36
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Assessment of the toxicity of different antiretroviral drugs and their combinations on Sertoli and Leydig cells

ORCID Icon, , , &
Received 29 Jan 2024, Accepted 25 Mar 2024, Published online: 22 Apr 2024

References

  • Adamczewska, D., Słowikowska-Hilczer, J., & Walczak-Jędrzejowska, R. (2022). The fate of Leydig cells in men with spermatogenic failure. Life, 12(4), 570 https://doi.org/10.3390/life12040570
  • Adana, M. Y., Akang, E. N., Peter, A. I., Jegede, A. I., Naidu, E. C. S., Tiloke, C., Chuturgoon, A. A., & Azu, O. O. (2018). Naringenin attenuates highly active antiretroviral therapy-induced sperm DNA fragmentations and testicular toxicity in Sprague-Dawley rats. Andrology, 6(1), 166–175. https://doi.org/10.1111/andr.12439
  • Aitken, R. J., & Roman, S. D. (2008). Antioxidant systems and oxidative stress in the testes. Oxidative Medicine and Cellular Longevity, 1(1), 15–24. https://doi.org/10.4161/OXIM.1.1.6843
  • Akay, C., Cooper, M., Odeleye, A., Jensen, B. K., White, M. G., Vassoler, F., Gannon, P. J., Mankowski, J., Dorsey, J. L., Buch, A. M., Cross, S. A., Cook, D. R., Peña, M.-M., Andersen, E. S., Christofidou-Solomidou, M., Lindl, K. A., Zink, M. C., Clements, J., Pierce, R. C., Kolson, D. L., & Jordan-Sciutto, K. L. (2014). Antiretroviral drugs induce oxidative stress and neuronal damage in the central nervous system. Journal of Neurovirology, 20(1), 39–53. https://doi.org/10.1007/s13365-013-0227-1
  • Akinjo, O. O., Gant, T. W., & Marczylo, E. L. (2018). Perturbation of microRNA signalling by doxorubicin in spermatogonial, Leydig and Sertoli cell lines in vitro. Toxicology Research, 7(5), 760–770. https://doi.org/10.1039/c7tx00314e
  • Awodele, O., Popoola, T. D., Idowu, O., Bashua, B. M., Awolola, N. A., & Okunowo, W. O. (2018). Investigations into the risk of reproductive toxicity following exposure to highly active anti-retroviral drugs in rodents. The Tokai Journal of Experimental and Clinical Medicine, 43(2), 54–63.
  • Azu, O. O. (2012). Highly active antiretroviral therapy (HAART) and testicular morphology: current status and a case for a stereologic approach. Journal of Andrology, 33(6), 1130–1142. https://doi.org/10.2164/jandrol.112.016758
  • Azu, O. O., Naidu, E. C. S., Naidu, J. S., Masia, T., Nzemande, N. F., Chuturgoon, A., & Singh, S. (2014). Testicular histomorphologic and stereological alterations following short-term treatment with highly active antiretroviral drugs (HAART) in an experimental animal model. Andrology, 2(5), 772–779. https://doi.org/10.1111/j.2047-2927.2014.00233.x
  • Barratt, C. L. R., De Jonge, C. J., Anderson, R. A., Eisenberg, M. L., Garrido, N., Rautakallio Hokkanen, S., Krausz, C., Kimmins, S., O’Bryan, M. K., Pacey, A. A., Tüttelmann, F., & Veltman, J. A. (2021). A global approach to addressing the policy, research and social challenges of male reproductive health. Human Reproduction Open, 2021(1), hoab009. https://doi.org/10.1093/hropen/hoab009
  • Baysal, M., & Atlı-Eklioğlu, Ö. (2021). Comparison of the toxicity of pure compounds and commercial formulations of imidacloprid and acetamiprid on HT-29 cells: Single and mixture exposure. Food and Chemical Toxicology , 155, 112430. https://doi.org/10.1016/j.fct.2021.112430
  • Brinkman, K., Smeitink, J. A., Romijn, J. A., & Reiss, P. (1999). Mitochondrial toxicity induced by nucleoside-analogue reverse-transcriptase inhibitors is a key factor in the pathogenesis of antiretroviral-therapy-related lipodystrophy. Lancet, 354(9184), 1112–1115. https://doi.org/10.1016/S0140-6736(99)06102-4
  • Chatterjee, N., & Walker, G. C. (2017). Mechanisms of DNA damage, repair, and mutagenesis. Environmental and Molecular Mutagenesis, 58(5), 235–263. https://doi.org/10.1002/em.22087
  • Chen-Chen, L., de Jesus Silva Carvalho, C., de Moraes Filho, A. V., Véras, J. H., Cardoso, C. G., Bailão, E., Spanó, M. A., & Cunha, K. S. (2019). Toxicity and genotoxicity induced by abacavir antiretroviral medication alone or in combination with zidovudine and/or lamivudine in Drosophila melanogaster. Human & Experimental Toxicology, 38(4), 446–454. https://doi.org/10.1177/0960327118818248
  • Cheney, L., Barbaro, J. M., & Berman, J. W. (2021). Antiretroviral drugs ımpact autophagy with toxic outcomes. Cells, 10(4), 909 https://doi.org/10.3390/cells10040909
  • Collin, F. (2019). Chemical basis of reactive oxygen species reactivity and ınvolvement in neurodegenerative diseases. International Journal of Molecular Sciences, 20(10), 2407. https://doi.org/10.3390/ijms20102407
  • De Jonge, C., & Barratt, C. L. R. (2019). The present crisis in male reproductive health: an urgent need for a political, social, and research roadmap. Andrology, 7(6), 762–768. https://doi.org/10.1111/andr.12673
  • de Moraes Filho, A. V., Carvalho, C. d J. S., Carneiro, C. C., Vale, C. R. d., Lima, D. C. d S., Carvalho, W. F., Vieira, T. B., Silva, D. d M. E., Cunha, K. S., & Chen-Chen, L. (2016). Genotoxic and cytotoxic effects of antiretroviral combinations in mice bone marrow. PLoS One, 11(11), e0165706. https://doi.org/10.1371/journal.pone.0165706
  • Deeks, S. G., Lewin, S. R., & Havlir, D. V. (2013). The end of AIDS: HIV infection as a chronic disease. Lancet, 382(9903), 1525–1533. https://doi.org/10.1016/S0140-6736(13)61809-7
  • Deng, W., Baki, L., Yin, J., Zhou, H., & Baumgarten, C. M. (2010). HIV protease inhibitors elicit volume-sensitive Cl- current in cardiac myocytes via mitochondrial ROS. Journal of Molecular and Cellular Cardiology, 49(5), 746–752. https://doi.org/10.1016/j.yjmcc.2010.08.013
  • Forrester, S. J., Kikuchi, D. S., Hernandes, M. S., Xu, Q., & Griendling, K. K. (2018). Reactive oxygen species in metabolic and ınflammatory signaling. Circulation Research, 122(6), 877–902. https://doi.org/10.1161/CIRCRESAHA.117.311401
  • Frungieri, M. B., Calandra, R. S., & Rossi, S. P. (2017). Local actions of melatonin in somatic cells of the testis. International Journal of Molecular Sciences, 18(6), 1170. https://doi.org/10.3390/ijms18061170
  • Fu, Z., Jiang, Z., Guo, G., Liao, X., Liu, M., & Xiong, Z. (2021). rhKGF-2 attenuates smoke ınhalation lung ınjury of rats via activating PI3K/Akt/Nrf2 and repressing FoxO1-NLRP3 inflammasome. Frontiers in Pharmacology, 12, 641308 https://doi.org/10.3389/fphar.2021.641308
  • Grando, A. C., Guimarães, N. N., de Souza, A. P., Lehmann, M., Cunha, K. S., & Dihl, R. R. (2020). Assessment of complex genomic alterations induced by AZT, 3TC, and the combination AZT +3TC. Drug and Chemical Toxicology, 43(4), 429–434. https://doi.org/10.1080/01480545.2018.1504959
  • Gratton, R., Tricarico, P. M., Guimaraes, R. L., Celsi, F., & Crovella, S. (2018). Lopinavir/ritonavir treatment ınduces oxidative stress and caspaseindependent apoptosis in human glioblastoma U-87 MG cell line. Current HIV Research, 16(2), 106–112. https://doi.org/10.2174/1570162X16666180528100922
  • Guimarães, N. N., Silva, C. J., de Andrade, H. H., Dihl, R. R., Lehmann, M., & Cunha, K. S. (2013). Comparative analysis of genetic toxicity of antiretroviral combinations in somatic cells of Drosophila melanogaster. Food and Chemical Toxicology, 53, 299–309. https://doi.org/10.1016/j.fct.2012.12.005
  • Guo, C., Sun, L., Chen, X., & Zhang, D. (2013). Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regeneration Research, 8(21), 2003–2014. https://doi.org/10.3969/j.issn.1673-5374.2013.21.009
  • Gurung, P., Yetiskul, E., & Jialal, I. (2023). Physiology, male reproductive system. In StatPearls. StatPearls Publishing.
  • Gurupriya, V. S., & Roy, S. C. (2017). Proteases and protease ınhibitors in male reproduction. In S. Chakraborti & N. Dhalla (Eds.), Proteases in physiology and pathology. Springer. https://doi.org/10.1007/978-981-10-2513-6_10
  • Ham, J., Lim, W., & Song, G. (2021). Pendimethalin induces apoptosis in testicular cells via hampering ER-mitochondrial function and autophagy. Environmental Pollution, 278, 116835. https://doi.org/10.1016/j.envpol.2021.116835
  • Ham, J., You, S., Lim, W., & Song, G. (2020). Bifenthrin impairs the functions of Leydig and Sertoli cells in mice via mitochondrion-endoplasmic reticulum dysregulation. Environmental Pollution, 266(Pt 3), 115174. https://doi.org/10.1016/j.envpol.2020.115174
  • Hong, X., Shao, N., Yin, L., Li, C., Tao, G., Sun, Y., Qian, K., Yang, J., Xiao, P., Yu, X., & Zhou, Z. (2022). Exposure to zinc oxide nanoparticles affects testicular structure, reproductive development and spermatogenesis in parental and offspring male rats. Annals of Translational Medicine, 10(13), 751–751. https://doi.org/10.21037/atm-22-3047
  • Imahashi, M., Ode, H., Kobayashi, A., Nemoto, M., Matsuda, M., Hashiba, C., Hamano, A., Nakata, Y., Mori, M., Seko, K., Nakahata, M., Kogure, A., Tanaka, Y., Sugiura, W., Yokomaku, Y., & Iwatani, Y. (2021). Impact of long-term antiretroviral therapy on gut and oral microbiotas in HIV-1-infected patients. Scientific Reports, 11(1), 960. https://doi.org/10.1038/s41598-020-80247-8
  • Jerónimo, A., Baza, M. B., Río, I., Vera, M., Hernando, V., Castilla, J., Rodriguez, C., & Del Romero, J. (2017). Factors associated with seminal impairment in HIV-infected men under antiretroviral therapy. Human Reproduction, 32(2), 265–271. https://doi.org/10.1093/humrep/dew321
  • Jiang, B., Hebert, V. Y., Li, Y., Mathis, J. M., Alexander, J. S., & Dugas, T. R. (2007). HIV antiretroviral drug combination induces endothelial mitochondrial dysfunction and reactive oxygen species production, but not apoptosis. Toxicology and Applied Pharmacology, 224(1), 60–71. https://doi.org/10.1016/j.taap.2007.06.010
  • Kashyap, S., Mukker, A., Gupta, D., Datta, P. K., Rappaport, J., Jacobson, J. M., Ebert, S. N., & Gupta, M. K. (2021). Antiretroviral drugs regulate epigenetic modification of cardiac cells through modulation of H3K9 and H3K27 acetylation. Frontiers in Cardiovascular Medicine, 8, 634774 https://doi.org/10.3389/fcvm.2021.634774
  • Kausar, S., Wang, F., & Cui, H. (2018). The role of mitochondria in reactive oxygen species generation and its implications for neurodegenerative diseases. Cells, 7(12), 274 https://doi.org/10.3390/cells7120274
  • Koppen, G., Azqueta, A., Pourrut, B., Brunborg, G., Collins, A. R., & Langie, S. A. S. (2017). The next three decades of the comet assay: A report of the 11th International Comet Assay Workshop. Mutagenesis, 32(3), 397–408. https://doi.org/10.1093/mutage/gex002
  • Lagathu, C., Eustace, B., Prot, M., Frantz, D., Gu, Y., Bastard, J.-P., Maachi, M., Azoulay, S., Briggs, M., Caron, M., & Capeau, J. (2007). Some HIV antiretrovirals increase oxidative stress and alter chemokine, cytokine or adiponectin production in human adipocytes and macrophages. Antiviral Therapy, 12(4), 489–500.
  • Lambertini, M., Del Mastro, L., Pescio, M. C., Andersen, C. Y., Azim, H. A., Jr., Peccatori, F. A., Costa, M., Revelli, A., Salvagno, F., Gennari, A., Ubaldi, F. M., La Sala, G. B., De Stefano, C., Wallace, W. H., Partridge, A. H., & Anserini, P. (2016). Cancer and fertility preservation: International recommendations from an expert meeting. BMC Medicine, 14(1), 1. https://doi.org/10.1186/s12916-015-0545-7
  • Latronico, T., Pati, I., Ciavarella, R., Fasano, A., Mengoni, F., Lichtner, M., Vullo, V., Mastroianni, C. M., & Liuzzi, G. M. (2018). In vitro effect of antiretroviral drugs on cultured primary astrocytes: Analysis of neurotoxicity and matrix metalloproteinase inhibition. Journal of Neurochemistry, 144(3), 271–284. https://doi.org/10.1111/jnc.14269
  • Le Magueresse-Battistoni, B. (2008). Proteases and their cognate inhibitors of the serine and metalloprotease subclasses, in testicular physiology. Advances in Experimental Medicine and Biology, 636, 133–153. https://doi.org/10.1007/978-0-387-09597-4_8
  • Lorenzo, Y., Costa, S., Collins, A. R., & Azqueta, A. (2013). The comet assay, DNA damage, DNA repair and cytotoxicity: Hedgehogs are not always dead. Mutagenesis, 28(4), 427–432. https://doi.org/10.1093/mutage/get018
  • Lü, J. M., Jiang, J., Jamaluddin, M. S., Liang, Z., Yao, Q., & Chen, C. (2019). Ginsenoside Rb1 blocks ritonavir-induced oxidative stress and eNOS downregulation through activation of estrogen receptor-beta and upregulation of SOD in human endothelial cells. International Journal of Molecular Sciences, 20(2), 294 https://doi.org/10.3390/ijms20020294
  • Lv, R., Li, G., Wu, J., Zhu, Y., Qin, X., & Li, S. (2016). Research on AIDS patients’ survival time after highly active antiretroviral therapy, treatment effect and treatment modes. Saudi Pharmaceutical Journal , 24(3), 318–321. https://doi.org/10.1016/j.jsps.2016.04.004
  • Malisa, J., Manak, M., Michelo, C., Imami, N., & Kibirige, C. N. (2023). Use of laboratory-developed assays in global HIV-1 treatment-monitoring and research. Scientific Reports, 13(1), 4578 https://doi.org/10.1038/s41598-023-31103-y
  • Margolis, A. M., Heverling, H., Pham, P. A., & Stolbach, A. (2014). A review of the toxicity of HIV Medications. Journal of Medical Toxicology , 10(1), 26–39. https://doi.org/10.1007/S13181-013-0325-8/TABLES/4
  • Martemucci, G., Costagliola, C., Mariano, M., D’andrea, L., Napolitano, P., & D’Alessandro, A. G. (2022). Free radical properties, source and targets, antioxidant consumption and health. Oxygen, 2(2), 48–78. https://doi.org/10.3390/oxygen2020006
  • Menéndez-Arias, L., & Delgado, R. (2022). Update and latest advances in antiretroviral therapy. Trends in Pharmacological Sciences, 43(1), 16–29. https://doi.org/10.1016/j.tips.2021.10.004
  • Meroni, S. B., Galardo, M. N., Rindone, G., Gorga, A., Riera, M. F., & Cigorraga, S. B. (2019). Molecular mechanisms and signaling pathways ınvolved in sertoli cell proliferation. Frontiers in Endocrinology, 10, 224 https://doi.org/10.3389/fendo.2019.00224
  • Monageng, E., Offor, U., Takalani, N. B., Mohlala, K., & Opuwari, C. S. (2023). A review on the impact of oxidative stress and medicinal plants on Leydig cells. Antioxidants, 12(8), 1559 https://doi.org/10.3390/antiox12081559
  • Moreno, S., Perno, C. F., Mallon, P. W., Behrens, G., Corbeau, P., Routy, J. P., & Darcis, G. (2019). Two-drug vs. three-drug combinations for HIV-1: Do we have enough data to make the switch? HIV Medicine, 20 Suppl 4, 2–12. https://doi.org/10.1111/hiv.12716
  • Murphy, M. P. (2009). How mitochondria produce reactive oxygen species. The Biochemical Journal, 417(1), 1–13. https://doi.org/10.1042/BJ20081386
  • Nastri, B. M., Pagliano, P., Zannella, C., Folliero, V., Masullo, A., Rinaldi, L., Galdiero, M., & Franci, G. (2023). HIV and drug-resistant subtypes. Microorganisms, 11(1), 221. https://doi.org/10.3390/microorganisms11010221
  • Ogedengbe, O. O., Jegede, A. I., Onanuga, I. O., Offor, U., Naidu, E. C. S., Peter, A. I., & Azu, O. O. (2016). Coconut oil extract mitigates testicular injury following adjuvant treatment with antiretroviral drugs. Toxicological Research, 32(4), 317–325. https://doi.org/10.5487/TR.2016.32.4.317
  • Osmaniye, D., Levent, S., Karaduman, A. B., Ilgın, S., Özkay, Y., & Kaplancıklı, Z. A. (2018). Synthesis of new benzothiazole acylhydrazones as anticancer agents. Molecules, 23(5), 1054. https://doi.org/10.3390/molecules23051054
  • Park, S., Park, M. Y., Song, G., & Lim, W. (2019). Alpha-solanine inhibits cell proliferation via mitochondrial dysfunction and inhibin synthesis in mouse testis in vitro and in vivo. Chemosphere, 235, 271–279. https://doi.org/10.1016/j.chemosphere.2019.06.172
  • Petersen, C., & Soder, O. (2006). The sertoli cell–A hormonal target and ‘super’ nurse for germ cells that determines testicular size. Hormone Research, 66(4), 153–161. https://doi.org/10.1159/000094142
  • Pilatz, A., Discher, T., Lochnit, G., Wolf, J., Schuppe, H.-C., Schüttler, C. G., Hossain, H., Weidner, W., Lohmeyer, J., & Diemer, T. (2014). Semen quality in HIV patients under stable antiretroviral therapy is impaired compared to WHO 2010 reference values and on sperm proteome level. AIDS, 28(6), 875–880. https://doi.org/10.1097/QAD.0000000000000161
  • Richman, D. D., Margolis, D. M., Delaney, M., Greene, W. C., Hazuda, D., & Pomerantz, R. J. (2009). The challenge of finding a cure for HIV infection. Science, 323(5919), 1304–1307. https://doi.org/10.1126/science.1165706
  • Rudolph-Owen, L. A., Cannon, P., & Matrisian, L. M. (1998). Overexpression of the matrix metalloproteinase matrilysin results in premature mammary gland differentiation and male infertility. Molecular Biology of the Cell, 9(2), 421–435. https://doi.org/10.1091/MBC.9.2.421/ASSET/IMAGES/LARGE/MK0280519008.JPEG
  • Saldutti, L. P., Beyer, B. K., Breslin, W., Brown, T. R., Chapin, R. E., Campion, S., Enright, B., Faustman, E., Foster, P. M., Hartung, T., Kelce, W., Kim, J. H., Loboa, E. G., Piersma, A. H., Seyler, D., Turner, K. J., Yu, H., Yu, X., & Sasaki, J. C. (2013). In vitro testicular toxicity models: Opportunities for advancement via biomedical engineering techniques. Altex, 30(3), 353–377. https://doi.org/10.14573/altex.2013.3.353
  • Savasi, V., Oneta, M., Laoreti, A., Parisi, F., Parrilla, B., Duca, P., & Cetin, I. (2018). Effects of antiretroviral therapy on sperm DNA integrity of HIV-1-infected men. American Journal of Men’s Health, 12(6), 1824–1831. https://doi.org/10.1177/1557988318794282
  • Semet, M., Paci, M., Saïas-Magnan, J., Metzler-Guillemain, C., Boissier, R., Lejeune, H., & Perrin, J. (2017). The impact of drugs on male fertility: A review. Andrology, 5(4), 640–663. https://doi.org/10.1111/andr.12366
  • Stanley, E., Lin, C. Y., Jin, S., Liu, J., Sottas, C. M., Ge, R., Zirkin, B. R., & Chen, H. (2012). Identification, proliferation, and differentiation of adult Leydig stem cells. Endocrinology, 153(10), 5002–5010. https://doi.org/10.1210/en.2012-1417
  • Thein, P., Kalinec, G. M., Park, C., & Kalinec, F. (2014). In vitro assessment of antiretroviral drugs demonstrates potential for ototoxicity. Hearing Research, 310, 27–35. https://doi.org/10.1016/J.HEARES.2014.01.005
  • Tice, R. R., Agurell, E., Anderson, D., Burlinson, B., Hartmann, A., Kobayashi, H., Miyamae, Y., Rojas, E., Ryu, J.-C., & Sasaki, Y. F. (2000). Single cell gel/comet assay: Guidelines for in vitro and in vivo genetic toxicology testing. Environmental and Molecular Mutagenesis, 35(3), 206–221. https://doi.org/10.1002/(SICI)1098-2280(2000)35:3<206::AID-EM8>3.0.CO;2-J
  • Tirichen, H., Yaigoub, H., Xu, W., Wu, C., Li, R., & Li, Y. (2021). Mitochondrial reactive oxygen species and their contribution in chronic kidney disease progression through oxidative stress. Frontiers in Physiology, 12, 627837 https://doi.org/10.3389/fphys.2021.627837
  • Tong, N., Witherspoon, L., Dunne, C., & Flannigan, R. (2022). Global decline of male fertility: Fact or fiction? BC Medical Journal, 64(3), 126–130.
  • Ugwu, C. L. J., & Ncayiyana, J. R. (2022). Spatial disparities of HIV prevalence in South Africa. Do sociodemographic, behavioral, and biological factors explain this spatial variability? Frontiers in Public Health, 10, 994277 https://doi.org/10.3389/fpubh.2022.994277
  • Wei, J., Lu, T., Dong, F., Zhang, C., & Zhang, Y. (2021). Gene expression profiles of two testicular somatic cell lines respond differently to 4-nitrophenol mediating varied reproductive toxicity. Toxicology, 463, 152991. https://doi.org/10.1016/j.tox.2021.152991
  • Weinbauer, G. F., Luetjens, C. M., Simoni, M., & Nieschlag, E. (2010). Physiology of testicular function. In E. Nieschlag, H. M. Behre, & S. Nieschlag, (Eds.), Andrology. Springer. https://doi.org/10.1007/978-3-540-78355-8_2
  • White, A. J. (2001). Mitochondrial toxicity and HIV therapy. Sexually Transmitted İnfections, 77(3), 158–173. https://doi.org/10.1136/STI.77.3.158
  • Wu, D., Huang, C. J., Jiao, X. F., Ding, Z. M., Zhang, J. Y., Chen, F., Wang, Y. S., Li, X., & Huo, L. J. (2017). Olaquindox disrupts tight junction integrity and cytoskeleton architecture in mouse Sertoli cells. Oncotarget, 8(51), 88630–88644. https://doi.org/10.18632/oncotarget.20289
  • Xu, P., Wang, Y., Qin, Z., Qiu, L., Zhang, M., Huang, Y., & Zheng, J. C. (2017). Combined medication of antiretroviral drugs tenofovir disoproxil fumarate, emtricitabine, and raltegravir reduces neural progenitor cell proliferation ın vivo and ın vitro. Journal of Neuroimmune Pharmacology, 12(4), 682–692. https://doi.org/10.1007/s11481-017-9755-4
  • Xu, Y., Shrestha, N., Préat, V., & Beloqui, A. (2021). An overview of in vitro, ex vivo and in vivo models for studying the transport of drugs across intestinal barriers. Advanced Drug Delivery Reviews, 175, 113795. https://doi.org/10.1016/j.addr.2021.05.005
  • Yan, X., Du, J., & Ji, G. (2021). Prevalence and factors associated with fertility desire among people living with HIV: A systematic review and meta-analysis. PloS One, 16(3), e0248872. https://doi.org/10.1371/journal.pone.0248872
  • Yuan, Z., Matias, F. B., Yi, J. E., & Wu, J. (2016). T-2 toxin-induced cytotoxicity and damage on TM3 Leydig cells. Comparative biochemistry and physiology. Comparative Biochemistry and Physiology. Toxicology & Pharmacology , 181–182, 47–54. https://doi.org/10.1016/j.cbpc.2015.12.005
  • Zirkin, B. R., & Papadopoulos, V. (2018). Leydig cells: Formation, function, and regulation. Biology of Reproduction, 99(1), 101–111. https://doi.org/10.1093/biolre/ioy059

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.