GLUTATHIONE: INDUCTION OF APOPTOSIS AND AUTOPHAGY IN CANCER
HTML Full TextGLUTATHIONE: INDUCTION OF APOPTOSIS AND AUTOPHAGY IN CANCER
Smriti Gupta and Alakh N. Sahu *
Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi - 221005, Uttar Pradesh, India.
ABSTRACT: Glutathione (GSH) is the most abundant non-protein thiol in eukaryotic cells capable of carrying out antioxidant defense mechanisms in the cell for its survivability, mostly against free radicals, i.e., Reactive Oxygen Species (ROS). GSH protects normal cells against carcinogenic transformation, but high GSH levels in cancer cells decrease sensitivity to chemo- and radiotherapy, producing resistant cases of cancer. Hence, GSH depletion could be a potential mechanism by which resistant tumor cells can be sensitized to undergo apoptosis and autophagy. Thiol oxidation leads to the formation of permeability transition pore, promoting the extrusion of death-related molecular signals and activation of the intrinsic apoptotic pathway. Similarly, macroautophagic cell death involves the formation of auto-phagosomes that degrade organelles, including mitochondria, compromising normal cellular function and cellular death occurs by mechanisms such as mTorC1 inhibition. Therefore, the selective depletion of GSH in cancer cells could offer a promising approach in drug and radiation-resistant cases targeting multiple death-related cellular pathways. However, the selective depletion of such ubiquitous antioxidant, GSH in cancer cells yet remains to be a key challenge to the researchers worldwide.
Keywords: |
Glutathione, Apoptosis, Autophagy, mTorC1, Cancer
INTRODUCTION:
Glutathione and Cell Homeostasis: Cells respond to various metabolic and internal stressors through highly regulated pathways. One such stressor is free radicals such as reactive oxygen species (ROS) including superoxide anions (O2•−), hydroxyl radicals (OH•−), and peroxide radicals (ROO•) sources being mitochondria 1, 2. In mitochondria, the electron transport chain (ETC) produces ROS as an inevitable by-product 3. Complex I (NADH: ubiquinone oxidoreductase) and the mitochondrial cytochrome bc1 complex (complex III; ubiquinol:
cytochrome c oxidoreductase) are the main producers of superoxide which are released into the intermembrane space (approx. 80% of the generated superoxide) or the mitochondrial matrix (approx. 20%) within the mitochondrial respiratory chain 4. Superoxide is leaked into the cytoplasm through the mitochondrial permeability transition pore (MPTP) in the outer membrane of the mitochondria 5. H2O2 reacts with superoxide dismutase and inactivates it either in the mitochondrial matrix by MnSod or in the cytosol by Cu/ZnSOD 6, 7, 8.
Generally, the toxic cell oxidants are counteracted and neutralized using several defense strategies which include small reducing molecules maintained at high levels in the cells such as glutathione and ascorbate and several enzymes, such as superoxide dismutase, catalase, and other peroxidases, that further reduce ROS to water 9, 10.
GSH is an ubiquitous molecule, produced intra-cellularly, with 85-90% freely localized in the cytoplasm. Cytosolic GSH can also be found compartmentalized in several organelles that are subjected to higher oxidative stress, including the mitochondria, the peroxisomes, the nuclear matrix, and the endoplasmic reticulum (ER) 11. Mammalian cellular machinery maintains homeostatic pools of GSH via three different pathways, namely, de novo biosynthesis, uptake across plasma membrane of GSH derived from exogenous sources, and NADPH-dependent reduction of oxidized GSH (GSSG) through GSSG reductase-dependent reduction of oxidized GSH (GSSG) through GSSG reductase 12, 13.
Three amino acids, Cysteine, glutamic acid, and glycine, linked linearly by a ⍺-peptide and a ⍺-peptide bond form a tripeptide, glutathione, which is produced mainly by the liver to counteract the redox stress due to cells metabolic machinery 14, 15. GSH synthesis is catalyzed by enzymes, glutamate cysteine ligase (GCL), and GSH synthetase in an ATP dependent biosynthetic pathway, with cysteine being the rate-limiting substrate 16. Healthy cells have 90% of glutathione in reduced form, while only 10% is in the form of GSSG. The reduced form purports to be the biologically active form 12. An increase in oxidative stress leads to a homeostatic increment in the GSSG-to-GSH ratio 17, 18, 19. The cellular redox state is governed by the concentration and the ratio of NAD+/NADH, NADP+/NADPH and/or GSH/GSSG. GSH contains thiol (-SH) moiety that acts as reducing agent 20, 21. Thiol moiety in GSH serves to act as an antioxidant by serving as an electron donor, thereby reducing oxidized proteins in the cytoplasm to cysteine in a process called glutathionylation 21. In this process, thiol moiety (-SH) gets converted to the oxidized form to give glutathione disulfide (GSSG), also called L-(–)-glutathione 20, 21, 22, 23.
GSH reduces peroxides, catalyzed by glutathione peroxidases (GPx) by serving as an electron donor. H2O2 and organic hydroperoxides are metabolized by GPx Fig. 1 24, 25. Since there is no enzyme to reduce GPx, GSH acts reducing agent to GPx, and itself gets oxidized to GSSG. The oxidized glutathione (GSSG) is recycled to its reduced form (GSH), by an enzyme, GSH reductase (GR) using NADPH as an electron donor. Moreover, GSH also acts as a cofactor for oxidized glutaredoxin, formed during the reduction of disulfides 21.
FIG. 1: ROLE OF GSH IN REDUCTION OF PEROXIDES. NADPH ACTS AS AN ELECTRON DONOR
2. Glutathione in Cancer Cell Death: Cancer is the ultimate consequence of the failure of the cellular machinery to multiply normally and response to external or internal insult is overwhelmed such that the mitotic machinery develops aberrations 26.
The development of cancer does have genetic and epigenetic basis 27. Constant exposure to environmental pollution, dietary insufficiency, radiation, or hazardous chemical exposure along with chronic irritation, infection, or inflammation causes such homeostatic breakdown leading to a growth imbalance, attributable to the mutation of oncogenes and tumor suppressor genes, forfeiting response to cellular death signals. The overall perturbation is an anomalous expression of anti-death and pro-death proteins 28, 29.
The oncogenic stimulation causes an increased cellular metabolism leading to an aberrant increase in ROS stress, which manifolds than that in the normal cells; thus, changing mitochondrial membrane permeability 30, 31. An increase in free radicals activates cellular defense system for repair or neutralization 32. However, if the damage is irreversible and depending upon the extent and duration of redox imbalance, the cell progresses towards cell death 33, 34, 35. However, cancer cells adapt to a survival mode by an increased antioxidant capacity by overexpression of GSH 36 and enzymes responsible for GSH homeostasis, such as GSH peroxidase, GSH reductase, glutaredoxins, and GSH transferases. Overexpression of GSH has contributed to chemo- and radiotherapy resistance 2, 37.
GSH has shown to afford protection against stress-induced apoptosis 38, 39. Prevention of apoptosis has shown to be associated with the protection of redox-active catalytic sites at cysteines of the caspases 12. GSH is necessary for cell survivability as its exhaustion predisposes the cells to free radical attack 40. Various basic cellular components like lipid, protein, carbohydrate, and DNA are susceptible to attack by free radicals 41. An increase in cellular oxidative stress causes deleterious derangements supporting cell death such as single- and double-strand DNA fragmentation, damages in mitochondria causing permeability alterations by decreasing the transmembrane potential, and promoting the extrusion of death-related molecular signals 37, 42. Therefore, inhibiting GSH synthesis by an agent such as L-buthionine-(S, R)-sulfoximine (BSO) causes GSH depletion 43 and hence, can act as a potential target to sensitize resistant tumor cells 44 Fig. 2. Simon et al., have also shown that BSO tends to sensitize cancer cells to chemotherapeutic agents 45.
FIG. 2: EFFECT OF CELLULAR GSH DEPLETION IN CANCER CELLS: HIGH CELLULAR GSH LEVELS ARE A SURVIVAL ADAPTION OF CANCER CELLS TO ACCOMMODATE HIGH METABOLIC STRESSORS SUCH AS ROS, MAKING THEM RESISTANT TO CHEMOTHERAPY. HOWEVER, CELLULAR DEPLETION OF GSH RENDERS CELLS MORE VULNERABLE TO ROS ATTACKS
Apoptosis due to the oxidative stress can be activated either through an intrinsic pathway (mitochondria-derived) or extrinsic pathway (death receptor-mediated) former involving procaspase-9 activation while the latter involves activation of membrane receptors without activation of mitochondrial pro-apoptotic events 46. Furthermore, GSH depletion substantially increases ROS, thus activating autophagy 47. Increased ROS leaves the cell prone to oxidative damage, which in course if exceeds repairability of the cellular machinery, calls for activation of autophagy involving the formation of autophagosome that digests organelles 48. And if the damage is irreversible, autophagy becomes a pro-death mechanism 49.
3. GSH Depletion and Mechanisms of Cancer Cell Death: Cancer cells are immortal due to alterations in pathways that normally maintain the equilibrium between cell survivability and cell senescence 50. Any alteration in cell death can lead to multiple pathological diseases like cancer, aging, neurodegenerative disorders, cystic fibrosis 51.
Manipulating intracellular GSH by drugs such as BSO (inhibition of GSH synthesis) 52, Cys starvation (required for GSH synthesis) or γ-glutamate-cysteine ligase knockdown (in cultured cells) have shown to induce cell death, with or without inducing apoptosis 53, 54, 55.
Moreover, cytosolic efflux of GSH through MRP1 56, direct GSH oxidation by ROS or hindering GSH transport across mitochondria facilitates permeability transition pore opening, thereby activating mitochondrion-based death mechanism14 Fig. 3.
FIG. 3: ENDOGENOUS FACTORS AFFECTING OPENING OF PERMEABILITY TRANSITION PORE
GSH has found to exert most of its actions via modulating gene transcription 58. The effects of GSH are regulated by various transcription factors. For instance, redox-sensitive transcription factor, NF-E2 p45-related factor-2 (Nrf2) regulates GSH-related enzyme activities, which is itself under GSH regulation 59. Nuclear factor erythroid 2–related factor 2 (Nrf2), a basicleucine zipper (bZIP) protein regulates the basal and induced expression of antioxidant response element-dependent genes. Nrf2 binds to antioxidant response elements (ARE) and stimulates transcription of detoxification-related genes 60. Oxidation of thiol-containing proteins induces the release of Nrf-2, which thereafter translocate to the nucleus. Association of Nrf-2 to antioxidant response elements (ARE) in the control regions of multiple detoxification-related genes, activates gene transcriptions 61.
In the event of GSH depletion caused by BSO, Nrf2 is unregulated to provide antioxidant defense. Several studies demonstrate that Nrf-2-deficient murine embryonic fibroblasts lack this defense capacity leading to concentration, caspase-3 activation, and cellular death. Furthermore, Nrf-2 deficient cells have been found to be highly susceptible to doxorubicin and BSO treatment-induced cell death than wild cells 62. Furthermore, events leading to oxidation or reduction of critical Cys residues in the DNA binding domain of several transcription factors affect interactions with specific DNA bases 63. Oxidation of these Cys residues alters the 3D structure of transcription factors, which in turn affects its function 64, 65. Functional changes in any of these transcription factors affect gene expression of NF-κB, p53, MAPK, etc. by either upregulating or downregulating the process. Several MAPKs, including ERK, JNK, and p38 are known to have a crucial role in stress-induced apoptosis 66, 67, 68.
3.1 GSH and Apoptosis: Apoptosis or programmed cell death type 1 is a highly organized mechanism of controlled cell death, induced by diverse form of stimuli leading to terminal activation of cysteine-aspartate proteases 69. Various precise non-inflammatory apoptotic pathways undergo progressive activation, leading to specific bio-chemical and morphological cellular aberrations. Deregulation of apoptosis is found to be either a cause or consequence in several pathological conditions including cancer, auto-immune disorders, and neurodegeneration 70, 71.
Cytotoxic agents, such as chemotherapeutics, xenobiotics, and metals produce oxidative stress by inducing GSH depletion, which eventually lead to apoptosis 72, 73. This may be attributable to either oxidation of GSH to GSSG by reactive oxygen/nitrogen species or due to conjugation with highly reactive compounds. In contrast, apoptosis induced by stimuli other than reactive species, such as activation of death receptors, is found to be mediated by an efflux transport of GSH through plasma membrane 74. GSH is crucial for cell survival. Its depletion or extrusion renders the cell susceptible to death-receptor activation or mitochondrial apoptotic signaling making this strategy useful in rendering chemo- and radio-therapy resistant cells vulnerable to cell death 75, 76 Fig. 4.
FIG. 4: ROLE OF GSH MODULATION IN VARIOUS PATHWAYS OF CELL DEATH
BSO selectively inhibits γ-glutamate-cysteine ligase, depleting GSH without triggering apoptosis but facilitating other death-related mechanisms. For example, BSO potentiates death-receptor-induced apoptosis in cells 77. An increase in cellular ROS/RNS leads to loss of mitochondrial integrity, thereby activating the intrinsic apoptotic pathway 78, 79.
Permeability transition pore at the inner-outer membrane contact of mitochondria is voltage- and Ca2+-dependent, cyclosporine A-sensitive 80, 81, high conductance channel whose permeability suddenly increases for water and solutes with molecular masses of up to 1,500 Da 12. Compromised mitochodrial integrity is followed by an increased osmosis leading to swelling of mitochondria, rupture of the outer mitochondrial membrane, and release of pro-apoptogenic proteins into the cytosol 82. Pro-apoptotic factors released include apoptosis-inducing factor (AIF) 83 or second mitochondria-derived activator of caspases / direct IAP 84. Cyt c released from mitochondria binds to Apaf-1 (apoptotic protease-activating factor-1), forming apoptosome which recruits procaspase-9 84. Downstream cleavage/activation of effector caspases, -3 and -6/7 is signaled by ATP-dependent scission of procaspase-9 85.
Furthermore, GSH efflux by compounds such as resveratrol, a plant polyphenol induces experi-mental apoptosis by a mechanism involving BAX overexpression-mediated apoptosis, which is a ROS independent mechanism 28, 49, 86. ROS, electrophiles, and phenolic oxidants induce Nrf2 transcription factor to activate several detoxi-fication-related genes including those for γ-glutamate-cysteine ligase 28.
Genetically altered mice deficient in Nrf-2 (Nrf-2−/−) are found to be vulnerable to the damaging effects of hyperoxia, as indicated by an increase in pulmonary perme-ability, macrophage infiltration and epithelial injury in comparison to control wild mice 87, 88. Cell apoptosis is also found to be regulated by GSH/ GSSG redox status 17. According to one study, apoptosis was found to be regulated by a consistent GSH/GSSG imbalance, whereby a rise in GSSG produces kinetically accelerated loss of mitochondrial integrity, translocation of Cyt c from mitochondria-to-cytosol followed by caspase-3 activation 89. A significant rise in GSSG was found to occur within a narrow window of redox shift 90. During an initial 30 min post-oxidative challenge, followed by cellular GSH/GSSG balance recovery by 1 h had no influence on the apoptotic endpoint, a response that is coherent with early initiation of redox signaling. Pretreatment with a thiol antioxidant, N-acetylcysteine (NAC) could block oxidant-induced cell apoptosis by preventing an increase in GSSG 91. Howsoever, NAC does not prevent the oxidative stress once the rise in GSSG has occurred 92.
Mitochondria do not synthesize GSH but uptakes it from cytosol through a multicomponent transport system 14. The influxed GSH is the source of antioxidant defense against peroxides generated via ETC; regulating mitochondrial permeability transition and permeability transition pore opening 93. Mitochondrion-based cell death can be made feasible by either targeting GSH uptake of mitochondrion or through direct mtGSH depletion. This could be an attractive strategy to sensitize malignant cells to molecular effectors (e.g., oxidative stress inducers and/or cytotoxic drugs) and cause mitochondrion targeted cell death 94.
3.2 GSH and Autophagy: Autophagy or programmed cell death type II is a natural, regulated, destructive mechanism of the cell whereby the targeted cytoplasmic organelles are engulfed within lysosome to form the double-membrane vesicle, autophagosome 95. The autophagosome eventually fuses with lysosomes to form autolysosome and the contents are degraded by different acid hydrolases and recycled 96. While autophagy is a mechanism to digest cellular debris, worn out organelles and intracellular pathogenic organisms, if the stimuli are persistent, it becomes a pro-death mechanism 97, 98. Lysosomes are home to more than 50 soluble acid hydrolases that are meant to perform a cellular digestive function and over 120 lysosomal membrane proteins to maintain the structural integrity of the organelle, regulate lysosomal trafficking, fusion, and intralysosomal pH. The intra-organelle pH is highly acidic i.e. pH 4.5-5.0 which is crucial for the optimal catalytic activity of its hydrolytic enzymes 99, 100. Defective autophagic machinery compromises cellular recycling mechanism and leads to several pathological conditions such as cancer and neurodegenerative diseases 101, 102. Several stimuli can induce autophagy experimentally. This includes starvation (by culturing cells in Earle's Balanced Salt Solution (EBSS)), ROS, amino acid deprivation, and three different mTOR inhibitors, rapamycin, PP242 and Torin1 103, 104. ROS is crucial for inducing starvation-induced autophagy. As demonstrated by Scherz-Shouval et al., 105 starvation-induced autophagy targets ATG4, a thiol-containing protein that initiates early step of autophagosome formation 106.
A relation between starvation and GSH levels were elucidated in a study carried out by Desideri et al., whereby, cervix carcinoma HeLa cells were cultured in HBSS (to mimic nutrient starvation) and the GSH levels were analyzed by HPLC technique. Results concluded a time-dependent decrement in GSH concentration from 3 h of nutrient removal. Moreover, a progressive decrement in GSH/GSSG ratio was observed owing to decreasing GSH levels but a non-significant change in GSSG levels. Nutrient starvation in HeLa cells, as well as in HepG2 and H1299 cells, has shown to produce a significant increase in extracellular GSH levels post 3h nutrient starvation. And observed GSH efflux was not due to cells undergoing apoptosis 66 as there was no significant activation of markers for caspase-dependent apoptosis i.e. caspase 9/3 (CASP9/3), 24 h prior to starvation in HeLa and H1299 cells and 48 h in HepG2 cell, further indicating that a reduction/extrusion of GSH was not a consequence of cells undergoing apoptosis 95, 96.
Autophagy protein 5 (ATG5) and its complex in ATG12-ATG5:ATG16L causes elongation of the phagophore in the autophagic pathway 105. Studies including a knockdown model of ATG5 were not found to counteract the decrease in GSH, showing this isn't brought about via autophagy initiation 98. As GSH is a ubiquitous molecule, its role in autophagy is not completely elucidated, but several studies demonstrate that N-acetyl-L-cysteine induced replenishment of cellular GSH halters autophagy induction along with autophagosome formation and protein degradation induced by starvation 107, 108.
There has been a correlation between intracellular GSH pools and induction of mitochondrial autophagy (mitophagy) as studied in yeast by Deffieu et al. In the study by Deffieu et al., involving regulation of mitophagy in yeast, 3h after nitrogen starvation, 75% of the cells show the vacuolization of mitochondria. However, in the presence of NAC, 13% of the cells had their mitochondria/vacuoles in contact after 2 h of starvation, while 13% of the cells exhibited vacuolar sequestration of mitochondria after 3 h of starvation. The observations were a conclusive indication of the protective effect of NAC, as shown by impaired mitochondria/vacuoles contacts and mitochondria sequestration in vacuoles. The study though established the protective role of NAC against nitrogen starvation-induced mitophagy but the correlation between GSH thiol redox state and the induction/execution of autophagy in mammalian cells, is insufficient 109.
CONCLUSION: GSH is crucial for the survival of cells against a battle with free radicals generated as a byproduct of cellular metabolism or at an encounter with xenobiotics, ionizing radiations, and oxidative stress-inducing biotherapy. Tumor cells have the advantage of producing a high level of intracellular GSH and hence, high antioxidant capacity, therefore, a novel strategy to selectively deplete GSH in cancer cells offers a promising treatment approach in drug and radiation-resistant cases. For instance, GSH concentration in mammary gland tumors range between 10–40 nmol/mg protein, while for disease-free breast tissue, this value falls down to a range between 1–10 nmol/mg-protein 110. The heterogeneous nature of tumor cells provides them with an excellent armor to counteract the death-inducing stimuli, where different cell subset show different states of resistance where one strategy may include overproduction of GSH. Depletion of cytosolic or mitochondrial GSH by promoting efflux or inhibiting uptake predisposes cells to oxidative stress, thereby facilitating the release of death-inducing molecular signals ending in apoptosis or autophagy.
BSO inhibiting γ-glutamate-cysteine ligase (depleting GSH without triggering apoptosis) or promoting GSH efflux by compound like resveratrol (triggering apoptosis) 110 or inducing autophagy through several stimuli like starvation (by culturing cells in Earle's Balanced Salt Solution (EBSS)), ROS, amino acid deprivation and three different mTOR inhibitors, rapamycin, PP242, and Torin1 111, 112 appears to be promising but considering the heterogeneous nature of tumors and non-uniform levels of GSH in different cell types, ways to selectively deplete it from the cancer cells without jeopardizing the normal healthy cells is challenging.
ACKNOWLEDGEMENT: We are grateful to the Indian Institute of Technology (Banaras Hindu University), Varanasi, India.
CONFLICTS OF INTEREST: None
REFERENCES:
- Waypa GB, Smith KA and Schumacker PT: O2 sensing, mitochondria and ROS signaling: the fog is lifting. Molecular Aspects of Medicine 2016; 47: 76-89.
- Panieri E and Santoro M: ROS homeostasis and metabolism: a dangerous liason in cancer cells. Cell death & Disease 2016; 7(6): e2253-e.
- Zhao RZ, Jiang S, Zhang L and Yu ZB: Mitochondrial electron transport chain, ROS generation and uncoupling. International Journal of Mole Medicine 2019; 44(1): 3-15.
- Brand MD: Mitochondrial generation of superoxide and hydrogen peroxide as the source of mitochondrial redox signaling. Free Radical Bio and Medi 2016; 100: 14-31.
- Mnatsakanyan N, Beutner G, Porter GA, Alavian KN and Jonas EA: Physiological roles of the mitochondrial permeability transition pore. Journal of Bioenergetics and Biomembranes 2017; 49(1): 13-25.
- Zou X, Ratti BA, O’Brien JG, Lautenschlager SO, Gius DR and Bonini MG: Manganese superoxide dismutase (SOD2): is there a center in the universe of mitochondrial redox signaling? Journal of Bioenergetics and Biomembranes 2017; 49(4): 325-33.
- Che M, Wang R, Li X, Wang HY and Zheng XS: Expanding roles of superoxide dismutases in cell regulation and cancer. Drug Discovery Today 2016; 21(1): 143-9.
- Mondola P, Damiano S, Sasso A and Santillo M: The Cu, Zn superoxide dismutase: not only a dismutase enzyme. Frontiers in Physiology 2016; 7: 594.
- Ighodaro OM and Akinloye OA: First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): Their fundamental role in the entire antioxidant defence grid. Alexandria Journal of Medicine 2018; 54(4): 287-93.
- Bardaweel SK, Gul M, Alzweiri M, Ishaqat A, and AL-Salamat: Reactive oxygen species: the dual role in physiological and pathological conditions of the human body. Eurasian J Med 2018; 50(3): 193-201.
- Ighodaro O and Akinloye O: First line defence antioxidants-superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX): Their fundamental role in the entire antioxidant defence grid. Alexandria Journal of Medicine 2018; 54(4): 287-93.
- Bansal A and Simon MC: Glutathione metabolism in cancer progression and treatment resistance. Journal of Cell Biology 2018; 217(7): 2291-8.
- Circu ML and Yee Aw T: Glutathione and apoptosis. Free Radical Research 2008; 42(8): 689-06.
- Saini R, Saini HS and Dahiya A: Iron treatment enhances the levels of reduced glutathione, oxidized glutathione and glutathione reductase activity in Rice (Oryza sativa). Journal of Pharma and Phytochemistry 2017; 6(4): 1321-8.
- Ribas V, García-Ruiz C and Fernández-Checa JC: Glutathione and mitochondria. Frontiers in Pharmacology 2014; 5: 151.
- Cichoż-Lach H and Michalak A: Oxidative stress as a crucial factor in liver diseases. World Journal of Gastroenterology 2014; 20(25): 8082.
- Wang L, Yan B, Liu N, Li Y and Wang Q: Effects of cadmium on glutathione synthesis in hepatopancreas of freshwater crab, Sinopotamon yangtsekiense. Chemosphere 2008; 74(1): 51-6.
- Aquilano K, Baldelli S and Ciriolo MR: Glutathione: new roles in redox signaling for an old antioxidant. Frontiers in Pharmacology 2014; 5: 196.
- Timm KN, Hu DE, Williams M, Wright AJ, Kettunen MI, Kennedy BW, Larkin TJ, Dzien P, Marco-Rius I, Bohndiek SE and Brindle KM: Assessing oxidative stress in tumors by measuring the rate of hyperpolarized [1-13C] dehydroascorbic acid reduction using 13C magnetic resonance spectroscopy. Journal of Biological Chemistry 2017; 292(5): 1737-48.
- Torres-Cuevas I, Parra-Llorca A, Sánchez-Illana A, Nuñez-Ramiro A, Kuligowski J and Cháfer-Pericás C: Oxygen and oxidative stress in the perinatal period. Redox Biology 2017; 12: 674-81.
- Berthiaume JM, Kurdys JG, Muntean DM and Rosca MG: Mitochondrial NAD+/NADH redox state and diabetic cardiomyopathy. Antioxidants & Redox Signaling 2019; 30(3): 375-98.
- Kalinina E, Chernov N and Novichkova M: Role of glutathione, glutathione transferase and glutaredoxin in regulation of redox-dependent processes. Biochemistry (Moscow) 2014; 79(13): 1562-83.
- Couto N, Wood J and Barber J: The role of glutathione reductase and related enzymes on cellular redox homoeo-stasis network. Free Rad Bio and Medi 2016; 95: 27-42.
- Xiao W, Wang R-S, Handy DE and Loscalzo J: NAD (H) and NADP (H) redox couples and cellular energy metabolism. Antioxidants & Redox Signaling 2018; 28(3): 251-72.
- Gaschler MM and Stockwell BR: Lipid peroxidation in cell death. Biochemical and Biophysical Research Communications 2017; 482(3): 419-25.
- Sies H: Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biology 2017; 11: 613-9.
- Harrington KJ: The biology of cancer. Medicine 2016; 44(1): 1-5.
- Ahmad A, Azim S, Zubair H, Khan MA, Singh S, Carter JE, Rocconi RP and Singh AP: Epigenetic basis of cancer health disparities: looking beyond genetic differences. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer 2017; 1868(1): 16-28.
- Ortega AL, Mena S and Estrela JM: Glutathione in cancer cell death. Cancers 2011; 3(1): 1285-10.
- Zaorsky N, Churilla T, Egleston B, Fisher S, Ridge J and Horwitz E: Causes of death among cancer patients. Annals of oncology 2017; 28(2): 400-7.
- Treulen F, Uribe P, Boguen R and Villegas JV: Mitochondrial permeability transition increases reactive oxygen species production and induces DNA fragmentation in human spermatozoa. Human Reproduction 2015; 30(4): 767-76.
- Ralph S, Rodriguez-Enriquez S, Neuzil J, Saavedra E, Moreno-Sanchez R. Ralph SJ, Rodriguez-Enriquez S, Neuzil J, Saavedra E and Moreno-Sanchez R: The causes of cancer revisited: ‘mitochondrial malignancy’ and ROS-induced oncogenic transformation - why mitochondria are targets for cancer therapy. Molecular Aspects of Medicine 2010; 31: 145-70.
- Poprac P, Jomova K, Simunkova M, Kollar V, Rhodes CJ and Valko M: Targeting free radicals in oxidative stress-related human diseases. Trends in Pharmacological Sciences 2017; 38(7): 592-607.
- Georgieva E, Ivanova D, Zhelev Z, Bakalova R, Gulubova M and Aoki I: Mitochondrial dysfunction and redox imbalance as a diagnostic marker of “free radical diseases”. Anticancer Research 2017; 37(10): 5373-81.
- Kwon M-Y, Park E, Lee S-J and Chung SW: Heme oxygenase-1 accelerates erastin-induced ferroptotic cell death. Oncotarget 2015; 6(27): 24393.
- Pham-Huy LA, He H and Pham-Huy C: Free radicals, antioxidants in disease and health. International Journal of Biomedical Science 2008; 4(2): 89.
- Sznarkowska A, Kostecka A, Meller K and Bielawski KP: Inhibition of cancer antioxidant defense by natural compounds. Oncotarget 2017; 8(9): 15996.
- Liu J, Li D, Zhang T, Tong Q, Ye RD and Lin L: SIRT3 protects hepatocytes from oxidative injury by enhancing ROS scavenging and mitochondrial integrity. Cell Death & Disease 2017; 8(10): e3158-e.
- Gao Y, Zhuang Z, Gao S, Li X, Zhang Z and Ye Z: Tetrahydrocurcumin reduces oxidative stress-induced apoptosis via the mitochondrial apoptotic pathway by modulating autophagy in rats after traumatic brain injury. American Journal of Trans Research 2017; 9(3): 887.
- Kwon DH, Cha H-J, Lee H, Hong S-H, Park C and Park SH: Protective effect of glutathione against oxidative stress-induced cytotoxicity in RAW 264.7 macrophages through activating the nuclear factor erythroid 2-related factor-2/heme oxygenase-1 pathway. Antioxidants 2019; 8(4): 82.
- Leong P and Ko K: Induction of the glutathione antioxidant response glutathione redox cycling by nutraceuticals: mechanism of protection against oxidant-induced cell death. Journal of Nutraceuticals and Food Science 2016; 1: 1.
- Pacifici RE and Davies KJ: Protein, lipid and DNA repair systems in oxidative stress: the free-radical theory of aging revisited. Gerontology 1991; 37(1-3): 166-80.
- Poyton RO, Ball KA and Castello PR: Mitochondrial generation of free radicals and hypoxic signaling. Trends in Endocrinology & Metabolism 2009; 20(7): 332-40.
- Wrotek S, Domagalski K, Jędrzejewski T, Dec E and Kozak W: Buthionine sulfoximine, a glutathione depletor, attenuates endotoxic fever and reduces IL-1β and IL-6 level in rats. Cytokine 2017; 90: 31-7.
- Gana CC, Hanssen KM, Denise M, Flemming CL, Wheatley MS and Conseil G: MRP1 modulators synergize with buthionine sulfoximine to exploit collateral sensitivity and selectively kill MRP1-expressing cancer cells. Biochemical Pharmacology 2019; 168: 237-48.
- Simons AL, Ahmad IM, Mattson DM, Dornfeld KJ and Spitz DR: 2-Deoxy-D-glucose combined with cisplatin enhances cytotoxicity via metabolic oxidative stress in human head and neck cancer cells. Cancer Research 2007; 67(7): 3364-70.
- Sun Y, Zheng Y, Wang C and Liu Y: Glutathione depletion induces ferroptosis, autophagy, and premature cell senescence in retinal pigment epithelial cells. Cell Death & Disease 2018; 9(7): 1-5.
- Lemasters JJ, Nieminen A-L, Qian T, Trost LC, Elmore SP and Nishimura Y: The mitochondrial permeability transition in cell death: a common mechanism in necrosis, apoptosis and autophagy. Biochimica et Biophysica Acta (BBA)-bioenergetics 1998; 1366(1-2): 177-96.
- Kumar B, Iqbal MA, Singh RK and Bamezai RN: Resveratrol inhibits TIGAR to promote ROS induced apoptosis and autophagy. Biochimie 2015; 118: 26-35.
- Chen S, Zhou N, Zhang Z, Li W and Zhu W: Resveratrol induces cell apoptosis in adipocytes via AMPK activation. Biochemical and Biophysical Research Communications 2015; 457(4): 608-13.
- Lapinska K, Faria G, Mcgonagle S, Macumber KM, Heerboth S and Sarkar S: Cancer progenitor cells: the result of an epigenetic event? Antica Res 2018; 38(1): 1-6.
- Favaloro B, Allocati N, Graziano V, Di Ilio C and De Laurenzi V: Role of apoptosis in disease. Aging (Albany NY) 2012; 4(5): 330.
- Rodman SN, Spence JM, Ronnfeldt TJ, Zhu Y, Solst SR and O'Neill RA: Enhancement of radiation response in breast cancer stem cells by inhibition of thioredoxin-and glutathione-dependent metabolism. Radiation Research 2016; 186(4): 385-95.
- Armstrong JS, Whiteman M, Yang H, Jones DP and Sternberg P: Cysteine starvation activates the redox-dependent mitochondrial permeability transition in retinal pigment epithelial cells. Investigative Ophthalmology & Visual Science 2004; 45(11): 4183-9.
- Dalton TP, Chen Y, Schneider SN, Nebert DW and Shertzer HG: Genetically altered mice to evaluate glutathione homeostasis in health and disease. Free Radical Biology and Medicine 2004; 37(10): 1511-26.
- Lin A-H, Chen H-W, Liu C-T, Tsai C-W and Lii CK: Activation of Nrf2 is required for up-regulation of the π class of glutathione S-transferase in rat primary hepatocytes with L-methionine starvation. Journal of Agricultural and Food Chemistry 2012; 60(26): 6537-45.
- Oestreicher J and Morgan B: Glutathione: subcellular distribution and membrane transport. Biochemistry and Cell Biology 2019; 97(3): 270-89.
- Schumacker PT: Reactive oxygen species in cancer: a dance with the devil. Cancer Cell 2015; 27(2): 156-7.
- Mukherjee A, Malik H, Saha AP, Dubey A, Singhal DK and Boateng S: Resveratrol treatment during goat oocytes maturation enhances developmental competence of parthenogenetic and hand-made cloned blastocysts by modulating intracellular glutathione level and embryonic gene expression. Journal of assisted reproduction and genetics 2014; 31(2): 229-39.
- Wang P, Li CG, Qi Z, Cui D and Ding S: Acute exercise stress promotes Ref1/Nrf2 signalling and increases mitochondrial antioxidant activity in skeletal muscle. Experimental Physiology 2016; 101(3): 410-20.
- Janssen-Heininger YM, Aesif SW, van der Velden J, Guala AS, Reiss JN and Roberson EC: Regulation of apoptosis through cysteine oxidation: implications for fibrotic lung disease. Annals of the New York Academy of Sciences 2010; 1203: 23.
- Kensler TW, Wakabayashi N and Biswal S: Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annual Review of Pharmacology and Toxicology 2007; 47: 89-116.
- Biswas M and Chan JY: Role of Nrf1 in antioxidant response element-mediated gene expression and beyond. Toxicology and applied pharmacology 2010;244(1):16-20.
- Arrigo AP: Gene expression and the thiol redox state. Free Radical Biology and Medicine 1999; 27(9-10): 936-44.
- Chio IIC, Jafarnejad SM, Ponz-Sarvise M, Park Y, Rivera K and Palm W: NRF2 promotes tumor maintenance by modulating mRNA translation in pancreatic cancer. Cell 2016; 166(4): 963-76.
- Li H, Tang Z, Chu P, Song Y, Yang Y and Sun B: Neuroprotective effect of phosphocreatine on oxidative stress and mitochondrial dysfunction induced apoptosis in vitro and in vivo: Involvement of dual PI3K/Akt and Nrf2/HO-1 pathways. Free Radical Biology and Medicine 2018; 120: 228-38.
- Sui X, Kong N, Ye L, Han W, Zhou J and Zhang Q: p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Letters 2014; 344(2): 174-9.
- Sun Y, Liu W-Z, Liu T, Feng X, Yang N and Zhou HF: Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. Journal of Receptors and Signal Trans 2015; 35(6): 600-4.
- Lee JS, Kim YR, Song IG, Ha SJ, Kim YE and Baek NI: Cyanidin-3-glucoside isolated from mulberry fruit protects pancreatic β-cells against oxidative stress-induced apoptosis. International Journal of Molecular Medicine 2015; 35(2): 405-12.
- Banerjee M, Chattopadhyay S, Choudhuri T, Bera R, Kumar S and Chakraborty B: Cytotoxicity and cell cycle arrest induced by andrographolide lead to programmed cell death of MDA-MB-231 breast cancer cell line. Journal of Biomedical Science 2016; 23(1): 40.
- Bhat AH, Dar KB, Anees S, Zargar MA, Masood A and Sofi MA: Oxidative stress, mitochondrial dysfunction and neurodegenerative diseases; a mechanistic insight. Biomedicine & Pharmacotherapy 2015; 74: 101-10.
- Chen Q, Kang J and Fu C: The independence of and associations among apoptosis, autophagy, and necrosis. Signal Transduction and Targeted Ther 2018; 3(1): 1-11.
- Zanotto-Filho A, Masamsetti VP, Loranc E, Tonapi SS and Gorthi A: Alkylating agent–induced NRF2 blocks endoplasmic reticulum stress–mediated apoptosis via control of glutathione pools and protein thiol homeostasis. Molecular Cancer Therapeutics 2016; 15(12) 3000-14.
- Varghese MV, Manju A, Abhilash M, Paul MS, Abhilash S and Nair RH: Oxidative stress induced by the chemotherapeutic agent arsenic trioxide. 3 Biotech 2014; 4(4): 425-30.
- Franco R, Bortner CD, Schmitz I and Cidlowski JA: Glutathione depletion regulates both extrinsic and intrinsic apoptotic signaling cascades independent from multidrug resistance protein 1. Apoptosis 2014; 19(1): 117-34.
- Bansal A and Simon MC: Glutathione metabolism in cancer progression and treatment resistance. Journal of Cell Biology 2018; 217(7): 2291-8.
- Roh J-L, Kim EH, Jang H and Shin D: Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis. Redox Biology 2017; 11: 254-62.
- Caro AA, Commissariat A, Dunn C, Kim H, García SL and Smith A: Prooxidant and antioxidant properties of salicylaldehyde isonicotinoyl hydrazone iron chelators in HepG2 cells. Biochimica et Biophysica Acta (BBA)-General Subjects 2015; 1850(11): 2256-64.
- Redza-Dutordoir M and Averill-Bates DA: Activation of apoptosis signalling pathways by reactive oxygen species. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research 2016; 1863(12): 2977-92.
- Maharjan S, Oku M, Tsuda M, Hoseki J and Sakai Y: Mitochondrial impairment triggers cytosolic oxidative stress and cell death following proteasome inhibition. Scientific Reports 2014; 4: 5896.
- Bernardi P and Di Lisa F: The mitochondrial permeability transitions pore: molecular nature and role as a target in cardioprotection. Journal of Molecular and Cellular Cardiology 2015; 78: 100-6.
- Briston T, Roberts M, Lewis S, Powney B, Staddon JM and Szabadkai G: Mitochondrial permeability transitions pore: sensitivity to opening and mechanistic dependence on substrate availability. Scientific Reports 2017; 7(1): 1-13.
- Desagher S and Martinou JC: Mitochondria as the central control point of apoptosis. Trends in Cell Biology 2000; 10(9): 369-77.
- Garrido C, Galluzzi L, Brunet M, Puig P, Didelot C and Kroemer G: Mechanisms of cytochrome c release from mitochondria. Cell Death & Differentiation 2006; 13(9): 1423-33.
- Zhou M, Li Y, Hu Q, Bai X-c, Huang W and Yan C: Atomic structure of the apoptosome: mechanism of cytochrome c-and dATP-mediated activation of Apaf-1. Genes & Development 2015; 29(22): 2349-61.
- Ma Y, Zhu B, Yong L, Song C, Liu X and Yu H: Regulation of intrinsic and extrinsic apoptotic pathways in osteosarcoma cells following oleandrin treatment. International Journal of Molecular Sciences 2016; 17(11): 1950.
- Guha P, Dey A, Sen R, Chatterjee M, Chattopadhyay S and Bandyopadhyay SK: Intracellular GSH depletion triggered mitochondrial Bax translocation to accomplish resveratrol-induced apoptosis in the U937 cell line. Journal of Pharmacology and Experimental Therapeutics 2011; 336(1): 206-14.
- Cho HY and Kleeberger SR: Association of Nrf2 with airway pathogenesis: lessons learned from genetic mouse models. Archives of Toxicology 2015; 89(11): 1931-57.
- Schafer FQ and Buettner GR: Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radical Biology and Medicine 2001; 30(11): 1191-12.
- Pias E and Aw T: Early redox imbalance mediates hydroperoxide-induced apoptosis in mitotic competent undifferentiated PC-12 cells. Cell Death & Differentiation 2002; 9(9): 1007-16.
- Suntres ZE: Role of antioxidants in paraquat toxicity. Toxicology 2002; 180(1): 65-77.
- Powers SK and Jackson MJ: Exercise-induced oxidative stress: cellular mechanisms and impact on muscle force production. Physiological Reviews 2008; 88(4): 1243-76.
- Calabrese G, Morgan B and Riemer J: Mitochondrial glutathione: regulation and functions. Antioxidants & Redox Signaling 2017; 27(15): 1162-77.
- Kowluru RA and Mishra M: Oxidative stress, mitochondrial damage and diabetic retinopathy. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 2015; 1852(11): 2474-83.
- Hileman EO, Liu J, Albitar M, Keating MJ and Huang P: Intrinsic oxidative stress in cancer cells: a biochemical basis for therapeutic selectivity. Cancer Chemotherapy and Pharmacology 2004; 53(3): 209-19.
- Levy JMM, Towers CG and Thorburn A: Targeting autophagy in cancer. Nature Reviews Cancer 2017; 17(9): 528.
- Yu L, Chen Y and Tooze SA: Autophagy pathway: cellular and molecular mechanisms. Autophagy 2018; 14(2): 207-15.
- Plaza-Zabala A, Sierra-Torre V and Sierra A: Autophagy and microglia: novel partners in neurodegeneration and aging. Int Journal of Molecular Sciences 2017; 18(3): 598.
- Zhou J, Tan SH, Nicolas V, Bauvy C, Yang ND and Zhang J: Activation of lysosomal function in the course of autophagy via mTORC1 suppression and autophagosome-lysosome fusion. Cell Research 2013; 23(4): 508-23.
- Cai Y, Gui C, Samedov K, Su H, Gu X, Li S, Luo W, Sung HH, Lam JW, Kwok RT and Williams ID: An acidic pH independent piperazine–TPE AIEgen as a unique bioprobe for lysosome tracing. Chemical Science 2017; 8(11): 7593-603.
- Settembre C, Fraldi A, Medina DL and Ballabio A: Signals from the lysosome: a control centre for cellular clearance and energy metabolism. Nature reviews Molecular Cell Biology 2013; 14(5): 283-96.
- Menzies FM, Fleming A, Caricasole A, Bento CF, Andrews SP, Ashkenazi A, Füllgrabe J, Jackson A, Sanchez MJ, Karabiyik C and Licitra F: Autophagy and neurodegeneration: pathogenic mechanisms and therapeutic opportunities. Neuron 2017; 93(5): 1015-34.
- Amaravadi R, Kimmelman AC and White E: Recent insights into the function of autophagy in cancer. Genes & Development 2016; 30(17): 1913-30.
- Gao L, Jauregui CE and Teng Y: Targeting autophagy as a strategy for drug discovery and therapeutic modulation. Future Medicinal Chemistry 2017; 9(3):335-45.
- Murugan S and Amaravadi RK: Methods for studying autophagy within the tumor microenvironment. Advances in Experimental Medicine and Biology 2016; 899: 145-66.
- Scherz-Shouval R and Elazar Z: ROS, mitochondria and the regulation of autophagy. Trends in Cell Biology 2007; 17(9): 422-7.
- Zhang L, Li J, Ouyang L, Liu B and Cheng Y: Unraveling the roles of Atg4 proteases from autophagy modulation to targeted cancer therapy. Cancer Letters 2016; 373(1): 19-26.
- Boşgelmez İİ and Güvendik G: N-acetyl-L-cysteine protects liver and kidney against chromium (VI)-induced oxidative stress in mice. Biological Trace Element Research 2017; 178(1): 44-53.
- Kurahashi T, Lee J, Nabeshima A, Homma T, Kang ES and Saito Y: Ascorbic acid prevents acetaminophen-induced hepatotoxicity in mice by ameliorating glutathione recovery and autophagy. Archives of Biochemistry and Biophysics 2016; 604: 36-46.
- Deffieu M, Bhatia-Kiššová I, Salin B, Galinier A, Manon S and Camougrand N: Glutathione participates in the regulation of mitophagy in yeast. Journal of Biological Chemistry 2009; 284(22): 14828-37.
- Kelkel M, Jacob C, Dicato M and Diederich M: Potential of the dietary antioxidants resveratrol and curcumin in prevention and treatment of hematologic malignancies. Molecules 2010; 15(10): 7035-74.
- Levine B and Kroemer G: Autophagy in the pathogenesis of disease. Cell 2008; 132(1): 27-42.
How to cite this article:
Gupta S and Sahu AN: Glutathione: induction of apoptosis and autophagy in cancer. Int J Pharm Sci & Res 2020; 11(8): 3608-18. doi: 10.13040/IJPSR.0975-8232.11(8).3608-18.
All © 2013 are reserved by the International Journal of Pharmaceutical Sciences and Research. This Journal licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 3.0 Unported License.
Article Information
8
3608-3618
788
864
English
IJPSR
S. Gupta and A. N. Sahu *
Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India.
ansahu.phe@iitbhu.ac.in
03 November 2019
17 February 2020
19 April 2020
10.13040/IJPSR.0975-8232.11(8).3608-18
01 August 2020