243
Views
10
CrossRef citations to date
0
Altmetric
Original Research

Acute Damage to the Sperm Quality and Spermatogenesis in Male Mice Exposed to Curcumin-Loaded Nanoparticles

, , , &
Pages 1853-1862 | Published online: 17 Mar 2020

References

  • Aggarwal BB, Sundaram C, Malani N, Ichikawa H. Curcumin: the Indian solid gold. Adv Exp Med Biol. 2007;595:1–75.17569205
  • Kotha RR, Luthria DL. Curcumin: biological, pharmaceutical, nutraceutical, and analytical aspects. Molecules. 2019;24(16):E2930. doi:10.3390/molecules2416293031412624
  • Di Martino RM, Luppi B, Bisi A, et al. Recent progress on curcumin-based therapeutics: a patent review (2012–2016). Part I: curcumin. Expert Opin Ther Pat. 2017;27(5):579–590. doi:10.1080/13543776.2017.127656628024125
  • Bisht S, Maitra A. Systemic delivery of curcumin: 21st century solutions for an ancient conundrum. Curr Drug Discov Technol. 2009;6(3):192–199. doi:10.2174/15701630978905493319496751
  • Liu W, Zhai Y, Heng X, et al. Oral bioavailability of curcumin: problems and advancements. J Drug Target. 2016;24(8):694–702. doi:10.3109/1061186X.2016.115788326942997
  • Tan BL, Norhaizan ME. Curcumin combination chemotherapy: the implication and efficacy in cancer. Molecules. 2019;24(14):E2527. doi:10.3390/molecules2414252731295906
  • Yavarpour-bali H, Ghasemi-kasman M, Pirzadeh M. Curcumin-loaded nanoparticles: a novel therapeutic strategy in treatment of central nervous system disorders. Int J Nanomedicine. 2019;14:4449–4460. doi:10.2147/IJN.S20833231417253
  • Mohebbati R, Anaeigoudari A, Khazdair MR. The effects of Curcuma longa and curcumin on reproductive systems. Endocr Regul. 2017;51(4):220–228. doi:10.1515/enr-2017-002429232190
  • Naz RK. Can curcumin provide an ideal contraceptive? Mol Reprod Dev. 2011;78(2):116–123. doi:10.1002/mrd.v78.221337449
  • Naz RK. The effect of curcumin on intracellular pH (pHi), membrane hyperpolarization and sperm motility. J Reprod Infertil. 2014;15(2):62–70.24918078
  • Naz RK, Lough ML. Curcumin as a potential non-steroidal contraceptive with spermicidal and microbicidal properties. Eur J Obstet Gynecol Reprod Biol. 2014;176:142–148. doi:10.1016/j.ejogrb.2014.01.02424702904
  • Rithaporn T, Monga M, Rajasekaran M. Curcumin: a potential vaginal contraceptive. Contraception. 2003;68(3):219–223. doi:10.1016/S0010-7824(03)00163-X14561543
  • Murphy CJ, Tang H, Van Kirk EA, Shen Y, Murdoch WJ. Reproductive effects of a pegylated curcumin. Reprod Toxicol. 2012;34(1):120–124. doi:10.1016/j.reprotox.2012.04.00522576113
  • Moshari S, Nejati V, Najafi G, Razi M. Nanomicelle curcumin-induced DNA fragmentation in testicular tissue; Correlation between mitochondria dependent apoptosis and failed PCNA-related hemostasis. Acta Histochem. 2017;119(4):372–381. doi:10.1016/j.acthis.2017.03.00728385400
  • Moshari S, Nejati V, Najafi G, Razi M. Insight into curcumin nanomicelle-induced derangements in male reproduction potential: an experimental study. Andrologia. 2018;50(2):e12842. doi:10.1111/and.12842
  • Nakamura K, Yasunaga Y, Segawa T, et al. Curcumin down-regulates AR gene expression and activation in prostate cancer cell lines. Int J Oncol. 2002;21(4):825–830.12239622
  • Hu GX, Liang G, Chu Y, et al. Curcumin derivatives inhibit testicular 17beta-hydroxysteroid dehydrogenase 3. Bioorg Med Chem Lett. 2010;20(8):2549–2551. doi:10.1016/j.bmcl.2010.02.08920346654
  • Ide H, Lu Y, Noguchi T, et al. Modulation of AKR1C2 by curcumin decreases testosterone production in prostate cancer. Cancer Sci. 2018;109(4):1230–1238. doi:10.1111/cas.2018.109.issue-429369461
  • Lin YC, Chiu CH, Liu HC, Wang JY. Curcumin downregulates 8-br-cAMP-induced steroidogenesis in mouse Leydig cells by suppressing the expression of Cyp11a1 and StAR independently of the PKA-CREB pathway. Endocr J. 2018;65(8):833–840. doi:10.1507/endocrj.EJ18-001029887570
  • Xie X, Tao Q, Zou Y, et al. PLGA nanoparticles improve the oral bioavailability of curcumin in rats: characterizations and mechanisms. J Agric Food Chem. 2011;59(17):9280–9289. doi:10.1021/jf202135j21797282
  • Hofmann MC, Braydich-stolle L, Dettin L, Johnson E, Dym M. Immortalization of mouse germ line stem cells. Stem Cells. 2005;23(2):200–210. doi:10.1634/stemcells.2003-003615671143
  • Fan Y, Liu Y, Xue K, et al. Diet-induced obesity in male C57BL/6 mice decreases fertility as a consequence of disrupted blood-testis barrier. PLoS One. 2015;10(4):e0120775. doi:10.1371/journal.pone.012077525886196
  • Noorafshan A. Stereology as a valuable tool in the toolbox of testicular research. Ann Anat. 2014;196(1):57–66. doi:10.1016/j.aanat.2012.07.00823069191
  • Ma L, Guo Y, Yuan Y, Li YG, Deng XZ, Yang ZW. Morphometric study of the testis and reproductive tract (including sperm granuloma) after vasectomy in mature rats. Asian J Androl. 2016;18(1):66–73. doi:10.4103/1008-682X.15003825791731
  • Ahmed EA, de Rooij DG. Staging of mouse seminiferous tubule cross-sections. Methods Mol Biol. 2009;558:263–277.19685330
  • Xia X, Cai H, Qin S, Xu C. Histone acetylase inhibitor curcumin impairs mouse spermiogenesis-an in vitro study. PLoS One. 2012;7(11):e48673. doi:10.1371/journal.pone.004867323144926
  • Kim YS, Park JS, Park M, et al. PLGA nanoparticles with multiple modes are a biologically safe nanocarrier for mammalian development and their offspring. Biomaterials. 2018;183:43–53. doi:10.1016/j.biomaterials.2018.08.04230149229
  • Tiwari SK, Agarwal S, Seth B, et al. Curcumin-loaded nanoparticles potently induce adult neurogenesis and reverse cognitive deficits in Alzheimer’s disease model via canonical Wnt/beta-catenin pathway. ACS Nano. 2014;8(1):76–103. doi:10.1021/nn405077y24467380
  • Hansen DA, Esakky P, Drury A, Lamb L, Moley KH. The aryl hydrocarbon receptor is important for proper seminiferous tubule architecture and sperm development in mice. Biol Reprod. 2014;90(1):8. doi:10.1095/biolreprod.113.10884524174576
  • Balasubramanyam K, Varier RA, Altaf M, et al. Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase-dependent chromatin transcription. J Biol Chem. 2004;279(49):51163–51171. doi:10.1074/jbc.M40902420015383533
  • Kang J, Chen J, Shi Y, Jia J, Zhang Y. Curcumin-induced histone hypoacetylation: the role of reactive oxygen species. Biochem Pharmacol. 2005;69(8):1205–1213. doi:10.1016/j.bcp.2005.01.01415794941
  • Johnson L, Thompson DL, Varner DD. Role of Sertoli cell number and function on regulation of spermatogenesis. Anim Reprod Sci. 2008;105(1–2):23–51. doi:10.1016/j.anireprosci.2007.11.02918242891
  • Larson JK, Carvan MJ, Hutz RJ. Engineered nanomaterials: an emerging class of novel endocrine disruptors. Biol Reprod. 2014;91(1):20. doi:10.1095/biolreprod.113.11624424899576
  • Brohi RD, Wang L, Talpur HS, et al. Toxicity of nanoparticles on the reproductive system in animal models: a review. Front Pharmacol. 2017;8:606. doi:10.3389/fphar.2017.0060628928662
  • Ema M, Okuda H, Gamo M, Honda K. A review of reproductive and developmental toxicity of silver nanoparticles in laboratory animals. Reprod Toxicol. 2017;67:149–164. doi:10.1016/j.reprotox.2017.01.00528088501
  • Wang R, Song B, Wu J, Zhang Y, Chen A, Shao L. Potential adverse effects of nanoparticles on the reproductive system. Int J Nanomedicine. 2018;13:8487–8506. doi:10.2147/IJN30587973
  • Ahmed-farid OAH, Nasr M, Ahmed RF, Bakeer RM. Beneficial effects of curcumin nano-emulsion on spermatogenesis and reproductive performance in male rats under protein deficient diet model: enhancement of sperm motility, conservancy of testicular tissue integrity, cell energy and seminal plasma amino acids content. J Biomed Sci. 2017;24(1):66.28865467
  • Alizadeh F, Javadi M, Karami AA, Gholaminejad F, Kavianpour M, Haghighian HK. Curcumin nanomicelle improves semen parameters, oxidative stress, inflammatory biomarkers, and reproductive hormones in infertile men: a randomized clinical trial. Phytother Res. 2018;32(3):514–521. doi:10.1002/ptr.v32.329193350
  • Zindy F, den Besten W, Chen B, et al. Control of spermatogenesis in mice by the cyclin D-dependent kinase inhibitors p18(Ink4c) and p19(Ink4d). Mol Cell Biol. 2001;21(9):3244–3255. doi:10.1128/MCB.21.9.3244-3255.200111287627
  • Qian MX, Pang Y, Liu CH, et al. Acetylation-mediated proteasomal degradation of core histones during DNA repair and spermatogenesis. Cell. 2013;153(5):1012–1024.23706739
  • Goudarzi A, Shiota H, Rousseaux S, Khochbin S. Genome-scale acetylation-dependent histone eviction during spermatogenesis. J Mol Biol. 2014;426(20):3342–3349. doi:10.1016/j.jmb.2014.02.02324613302