Generic placeholder image

Current HIV Research

Editor-in-Chief

ISSN (Print): 1570-162X
ISSN (Online): 1873-4251

Research Article

Lopinavir/Ritonavir Treatment Induces Oxidative Stress and Caspaseindependent Apoptosis in Human Glioblastoma U-87 MG Cell Line

Author(s): Rossella Gratton*, Paola Maura Tricarico, Rafael Lima Guimaraes, Fulvio Celsi and Sergio Crovella

Volume 16, Issue 2, 2018

Page: [106 - 112] Pages: 7

DOI: 10.2174/1570162X16666180528100922

Price: $65

Abstract

Background: Lopinavir and Ritonavir (LPV/r) treatment is widely used to prevent HIV mother-to-child transmission. Nevertheless, studies related to the impact of these compounds on patients, in particular in the foetus and newborns, are strictly required due to the controversial findings reported in the literature concerning possible neurologic side effects following the administration of these drugs.

Objectives: In our study, we evaluated the impact of LPV/r treatment on the human glioblastoma U- 87 MG cell line.

Methods: In order to evaluate the influence of Lopinavir and Ritonavir in terms of oxidative stress (ROS production), mitochondrial morphology and apoptotic cell death, the latter either in the presence or in the absence of caspase-3 and -9 inhibitors, we treated U-87 MG with increasing doses (0.1-1-10-25-50 µM) of Lopinavir and Ritonavir for 24h, either in single formulation or in combination. ROS production was measured by flow cytometry using H2DCFDA dye, mitochondrial morphology was evaluated using MitoRed dye and apoptotic cell death was monitored by flow cytometry using Annexin V-FITC and Propidium Iodide.

Results: We observed that co-treatment with Lopinavir and Ritonavir (25 and 50 µM) promoted a significant increase in ROS production, caused mitochondrial network damage and induced apoptosis in a caspase-independent manner.

Conclusion: Based on our findings, concordant with others reported in the literature, we hypothesize that LPV/r treatment could not be entirely free from side effects, being aware of the need of validation in in vivo models, necessary to confirm our results.

Keywords: Apoptosis, caspase-independent apoptosis, mitochondrial damage, protease inhibitors, ROS, U87-MG glioblastoma cell line.

Next »
Graphical Abstract
[1]
Chougrani I, Luton D, Matheron S, Mandelbrot L, Azria E. Safety of protease inhibitors in HIV-infected pregnant women. HIV AIDS 2013; 5: 253-62.
[2]
Chandwani A, Shuter J. Lopinavir/ritonavir in the treatment of HIV-1 infection: a review. Ther Clin Risk Manag 2008; 4: 1023-33.
[3]
Potard V, Rey D, Poizot-Martin I, et al. Lopinavir/r no longer recommended as a first-line regimen: a comparative effectiveness analysis. J Int AIDS Soc 2014; 17: 19070.
[4]
Heidari S, Mofenson L, Cotton MF, Marlink R, Cahn P, Katabira E. Antiretroviral drugs for preventing mother-to-child transmission of HIV: a review of potential effects on HIV-exposed but uninfected children. J Acquir Immune Defic Syndr 2011; 57: 290-6.
[5]
Tubiana R, Mandelbrot L, Le Chenadec J, et al. Lopinavir/ritonavir monotherapy as a nucleoside analogue-sparing strategy to prevent HIV-1 mother-to-child transmission: the ANRS 135 PRIMEVA phase 2/3 randomized trial. Clin Infect Dis 2013; 57: 891-902.
[6]
Decloedt EH, Maartens G. Neuronal toxicity of efavirenz: A systematic review. Expert Opin Drug Saf 2013; 12: 841-6.
[7]
Purnell PR, Fox HS. Efavirenz induces neuronal autophagy and mitochondrial alterations. J Pharmacol Exp Ther 2014; 351: 250-8.
[8]
Clarke DF, Penazzato M, Capparelli E, et al. Prevention and treatment of HIV infection in neonates: evidence based for existing WHO dosing recommendations and implementation considerations. Expert Rev Clin Pharmacol 2018; 11(1): 83-93.
[9]
Cotton MF, Holgate S, Nelson A, Rabie H, Wedderburn C, Mirochnick M. The last and first frontier–emerging challenges for HIV treatment and prevention in the first week of life with emphasis on premature and low birth weight infants. J Int AIDS Soc 2015; 18: 20271.
[10]
Stek AM, Mirochnick M, Capparelli E, et al. Reduced lopinavir exposure during pregnancy. AIDS 2006; 20(15): 1931-9.
[11]
McCormack SA, Best BM. Protecting the fetus against HIV infection: A systematic review of placental transfer of antiretrovirals. Clin Pharmacokinet 2014; 53: 989-1004.
[12]
Tookey PA, Thorne C, van Wyk J, Norton M. Maternal and foetal outcomes among 4118 women with HIV infection treated with lopinavir/ritonavir during pregnancy: analysis of population-based surveillance data from the national study of HIV in pregnancy and childhood in the United Kingdom and Ireland. BMC Infect Dis 2016; 16: 65.
[13]
Coelho AV, Tricarico PM, Celsi F, Crovella S. Antiretroviral treatment in HIV-1-positive mothers: Neurological implications in virus-free children. Int J Mol Sci 2017; 18(2)
[14]
Kim MJ, Kim SW, Chang HH, et al. Comparison of Antiretroviral Regimens: Adverse Effects and Tolerability Failure that Cause Regimen Switching. Infect Chemother 2015; 47: 231-8.
[15]
Zhou H, Pandak WM, Lyall V, Natarajan R, Hylemon PB. HIV protease inhibitors activate the unfolded protein response in macrophages: implication for atherosclerosis and cardiovascular disease. Mol Pharmacol 2005; 68(3): 690-700.
[16]
Gupta S, Knight AG, Losso BY, Ingram DK, Keller JN, Bruce-Keller AJ. Brain injury caused by HIV protease inhibitors: role of lipodystrophy and insulin resistance. Antiviral Res 2012; 95: 19-29.
[17]
Tricarico PM, de Oliveira Franca RF, Pacor S, Crovella S, Celsi F. HIV protease inhibitors apoptotic effect in SH-SY5Y neuronal cell line. Cell Physiol Biochem 2016; 39: 1463-70.
[18]
Ngoma MS, Hunter JA, Harper JA, et al. Cognitive and language outcomes in HIV-uninfected infants exposed to combined antiretroviral therapy in utero and through extended breast-feeding. AIDS 2014; 28(Suppl. 3): S323-30.
[19]
Bruce-Keller AJ, Keller JN, Morrison CD. Obesity and vulnerability of the CNS. Biochim Biophys Acta 2009; 1792: 395-400.
[20]
Valcour V, Maki P, Bacchetti P, et al. Insulin Resistance and Cognition among HIV-infected and HIV-uninfected Adult Women - The Women’s Interagency HIV Study. AIDS Res Hum Retroviruses 2012; 28: 447-53.
[21]
Wu X, Sun L, Zha W, et al. (2010). HIV protease inhibitors induce endoplasmic reticulum stress and disrupt barrier integrity in intestinal epithelial cells. Gastroenterology 2010; 138: 197-209.
[22]
Deng W, Baki L, Yin J, Zhou H, Baumgarten CM. HIV protease inhibitors elicit volume-sensitive Cl- current in cardiac myocytes via mitochondrial ROS. J Mol Cell Cardiol 2010; 49: 746-52.
[23]
Taura M, Kariya R, Kudo E, et al. Comparative analysis of ER stress response into HIV protease inhibitors: lopinavir but not darunavir induces potent ER stress response via ROS/JNK pathway. Free Radic Biol Med 2013; 65: 778-88.
[24]
Estaquier J, Lelièvre JD, Petit F, et al. Effects of antiretroviral drugs on human immunodeficiency virus type 1-induced CD4(C) T-cell death. J Virol 2002; 76: 5966-73.
[25]
Vivithanaporn P, Asahchop EL, Acharjee S, Baker GB, Power C. HIV protease inhibitors disrupt astrocytic glutamate transporter function and neurobehavioral performance. AIDS 2016; 30: 543-52.
[26]
Retallack H, Di Lullo E, Arias C, et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc Natl Acad Sci USA 2016; 113(50): 14408-13.
[27]
Nowakowsky TJ, Pollen AA, Di Lullo E, Sandoval-Espinosa C, Bershteyn M, Kriegstein AR. Expression analysis highlights AXL as a candidate Zika virus entry receptor in human neural stem cells. Cell Stem Cell 2016; 18(5): 591-6.
[28]
Woulfe J, Gray DA, Mackenzie IR. FUS-immunoreactive intranuclear inclu- sions in neurodegenerative disease. Brain Pathol 2010; 20: 589-97.
[29]
Li Y, Cheng D, Cheng R, et al. Mechanisms of U87 astrocytoma cell uptake and trafficking of monomeric versus protofibril Alzheimer’s disease amyloid-β proteins. PLoS One 2014; 9: e99939.
[30]
Zabłocka A, Mitkiewicz M, Macała J, Janusz M. Neurotrophic activity of cultured cell line U87 is up-regulated by proline-rich polypeptide complex and its constituent nonapeptide. Cell Mol Neurobiol 2015; 35: 977-86.
[31]
Lan X, Kiyota T, Hanamsagar R, et al. The effect of HIV protease inhibitors on amyloid-beta peptide degradation and synthesis in human cells and Alzhaimer’s disease animal model. J Neuroimmune Pharmacol 2012; 7: 412-23.
[32]
Arend C, Brandmann M, Dringen R. The antiretroviral protease inhibitor ritonavir accelerates glutathione export from culture primary astrocytes. Neurochem Res 2013; 38: 732-41.
[33]
Emmett SD, Cunningham CK, Mmbaga BT, et al. Predicting virologic failure among HIV-1 infected children receiving antiretroviral therapy in Tanzania: a cross-sectional study. J Acquir Immune Defic Syndr 2010; 54(4): 368-75.
[34]
Wang L, Liu L, Shi Y, et al. Berberine induces caspase-independent cell death in colon tumor cells through activation of apoptosis-inducing factor. PLoS One 2012; 7: e36418.
[35]
Liu G, Zou H, Luo T, et al. Caspase-dependent and caspase-independent pathways are involved in cadmium-induced apoptosis in primary rat proximal tubular cell culture. Plos One 2016; 11(11): e 0166823.
[36]
Kumar S, Eroglu E, Stokes JA, et al. Resveratrol induces mitochondria-mediated, caspase-independent apoptosis in murine prostate cancer cells. Oncotarget 2017; 8(13): 20895-908.
[37]
Wang CC, Fang KM, Yang CS, Tzeng SF. Reactive oxygen species-induced cell death of rat primary astrocytes through mitochondria-mediated mechanism. J Cell Biochem 2009; 107: 933-43.
[38]
Retallack H, Di Lullo E, Arias C, et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc Natl Acad Sci USA 2016; 113: 14408-13.
[39]
Saeed Y, Xie B, Xu J, et al. Glial U87 cells protect neuronal SH-SY5Y cells from indirect effect of radiation by reducing oxidative stress and apoptosis. Acta Biochim Biophys Sin 2015; 47: 250-7.
[40]
Li Y, Cheng D, Cheng R, et al. Mechanisms of U87 astrocytoma cell uptake and trafficking of monomeric versus protofibril alzheimer’s disease amyloid-β proteins. PLoS One 2014; 9: e99939.
[41]
Liu SJ, Nowakowsky TJ, Pollen AA, et al. Single-cell analysis of long non-coding RNAs in the developing human neocortex. Genome Biol 2016; 17: 67.
[42]
Retallack H, Di Lullo E, Arias C, et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc Natl Acad Sci USA 2016; 113(50): 14408-13.
[43]
Nowakowsky TJ, Pollen AA, Di Lullo E, Sandoval-Espinosa C, Bershteyn M, Kriegstein AR. Expression analysis highlights AXL as a candidate Zika virus entry receptor in human neural stem cells. Cell Stem Cell 2016; 18(5): 591-6.
[44]
Saunders NR, Liddelow SA, Dziegielewska KM. Barrier mechanisms in the developing brain. Front Pharmacol 2012; 3: 46.
[45]
Saunders NR, Dreifuss JJ, Dziegielewska KM, et al. The rights and wrongs of blood-brain barrier permeability studies: a walk through 100 years of history. Front Neurosci 2014; 8: 404.
[46]
Adinolfi M, Beck SE, Haddad SA, Seller MJ. Permeability of the blood–cerebrospinal fluid barrier to plasma proteins during foetal and perinatal life. Nature 1976; 259: 140-1.
[47]
Evers S, Rahmann A, Schwaag S, Frese A, Reichelt D, Husstedt IW. Prevention of AIDS dementia by HAART does not depend on cerebrospinal fluid drug penetrance. AIDS Res Hum Retroviruses 2004; 20: 483-91.
[48]
Strazza M, Pirrone V, Wigdhal B, Nonnemacher MR. Breaking down the barrier: the effects of HIV-1 on the blood-brain barrier. Brain Res 2011; 1399: 96-115.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy