 2011;25(2):2251–70.
  • Surampudi PN, Wang C, Swerdloff R. Hypogonadism in the aging male diagnosis, potential benefits, and risks of testosterone replacement therapy. Int J Endocrinol. 2012;2012:625434.
  • Wu FC, Tajar A, Beynon JM, et al. Identification of late-onset hypogonadism in middle-aged and elderly men. N Engl J Med. 2010;363(2):123–135.
  • Huhtaniemi IT. Andropause–lessons from the European male ageing study. Ann Endocrinol (Paris). 2014;75(2):128–131.
  • Finkle WD, Greenland S, Ridgeway GK, et al. Increased risk of non-fatal myocardial infarction following testosterone therapy prescription in men. PLoS One. 2014;9(1):e85805.
  • Vigen R, O’Donnell CI, Baron AE, et al. Association of testosterone therapy with mortality, myocardial infarction, and stroke in men with low testosterone levels. JAMA. 2013;310(17):1829–1836.
  • Xu L, Freeman G, Cowling BJ, et al. Testosterone therapy and cardiovascular events among men: a systematic review and meta-analysis of placebo-controlled randomized trials. BMC Med. 2013;11(1):108.
  • Bosland MC. Testosterone treatment is a potent tumor promoter for the rat prostate. Endocrinology. 2014;155(12):4629–4633.
  • Pavlovich CP, King P, Goldstein M, et al. Evidence of a treatable endocrinopathy in infertile men. J Urol. 2001;165(3):837–841.
  • Ramasamy R, Stahl PJ, Schlegel PN. Medical therapy for spermatogenic failure. Asian J Androl. 2012;14(1):57–60.
  • Kaprara A, Huhtaniemi IT. The hypothalamus-pituitary-gonad axis: tales of mice and men. Metabolism. 2018;86:3–17.
  • Miller WL, Bose HS. Early steps in steroidogenesis: intracellular cholesterol trafficking. J Lipid Res. 2011;52(12):2111–2135.
  • Rone MB, Fan J, Papadopoulos V. Cholesterol transport in steroid biosynthesis: role of protein-protein interactions and implications in disease states. Biochim Biophys Acta. 2009;1791(7):646–658.
  • Biehs B, Hu JK, Strauli NB, et al. BMI1 represses Ink4a/arf and hox genes to regulate stem cells in the rodent incisor. Nat Cell Biol. 2013;15(7):846–852.
  • Li B, Chen Y, Wang F, et al. BMI1 drives hepatocarcinogenesis by repressing the TGFbeta2/SMAD signalling axis. Oncogene. 2020;39(5):1063–1079.
  • Park IK, Morrison SJ, Clarke MF. BMI1, stem cells, and senescence regulation. J Clin Invest. 2004;113(2):175–179.
  • Molofsky AV, Pardal R, Iwashita T, et al. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature. 2003;425(6961):962–967.
  • Park IK, Qian D, Kiel M, et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature. 2003;423(6937):302–305.
  • Zhang HW, Ding J, Jin JL, et al. Defects in mesenchymal stem cell self-renewal and cell fate determination lead to an osteopenic phenotype in Bmi-1 null mice. J Bone Miner Res. 2010;25(3):640–652.
  • Jin J, Lv X, Chen L, et al. Bmi-1 plays a critical role in protection from renal tubulointerstitial injury by maintaining redox balance. Aging Cell. 2014;13(5):797–809.
  • Huang Y, Chen N, Miao D. Biological effects of pyrroloquinoline quinone on liver damage in Bmi-1 knockout mice. Exp Ther Med. 2015;10(2):451–458.
  • Wang R, Xue X, Wang Y, et al. BMI1 deficiency results in female infertility by activating p16/p19 signaling and increasing oxidative stress. Int J Biol Sci. 2019;15(4):870–881.
  • Bracken AP, Kleine-Kohlbrecher D, Dietrich N, et al. The polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells. Genes Dev. 2007;21(5):525–530.
  • Liu J, Cao L, Chen J, et al. BMI1 regulates mitochondrial function and the DNA damage response pathway. Nature. 2009;459(7245):387–392.
  • Dai X, Zhang Q, Yu Z, et al. BMI1 deficient mice exhibit male infertility. Int J Biol Sci. 2018;14(3):358–368.
  • Zheng B, Zhou Q, Guo Y, et al. Establishment of a proteomic profile associated with gonocyte and spermatogonial stem cell maturation and differentiation in neonatal mice. Proteomics. 2014;14(2–3):274–285.
  • Zheng B, Yu J, Guo Y, et al. Cellular nucleic acid-binding protein is vital to testis development and spermatogenesis in mice. Reproduction. 2018;156(1):59–69.
  • Shen C, Yu J, Zhang X, et al. Strawberry Notch 1 (SBNO1) promotes proliferation of spermatogonial stem cells via the noncanonical Wnt pathway in mice. Asian J Androl. 2019;21(4):345–350.
  • Shen C, Zhang K, Yu J, et al. Stromal interaction molecule 1 is required for neonatal testicular development in mice. Biochem Biophys Res Commun. 2018;504(4):2111–2135.
  • Zhao D, Shen C, Gao T, et al. Myotubularin related protein 7 is essential for the spermatogonial stem cell homeostasis via PI3K/AKT signaling. Cell Cycle. 2019;18(20):2800–2813.
  • Zheng B, Zhao D, Zhang P, et al. Quantitative proteomics reveals the essential roles of stromal interaction molecule 1 (STIM1) in the testicular cord formation in mouse testis. Mol Cell Proteomics. 2015;14(10):2682–2691.
  • Zhang B, Ma W, Zhu Q, et al. The SET protein promotes androgen production in testicular Leydig cells. Andrology. 2018;6(3):478–487.
  • Xu W, Zhu Q, Liu S, et al. Calretinin participates in regulating steroidogenesis by PLC-Ca(2+)-PKC pathway in Leydig cells. Sci Rep. 2018;113(1):7403.
  • Mendoza-Villarroel RE, Robert NM, Martin LJ, et al. The nuclear receptor NR2F2 activates star expression and steroidogenesis in mouse MA-10 and MLTC-1 Leydig cells. Biol Reprod. 2014;914(1):26.
  • Kong Y, Ai C, Dong F, et al. Targeting of BMI-1 with PTC-209 inhibits glioblastoma development. Cell Cycle. 2018;17(10):1199–1211.
  • Zhu S, Zhao D, Li C, et al. BMI1 is directly regulated by androgen receptor to promote castration-resistance in prostate cancer. Oncogene. 2020;39(1):17–29.
  • Lombard DB, Chua KF, Mostoslavsky R, et al. DNA repair, genome stability, and aging. Cell. 2005;120(4):497–512.
  • Staples CJ, Barone G, Myers KN, et al. MRNIP/C5orf45 interacts with the MRN complex and contributes to the DNA damage response. Cell Rep. 2016;16(10):2565–2575.
  • Yin Y, Xue X, Wang Q, et al. BMI1 plays an important role in dentin and mandible homeostasis by maintaining redox balance. Am J Transl Res. 2016;8(11):4716–4725.
  • Molofsky AV, He S, Bydon M, et al. Bmi-1 promotes neural stem cell self-renewal and neural development but not mouse growth and survival by repressing the p16Ink4a and p19Arf senescence pathways. Genes Dev. 2005;19(12):1432–1437.
  • Ignatiadis M, Azim HA Jr., Desmedt C, et al. The genomic grade assay compared with Ki67 to determine risk of distant breast cancer recurrence. JAMA Oncol. 2016;2(2):217–224.
  • Bruggeman SW, Valk-Lingbeek ME, van der Stoop PP, et al. Ink4a and Arf differentially affect cell proliferation and neural stem cell self-renewal in BMI1-deficient mice. Genes Dev. 2005;19(12):1438–1443.
  • Chagraoui J, Hebert J, Girard S, et al. An anticlastogenic function for the polycomb group gene BMI1. Proc Natl Acad Sci U S A. 2011;108(13):5284–5289.
  • Wang H, Wang L, Erdjument-Bromage H, et al. Role of histone H2A ubiquitination in polycomb silencing. Nature. 2004;431(7010):873–878.
  • Zhou Y, Wang L, Vaseghi HR, et al. BMI1 is a key epigenetic barrier to direct cardiac reprogramming. Cell Stem Cell. 2016;18(3):382–395.
  • Banerjee Mustafi S, Aznar N, Dwivedi SK, et al. Mitochondrial BMI1 maintains bioenergetic homeostasis in cells. Faseb J. 2016;30(12):4042–4055.
  • Tremellen K. Oxidative stress and male infertility–a clinical perspective. Hum Reprod Update. 2008;14(3):243–258.
  • Clyne M. Male factor infertility: effects of ROS and vitamin E on sperm. Nat Rev Urol. 2012;9(2):62.
  • Moustafa MH, Sharma RK, Thornton J, et al. Relationship between ROS production, apoptosis and DNA denaturation in spermatozoa from patients examined for infertility. Hum Reprod. 2004;19(1):129–138.
  • Adeoye O, Olawumi J, Opeyemi A, et al. Review on the role of glutathione on oxidative stress and infertility. JBRA Assist Reprod. 2018;22(1):61–66.
  • Chen H, Cangello D, Benson S, et al. Age-related increase in mitochondrial superoxide generation in the testosterone-producing cells of brown Norway rat testes: relationship to reduced steroidogenic function? Exp Gerontol. 2001;36(8):1361–1373.
  • Cao L, Leers-Sucheta S, Azhar S. Aging alters the functional expression of enzymatic and non-enzymatic anti-oxidant defense systems in testicular rat Leydig cells. J Steroid Biochem Mol Biol. 2004;88(1):2682–2691.
  • Abidi P, Leers-Sucheta S, Azhar S. Suppression of steroidogenesis and activator protein-1 transcription factor activity in rat adrenals by vitamin E deficiency-induced chronic oxidative stress. J Nutr Biochem. 2004;15(4):210–219.
  • Chen H, Jin S, Guo J, et al. Knockout of the transcription factor Nrf2: effects on testosterone production by aging mouse Leydig cells. Mol Cell Endocrinol. 2015;409:113–120.
  • Shao L, Li H, Pazhanisamy SK, et al. Reactive oxygen species and hematopoietic stem cell senescence. Int J Hematol. 2011;94(1):24–32.
  • Small DM, Coombes JS, Bennett N, et al. Oxidative stress, anti-oxidant therapies and chronic kidney disease. Nephrology (Carlton). 2012;17(4):311–321.
  • Bulacio RP, Anzai N, Ouchi M, et al. Organic anion transporter 5 (Oat5) urinary excretion is a specific biomarker of kidney injury: evaluation of urinary excretion of exosomal Oat5 after N-Acetylcysteine prevention of cisplatin induced nephrotoxicity. Chem Res Toxicol. 2015;28(8):1595–1602.
  • Cazzola M, Calzetta L, Page C, et al. Influence of N-acetylcysteine on chronic bronchitis or COPD exacerbations: a meta-analysis. Eur Respir Rev. 2015;24(137):451–461.