The Role of mTOR, Autophagy, Apoptosis, and Oxidative Stress During Toxic Metal Injury

The Role of mTOR, Autophagy, Apoptosis, and Oxidative Stress During Toxic Metal Injury

C H A P T E R 5 The Role of mTOR, Autophagy, Apoptosis, and Oxidative Stress During Toxic Metal Injury Sarmishtha Chatterjee, Chayan Munshi and Shell...

1MB Sizes 1 Downloads 21 Views

C H A P T E R

5 The Role of mTOR, Autophagy, Apoptosis, and Oxidative Stress During Toxic Metal Injury Sarmishtha Chatterjee, Chayan Munshi and Shelley Bhattacharya Environmental Toxicology Laboratory, Department of Zoology, Centre for Advanced Studies, Visva-Bharati (A Central University), Santiniketan, India

5.1 INTRODUCTION

mTORC1, and mTORC2 binds to rictor. Signaling through mTORC1 is much better understood than signaling through mTORC2. mTORC1 is an important node in cellular regulation impacting on cell growth that is linked to aging [15,16]. Structurally, mTOR contains 2549 amino acids and the region of first 1200 N-terminal amino acids contains up to 20 tandem repeated HEAT (a proteinprotein interaction structure of two tandem antiparallel α-helices found in Huntingtin Elongation factor 3, PR65/A subunit of protein phosphatase 2A, and TOR) motifs [17]. HEAT repeat region is followed by a FAT [FRAP, ATM, and TRRAP (PIKK family members)] domain and FKPB12rapamycin binding domain, which serves as a docking site for the rapamycinFKBP12 complex. Downstream lies a catalytic kinase domain and a FATC (FAT carboxyterminal) domain, located at the C-terminus of the protein [18]. The interactions between FAT and FATC might contribute to the catalytic kinase activity of mTOR [19]. The domain structure of mTOR is sketched in Figure 5.1.

Target of rapamycin (TOR) discovered in 1991, is a highly conserved protein kinase, essential for both fundamental and clinical biology [1]. The story of TORsignaling begins with a remarkable drug, rapamycin [2]. Rapamycin is a lipophlic macrolide as well as a natural secondary metabolite, produced by Streptomyces hygroscopicus. S. hygroscopicus is a bacterium, isolated from soil in Easter Island, called Rapa-Nui in 1965, thus the name rapamycin [1,3] was coined. Intracellular rapamycin receptor in all eukaryotes is a small ubiquitous protein, called FK506 binding protein 12 kDa (FKBP12) [4,5]. RapamycinFKBP12 complex interacts with evolutionarily conserved TOR proteins to potently inhibit downstream effectors. A single mammalian TOR (mTOR) protein was cloned from several species and alternatively termed as mTOR, FKBP12 and rapamycin associated protein (FRAP), rapamycin and FKBP12 target, sirolimus effector protein, or rapamycin target [611]. mTOR is 289 kDa, sharing B45% identity with the Streptomyces cerevisiae TOR proteins and B56% identity with Drosophila melanogaster TOR as well as the human, rat, and mouse mTOR proteins share .95% identity at the amino acid level [9,12]. The TOR proteins function as Ser/Thr protein kinases assigned to a protein family termed the phosphatidylinositol kinase-related kinases (PIKKs) [13,14]. mTOR is a multidomain protein kinase that interacts with other proteins to form two main types of complex, mTOR complexes 1 and 2 (mTORC1 and mTORC2) [15]. mTORC1 associates with raptor, which binds to proteins that are direct substrates for

Molecules to Medicine with mTOR DOI: http://dx.doi.org/10.1016/B978-0-12-802733-2.00013-X

5.2 CELLULAR FUNCTIONS OF mTOR mTOR is involved in many different cell functions to regulate cellular protein anabolism and catabolism [20,21]. If the cell has enough available amino acids, mTOR is free to signal to other molecules in the cell to build new proteins. On the other hand, if the cell is running low on nutrients, existing proteins and other cell components break down to free the building blocks which can be reused. The process by which the

69

© 2016 Elsevier Inc. All rights reserved.

70

5. THE ROLE OF mTOR, AUTOPHAGY, APOPTOSIS, AND OXIDATIVE STRESS DURING TOXIC METAL INJURY

cell breaks down its own components is called autophagy [22], programmed cell death type II, which basically means “eating of the self” [2326]. The degraded part of the cell is engulfed by a double membrane called phagophore, to separate from the rest of the cell, resulting in a membrane-enclosed bubble of cytosol, called autophagosome. Autophagosome eventually fuses with lysosomes, forming autophagolysosomes as the garbage disposal of the cell that can break down all sorts of cellular components. Key regulators of autophagy include the class I phosphoinositide-3kinase (PI3K) [27] and adenosine-mono-phosphate activated protein kinase (AMPK) [28] in response to TOR kinase (Figure 5.2).

FIGURE 5.1 The domain structure of mTOR.

Activated PI3K signals TOR kinase, especially mTORC1 via protein kinase B (Akt kinase, belonging to the AGC protein kinase family) and modulates autophagy [22,29]. AMPK regulates autophagy through the control of mTORC1. AMPK indirectly inhibited TORC1 by phosphorylation of tuberous sclerosis complex-2 (TSC2) via deactivation of the Rheb GTPase (Ras homology enriched in brain-GTP binding protein) [3032]. AMPK activation also induces phosphorylation of p53 on Ser15 domain. Phosphorylated p53 inhibits mTORC1 activity and regulates its downstream targets including autophagy [3335]. In addition, mTOR has profound effects on the control of apoptosis [36]. Apoptosis, programmed cell death type I, plays an important role in various physiological and pathophysiological circumstances [37]. The intrinsic pathway of apoptosis deals with mitochondrial pathway, is initiated by a large variety of upstream stimuli and tightly modulated by various factors including, pro- and antiapoptotic proteins of the Bcl-2 family as well as PI3K/Akt/mTOR pathway [3840]. The Bcl-2 family of proteins regulates apoptosis by controlling mitochondrial permeability. The proapoptotic Bcl-2 proteins [Bcl-2 associated X-protein (BAX) and proapoptotic BH3 only Bcl-2 family protein (BIM)] may reside in the cytosol but translocate to

FIGURE 5.2 Role of mTOR in autophagy.

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

71

5.3 FREE RADICALS IN CELLS

FIGURE 5.3 Role of mTOR in apoptosis.

mitochondria following death signaling and promote the release of cytochrome c (Cyt c) [40]. The PI3K signaling network diversifies into many distinct downstream branches, one of which leads to the phosphorylation of mTORC1 [4145] on Thr 2446 and Ser 2448 sites by Akt kinase via TSC2 phosphorylation. Akt acts as an antiapoptotic factor that directly or indirectly antagonizes cell death signal transduction via the mitochondrial pathway [46]. Akt directly interferes with the phosphorylation of apoptosis-regulatory proteins which in turn results in a shift to pro- and antiapoptotic stimuli [4749]. The multidomain Bcl-2 protein BAX and BIM are phosphorylated at serine residues Ser 184 and Ser87, respectively, in an Aktdependent manner [49,50]. Akt-mediated direct and indirect (by mTORC1) phosphorylation of some antiapoptotic factors, such as myeloid leukemia cell sequence 1 (Mcl-1) located in mitochondrial matrix and vice versa, sometimes diminishes the antiapoptotic properties by decreasing their protein stability. Thus degradation of these proteins via the proteasomal machinery results in a reduction of Mcl-1 protein expression [48,51] which signals the release of Cyt c from mitochondria and induces the intrinsic apoptotic pathway. On the other hand, mTORC2 elicits its activity as a potent antiapoptotic signal through Mcl-1 [52] and Akt phosphorylation [45,53]. mTORC2 phosphorylates Akt on Ser 473 and Thr 308 region by protein-dependent kinase 1 (PDK1), however mTORC2 recognizes Ser 473 region in the hydrophobic motif of Akt as a kinase [53,54] and thereby converts inactive Akt to active Akt. Phosphorylated Akt/active Akt migrates to cytosol,

mitochondria, and nucleus. Nuclear Akt then fulfills the antiapoptotic role [55] through phosphorylation of transcription factors such as cAMP response elementbinding (CREB) protein [40,56,57]. Phosphorylated CREB binds to DNA as a dimer via leucine zipper motif and recognizes cAMP-response elements (CREs). CRE then stimulates the transcription of genes and results in the expression of the antiapoptotic factor, Bcl-2 (Bcl2-xL) [58]. The regulation of apoptosis by PI3K/Akt/mTOR pathway is schematically depicted in Figure 5.3.

5.3 FREE RADICALS IN CELLS Living cells are always subjected to the hazardous effects of exogenously or endogenously produced highly reactive oxidizing molecules and easily acquire electrons from oxidized molecules with which they remain in contact, such as all cellular biomolecules, generating chain reactions and ultimately leading to cell structure damage [59]. Products of oxidative metabolism that provoke oxidative injury are collectively called reactive oxygen species (ROS), which includes superoxide, H2O2, and the hydroxyl radical. They are formed either by the loss of a single electron from a nonradical or by the gain of a single electron by a nonradical. Lipid peroxidation is one of the wellstudied consequences of ROS. Lipid peroxidation initiators, such as ROS, contribute to the signal transduction cascade that controls cell death and proliferation [60]. The free radicals can trigger cellular damage by covalent binding to macromolecules and enhance

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

72

5. THE ROLE OF mTOR, AUTOPHAGY, APOPTOSIS, AND OXIDATIVE STRESS DURING TOXIC METAL INJURY

lipid peroxidation that depletes the stress of antioxidant enzymes. Inhibition of antioxidant enzymes leads to oxidative stress [61,62]. ROS has a major biological impact due to its endogenous production and high concentration in the cells. It is well known that the principal source of ROS in the cell is the mitochondrial respiratory chain [59,63]. Metal-mediated formation of free radicals causes various modifications to DNA bases, enhances lipid peroxidation, and alters calcium and sulfhydryl homeostasis [64]. Lipid peroxides, formed by the attack of radicals on polyunsaturated fatty acid residues of phospholipids, can further react with redox metals finally producing mutagenic and carcinogenic malondialdehyde, 4-hydroxynonenal, and other exocyclic DNA adducts (etheno and/or propano adducts). While iron (Fe), copper (Cu), chromium (Cr), and cobalt (Co) undergo redox-cycling reactions, for mercury (Hg), cadmium (Cd), and nickel (Ni), the primary route for their toxicity is depletion of glutathione and bonding to sulfhydryl groups of proteins [64,65]. Metal toxicity therefore generates oxidative stress in cell.

` -VIS mTOR 5.4 STRESS VIS-A The TOR pathway is an evolutionarily conserved signaling module regulating cell growth in response to a variety of internal and external stimuli, generating oxidative stress [66,67]. mTOR acts in two categories in the surveillance of stress conditions, illustrated in Figure 5.4. The oxidative stress-input to mTOR

generally lies upstream of the TSC complex. ROS induces transcriptional alteration of cellular proteins and impinges the mTOR network signaling [66]. ROS in turn inhibits mTORC1 by direct phosphorylation of TSC2 and indirect activation of AMPK. AMPK results ROS generation, phosphorylates, and thereby activates TSC2, resulting in mTORC1 inhibition [31]. On the contrary, oxidative stress can persuade mTORC1 to play a distinct role in the last stage of autophagy [68,69]. Generally, mTORC1 forms a complex with autophagy-related proteins (ATG) named ATG1, ATG 13, and ATG101, which in turn phosphorylates ATG1 and ATG13 to inhibit the ATG activity in autophagy [28,70]. Induction of stress can reactivate mTORC1 and thereby breaks the mTORC1ATG protein complex to free ATG1 [69]. ATG1 then leads to the formation of protolysosomal extension by lysosomal-associated membrane protein 1 and microtubule-associated protein 1A/1B light chain 3 (LC3) from autophagolysosome and ultimately mature into functional lysosome by autophagic lysosome reformation [68,69,71]. Toxicants can trigger cellular pathways classified broadly as death and survival signals. Each organ has its own critical threshold towards a toxicant, regulated by an intricate signaling system. Depending on the concentration and duration of exposure to toxicants, cells can utilize a coordinated, preprogrammed signaling pathway to maintain the normal homeostasis of the organ. Considering the complex and interactive mechanisms involved in maintenance of homeostasis of the organism, the present review is further aimed to FIGURE 5.4 mTOR.

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

Oxidative stress and

5.6 CADMIUM (CD) TOXICITY

monitor the role of mTOR during cellular injury imposed by toxic metals. Generally, oxidative stress is considered as the first response to toxic metals and a volley of interactions follows to maintain the integrity of the cellular and molecular homeostasis through several coordinated signaling pathways. Toxic metals are usually those which are highly reactive to the lipid membranes of the cells, resulting in formation of injurious free oxygen or hydroxyl radicals. An attempt has been made to weigh the deleteriousness of metals of environmental concern affecting biological systems and the interrelationship of mTOR with different types of cell death.

5.5 ARSENIC (AS) TOXICITY The main cause of toxic effects of metals is generation of ROS. It has been demonstrated that As is a potential inducer of oxidative stress as evidenced by DNA damage, by superoxide and hydrogen peroxide [72], and by disruption of mitosis while promoting apoptosis [73,74]. It has been amply demonstrated that inorganic As is more toxic than organo-As compounds [75]. Moreover, arsenite induces apoptosis much more strongly than arsenate [76,77] and there is a definite role of free radicals in As toxicity [78]. Various studies on the role of As in cellular apoptosis demonstrate 44.5% apoptosis at a concentration of 200 μM arsenite of the JB6C141 cell types [79]; 10 and 20 μM As2O3 initiated apoptosis in a neural cell line and 40 μM caused death in these cells [80]. Paradoxically, As has both carcinogenic [81,82] and effective chemotherapeutic activity [8385] in acute promyelocytic leukemia (APL), inducing apoptosis within a tumor cell population where a high cumulative dose (340430 mg) of As2O3 was found to cause a complete remission of APL [86]. Thus, normal organs and tissues are liable to As toxicity evidenced by triggering of signaling pathways, such as caspasedependent or -independent, mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase (JNK), extracellular-signal-regulated kinase (ERK), and p38 pathways. Arsenic-induced apoptosis at pharmacological concentrations was mediated by caspase-dependent pathways through mitochondrial checkpoints at 10 μM As2O3 where the activity of ERK and JNK is dependent on caspase activity, while p38 and MAPK were found to be operative in inducing apoptosis at 40 μM of As2O3 [87]. As-induced apoptosis of chronic lymphocytic leukemia cells involves inactivation of the kinase AKT and a blockade of the transcriptional factor NF-κB, as well as up-regulation of PTEN and downregulation of X-linked inhibitor of apoptosis protein [88]. As can paradoxically increase mTOR activation

73

and engagement of downstream mTOR effectors in BCR-ABL (tyrosine kinase inhibitor) expressing cells [89] or acute myeloid leukemia cells [90]. The combinations of As with mTORC1 inhibitor (mTORC1i) of rapamycin result in increased apoptosis and enhanced suppressive effects on primary leukemic progenitors [91]. Moreover, the combination of As and mTORC1i may overcome the feedback loop by decreasing activation of the MAPK and AKT signaling pathways in breast cancer [92]. Since carcinogenesis and apoptosis are both tightly linked to the PI3K/Akt/mTOR pathway, it could be expected that As31 would differentially target the pathway to accomplish these opposite effects on cell fate [93]. As2O3 has been reported to induce autophagic cell death without activation of caspase-dependent apoptotic cell death [9496]. Arsenic was reported to induce autophagy at a concentration of 2 μM in malignant glioma cell lines [94] and at 6 μM in lymphoblastoid cell lines [97]. Autophagy plays complex roles in Asinduced death of human promyelocytic leukemia 60 cells; it inhibits As-induced apoptosis in the initiation stage, but amplifies As-mediated apoptotic program on persistent activation of autophagy [98]. Furthermore, carcinogenicity of As can fabricate overproduction of interleukin 6, which sufficiently blocks autophagy by supporting Beclin-1/Mcl-1/mTOR interaction [99].

5.6 CADMIUM (CD) TOXICITY Cadmium (Cd), the toxic transitional metal, is mainly released from smelting and refining of metals and cigarette smoking, resulting in the pollution of water, air, and soil [65,100]. Cd compounds are used as stabilizers, color pigments, rechargeable batteries, alloys, and can be found in some fertilizers. Cd production, consumption, and emissions to the environment increased worldwide dramatically during the twentieth century [101]. Cd can be absorbed and accumulated in plants and animals and thereby can be accumulated in the human body either through direct exposure to Cd-contaminated environment or by the food chain. Such accumulation contributes to carcinogenesis, immunodepression, and neurodegeneration [100,102,103]. A very long biological half-life of Cd may be responsible for the accumulation in many human organs, including kidney [104], liver [105], lung [106], testis [107], bone [108], and blood system [109]. Cd has a high bloodbrain barrier permeability; therefore chronic Cd exposure affects the nervous system, including learning disabilities and hyperactivity in children [110,111], olfactory dysfunction, neurobehavioral defects in attention, psychomotor

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

74

5. THE ROLE OF mTOR, AUTOPHAGY, APOPTOSIS, AND OXIDATIVE STRESS DURING TOXIC METAL INJURY

speed, and memory in industrial workers [112] and also Parkinson’s disease by acute Cd poisoining [113]. Cd toxicity is amplified as a consequence of the long biological half-life of the metal and has been associated with blockage of oxidative phosphorylation, glutathione depletion, inhibition of antioxidant enzymatic activity, production of oxidative stress, DNA damage, reduction of protein synthesis, and cell death [65,114]. Cd-induced toxicity is closely associated with the production of ROS [115117]. Cd accelerates the generation of ROS by depleting cellular glutathione and antioxidant enzymes, such as superoxide dismutase and catalase [115,118] and by inhibiting the electron transfer chain in the mitochondria [119]. It is surmised that the effect of Cd toxicity depends on its concentration and the duration of exposure inducing both autophagic- and apoptotic-related pathways via ROS generation [65,120,121]. However, the mechanisms underlying Cd-induced autophagy are not yet completely understood. Cd (010 μM) has been found to induce autophagy through ROS-dependent activation of the serine/threonine kinase 11/liver kinase B1—AMPK—mTOR signaling in skin epidermal cells in a concentration- and time-dependent manner [122]. Cd could also provoke oxidative stress generating ROS inside JB6 cells, which in turn signals energy-sensing LKB1-AMP-activated protein kinase pathway after 12 h of Cd treatment (10 μM) and the effect is further augmented at 24 h after the treatment. AMPK, a heterotrimeric protein complex, is downstream of LKB1 in a signaling pathway that regulates energy homeostasis [123,124]. LKB/AMPK signaling can act as the upstream of mTOR signaling [125,126]. Cd-induced intracellular level of ROS signals LKB1, which in turn phosphorylates AMPK and thus inhibits mTORC1 triggering autophagy in JB6 cells in a significant manner [123,124]. It is commonly accepted that Cd induces apoptotic cell death in cellular systems [127,128]. However, Cd induces apoptosis by either caspase-dependent [129,130] or caspase-independent mechanisms [131,132] depending on the target cell type. Cd-induced apoptotic cell death in JB6 cells was found to be dependent on Ca21and H2O2-mediated JNK and p53 signaling [133]. Interestingly, autophagic cell death was also induced by Cd (010 μM) by the ROS-dependent LKB1-AMPK [122]. Moreover, 10 μM of Cd not only causes apoptosis in skin epidermal cells (JB6) but also leads to necrotic cell death [133] in a time-dependent manner from 1 to 24 h. Generation of ROS due to Cd toxicity elevates the intracellular calcium (Ca21) level. The increase of free calcium ions is believed to be one of the main mechanisms involved in Cd-induced apoptosis by stimulating the generation of H2O2 [133]. Ca21 regulates JNK signaling and also effects H2O2 generation. In turn H2O2 generated

by Cd regulates both activator protein-1 (AP-1) and p53 pathways in JB6 cells. Collectively, Cd promotes apoptosis mainly through Ca21-JNK-growth arrest and DNA-damaged inducible protein (GADD45α) as well as a caspase-independent pathway, H2O2-AP-1-p53-apoptosis-inducing-actor (AIF) signaling cascades [133]. Ca21 also leads to the activation of MAPKs in Cdexposed mesangial cells [120] and in tributyltin-treated human T-cell lines [134]. It has been reported [100,135] that Cd at 0120 mmol/L and 10 and/or 20 μM, respectively, induced neuronal apoptosis by the activation of MAPK and mTOR as well as JNK and PTEN-Akt/ mTOR network. Cd toxicity activates ROS production mediated by NADPH oxidase, which in turn activates MAPK [136138]. Ca21-dependent protein kinase and PI3K also stimulate MAPK during Cd toxicity [138]. Phospho-p38-MAPK signals GADD45α and induces apoptosis by caspase-3 activation [136,139,140]. JNK and PTEN-Akt/mTOR network diminishes antiapoptotic properties by decreasing their protein stability, which eventually releases Cyt c from mitochondria and induces caspase-dependent apoptosis [51,135]. The effects of Cd toxicity are schematically summarized in Figure 5.5.

5.7 LEAD (PB) TOXICITY Lead (Pb), a well-known heavy metal, can cause pathophysiological changes in several organ systems including the central nervous system (CNS), cardiovascular, hematopoietic, reproductive, gastrointestinal, and renal systems [141,142]. Pb had modest early uses in ancient medicines and cosmetics, however, in the modern era it has had industrial uses in building materials, paints, and gasoline [143]. Pb has become ubiquitous in the environment. Pb exposure mainly occurs through the respiratory and gastrointestinal systems and absorbed Pb is stored in soft tissues. Inhaled Pb (B3040%) enters the bloodstream [144]; once absorbed, 99% of Pb is retained in the blood for approximately 3035 days and over the following 46 weeks it is dispersed and accumulated in other tissues [145]. Pb serves no useful purpose in the human body and its unfortunate presence in the body can lead to toxic effects, regardless of the exposure pathway adopted [146]. The effects of Pb on Ca21 fluxes and Ca21-regulated events are the major mechanisms of lead neurotoxicity [142,147,148]. Pb can cause abnormal hyperphosphorylation of tubulin-associated unit (tau) protein and accumulation of α-synuclein, inducing hippocampal injury affecting the ability to learning and causing memory damage. Pathological hyperphosphorylation and aggregation of tau deposits as neurofibrillary tangles in the brains of those with Alzheimer’s disease and other related neurodegenerative disorders are called tauopathies [142,149]. α-Synuclein,

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

75

5.8 MERCURY (HG) TOXICITY

FIGURE 5.5

expressed in the CNS neurons and localized around synaptic vesicles in presynaptic terminals can cause Parkinson’s disease or a related disorder, dementia with Lewy bodies through genetic alteration or point mutation [142,150]. The deleterious effects of Pb exposures can involve endoplasmic reticulum (ER) stress in cardiofibroblasts which consecutively generate ROS and a direct depletion of the antioxidant reserves [145,148]. Significant effects have been found on various fundamental cellular processes like protein folding and maturation, ionic transportation, enzyme regulation leading to cell stress [151], and finally cell death, such as autophagy and apoptosis [142,148]. ER has an essential role in multiple cellular processes, such as the folding of secretory membrane proteins, calcium homeostasis, and lipid biosynthesis. When ER trans-membrane sensors detect the accumulation of unfolded proteins, the unfolded protein response (UPR) is initiated to cope with the resulting ER stress [152]. Under stress conditions, a cell performs several adaptive alterations such as autophagy and apoptosis. Pb toxicity can induce robust UPR and ER stress which can mediate autophagy and/or apoptosis to protect the internal system of the cell [142,148]. The regulation of autophagy and to some extent apoptosis is mediated by mTORC1 [32,36]. mTORC1 activity is critical for de novo protein synthesis; the reduced mTORC1 activity may decrease the rate of synthesis of new proteins, which may partly attenuate ER stress on protein folding and processing reactions [148]. Recently it has been reported that Pb

Toxic signals of Cd.

toxicity inhibits the mTORC1 pathway [142,148]. Depending on the concentration and duration of Pb exposure, the internal cellular system then decides to undertake the cellular homeostatic pathway, autophagy/ apoptosis, mediated by the ATG proteins. Inhibition of mTORC1 easily shifts the cell towards the induction of autophagy via ATG1ATG13ATG101 complex and Beclin-1 (ATG6)ATG14 complex [23,25]. On the other hand, Beclin-1 plays a significant role in the network of cellular homeostasis, focusing on the cross-regulation between autophagy and apoptosis [153,154]. Pb toxicity can induce apoptosis to promote the activation of caspase-3 via Beclin-1 [142]. It cleaves Beclin-1 from the Beclin-1ATG14 complex, triggers proapoptotic proteins in mitochondria to release Cyt c which consequently induces apoptosis [153]. The effects of Pb toxicity are portrayed in Figure 5.6.

5.8 MERCURY (HG) TOXICITY Hg is a well-established environmental toxicant. Hg is known to cause several physiological and biochemical disturbances in mammals. Most of these biochemical reactions are exerted through formation of covalent bonds between sulfur and Hg. Three forms of Hg exist: elemental, inorganic, and organic. Each of them has its own profile of toxicity. Elemental Hg vapor is highly lipid-soluble allowing it to readily cross cellular membranes. It can also be oxidized to mercuric salts from monovalent Hg

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

76

5. THE ROLE OF mTOR, AUTOPHAGY, APOPTOSIS, AND OXIDATIVE STRESS DURING TOXIC METAL INJURY

FIGURE 5.6 Toxic signals of Pb.

compounds. Thus, when ingested, Hg will be more rapidly absorbed and produce greater toxicity [155]. The distribution kinetics of radioactive Hg in different hepatocellular fractions revealed that Hg treatment increases nuclear and liposomal protein content significantly [156]. On the other hand, generation of an acutephase response was observed in rabbit treated with mercuric chloride (HgCl2) and remarkable changes in phospholipid profile and acetylcholinesterase activity, [157] while in rat Hg has been found to induce C reactive protein along with a metallothionein-like protein [158]. HgCl2 is also known to damage DNA in fibroblasts of rat and mouse embryos [159,160] and exposure to a nonlethal dose of Hg induces oxidative-stress-mediated apoptosis in rat liver [87,161]. Five micromolar HgCl2 was shown to damage cellular DNA by a nonapoptotic mechanism in the U-937 cell line [162]. Interestingly, recent research [35,163165] revealed that 5 μM HgCl2 drives autophagy and apoptosis in different cell types of rat liver. Covalent-conjugation of autophagic proteins (ATG5-ATG12-LC3B) stimulates cell death in rat hepatocytes, modulated by Keap1-p62, ERK-p38, and DRAMp53 regulator proteins through inhibition of mTOR [35,164]. Concurrently, autophagic proteins ATG12 and LC3B exerted a significant role in triggering apoptosis in 5 μM HgCl2-treated rat oval cells [165].

pathways. Oxidative stress and PI3K, Akt, and mTOR pathways are intimately related to determine the cell fate [166]. The significant biological role of PI3K, Akt, and mTOR is involved in the modulation of apoptosis, autophagy, disorders of cellular metabolism, acute nervous system injury, and chronic neurodegeneration [20,21,166168]. Moreover, MTOR activates CREB1 through suppression of autophagy, sensitizing subsequent apoptosis [169]. Different metals seem to prefer different signaling pathways and display diverse potential to trigger mTOR pathway cascades [49,135,148]. mTOR signaling pathways integrate both intracellular and extracellular signals and serves as a central regulator of metal toxicity in cellular adaptation. In conclusion, this review elucidates an important role for mTOR modulation of oxidative stress and cell death signaling due to toxic metal injury.

5.10 FUTURE PERSPECTIVES Metal-induced oxidative stress plays a significant role in the modulation of apoptosis and autophagy. In this aspect, the PI3K, Akt, and mTOR cascade offers new inroads to the development of novel therapies. Nowadays, with increasing use of nanoparticles in the cosmetics and food industries, attention needs to be focused on metal-nanoparticle-induced oxidative stress and PI3K/Akt/mTOR signaling pathway. It appears to be an important mechanism in manifesting pathways leading to several cardiovascular diseases. New research is focusing on mTOR signaling components in the maintenance of insulin signaling to prevent diabetes mellitus. However, mTOR signaling can also trigger cellular proliferative pathways that can promote aggressive tumor growth. On the other hand, prevention of PI3K and Akt activation can block medulloblastoma growth. Toxicology studies have unraveled that the PI3K/Akt/mTOR pathway is commonly targeted by toxins, especially metals, to produce several hazardous conditions. The PI3K/Akt/mTOR targeted therapy shows therapeutic efficacy, albeit with a variety of toxic side effects. A thorough understanding of the role of the mTOR pathway, with a systemic perspective and computational modeling appears to be the future center of interest.

5.9 CONCLUDING REMARKS The last decade has seen a rapid rise in interest in mTOR signaling cascades. Metal affronted oxidative stress determines the down-regulation and upregulation of mTOR signaling to maintain cellular homeostasis by different programmed cell death

Acknowledgments SC is grateful to the National Academy of Sciences, India (NASI), for the award of Research Associate and SB acknowledges NASI for the award of Senior Scientist Platinum Jubilee Fellowship. The authors are grateful to Shuvasree Sarkar, UGC-BSR SRF, for critically going through the manuscript.

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

REFERENCES

References [1] Loewith R, Hall MN. Target of rapamycin (TOR) in nutrient signaling and growth control. Genetics 2011;189:1177201. [2] Benjamin D, Colombi M, Moroni C, Hall MN. Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov 2011;10:86880. [3] Ve´zina C, Kudelski A, Sehgal SN. Rapamycin (AY-22,989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. J Antibiot 1975;28:7216. [4] Harding MW, Galat A, Uehling DE, Schreiber SL. A receptor for the immunosuppressant FK506 is a cis-trans peptidyl-prolyl isomerase. Nature 1989;341:75860. [5] Siekierka JJ, Wiederrecht G, Greulich H, Boulton D, Hung SH, et al. The cytosolic-binding protein for the immunosuppressant FK-506 is both a ubiquitous and highly conserved peptidylprolyl cis-trans isomerase. J Biol Chem 1990;265:2101115. [6] Brown EJ, Albers MW, Shin TB, Ichikawa K, Keith CT, et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature 1994;369:7568. [7] Chen Y, Chen H, Rhoad AE, Warner L, Caggiano TJ, et al. A putative sirolimus (rapamycin) effector protein. Biochem Biophys Res Commun 1994;203:17. [8] Chiu MI, Katz H, Berlin V. RAPT1, a mammalian homolog of yeast Tor, interacts with the FKBP12/rapamycin complex. Proc Natl Acad Sci USA 1994;91:125748. [9] Raught B, Gingras AC, Sonenberg N. The target of rapamycin (TOR) proteins. Proc Natl Acad Sci USA 2001;98:703744. [10] Sabatini DM, Erdjument-Bromage H, Lui M, Tempst P, Snyder SH. RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs. Cell 1994;78:3543. [11] Sabers CJ, Martin MM, Brunn GJ, Williams JM, Dumont FJ, et al. Isolation of a protein target of the FKBP12rapamycin complex in mammalian cells. J Biol Chem 1995;270:81522. [12] Abraham RT, Wiederrecht GJ. Immunopharmacology of rapamycin. Annu Rev Immunol 1996;14:483510. [13] Hoekstra MF. Responses to DNA damage and regulation of cell cycle checkpoints by the ATM protein kinase family. Curr Opin Genet Dev 1997;7:1705. [14] Hunter T. When is a lipid kinase not a lipid kinase—when it is a protein-kinase. Cell 1995;83:14. [15] Hands SL, Proud CG, Wyttenbach A. mTOR’s role in ageing: protein synthesis or autophagy? Aging 2009;1:58697. [16] Blagosklonny MV, Hall MN. Growth and aging: a common molecular mechanism. Aging 2009;1:35762. [17] Tchevkina E, Komelkov A. Protein phosphorylation as a key mechanism of mTORC1/2 signaling pathways. Intech Open Sci 2012. Available from: ,http://dx.doi.org/10.5772/48274. [accessed in April 2015]. [18] Gingras AC, Raught B, Sonenberg N. Regulation of translation initiation by FRAP/mTOR. Genes Dev 2001;15:80726. [19] Perry J, Kleckner N. The ATRs, ATMs, and TORs are giant HEAT repeat proteins. Cell 2003;112:1515. [20] Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell 2012;149:27493. [21] Sengupta S, Peterson TR, Sabatini DM. Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress. Mol Cell 2010;40:31022. [22] Bergmann A. Autophagy and cell death: no longer at odds. Cell 2007;131:10323. [23] Barth S, Glick D, Macleod KF. Autophagy: assays and artifacts. J Pathol 2010;221:11724. [24] Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol 2010;221:312.

77

[25] Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, et al. Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy 2008;4:15175. [26] Mizushima N, Ohsumi Y, Yoshimori T. Autophagosome formation in mammalian cells. Cell Struct Funct 2002;27:4219. [27] Blommaart EF, Krause U, Schellens JP, Vreeling-Sindelarova H, Meijer AJ. The phosphatidylinositol 3-kinase inhibitors wortmannin and LY294002 inhibit autophagy in isolated rat hepatocytes. Eur J Biochem 1997;243:2406. [28] Roach PJ. AMPK-ULK1-autophagy. Mol Cell Biol 2011;31:30824. [29] Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self digestion. Nature 2008;451:106974. [30] Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, et al. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell 2008;30:21426. [31] Inoki K, Li Y, Xu T, Guan KL. Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Gene Dev 2003;17:182934. [32] Martin TD, Chen XW, Kaplan REW, Saltiel AR, Walker CL, et al. Ral and Rheb GTPase activating proteins integrate mTOR and GTPase signaling in aging, autophagy, and tumor cell invasion. Mol Cell 2014;53:20920. [33] Budanov AV, Karin M. p53 target genes sestrin1 and sestrin2 connect genotoxic stress and mTOR signaling. Cell 2008;134:45160. [34] Feng Z, Zhang H, Levine AJ, Jin S. The coordinate regulation of the p53 and mTOR pathways in cells. Proc Natl Acad Sci USA 2005;102:82049. [35] Chatterjee S, Ray A, Mukherjee S, Agarwal S, Kundu R, et al. Low concentration of mercury induces autophagic cell death in rat hepatocytes. Toxicol Ind Health 2012;30:61120. [36] Castedo M, Ferri KF, Kroemer G. Mammalian target of rapamycin (mTOR): pro- and anti-apoptotic. Cell Death Differ 2002;9:99100. [37] Lockshin RA, Zakeri Z. Cell death in health and disease. J Cell Mol Med 2007;11:121424. [38] Engelman JA. Targeting PI3K signaling in cancer: opportunities, challenges and limitations. Nat Rev Cancer 2009;9:55062. [39] Fulda S, Galluzzi L, Kroemer G. Targeting mitochondria for cancer therapy. Nat Rev Drug Discov 2010;9:44764. [40] Fulda S. Shifting the balance of mitochondrial apoptosis: therapeutic perspectives. Front Oncol 2012;2:14. [41] Scott PH, Brunn GJ, Kohn AD, Roth RA, Lawrence JC. Evidence of insulin-stimulated phosphorylation and activation of the mammalian target of rapamycin mediated by a protein kinase B signaling pathway. Proc Natl Acad Sci USA 1998;95:77727. [42] Nave BT, Ouwens M, Withers DJ, Alessi DR, Shepherd PR. Mammalian target of rapamycin is a direct target for protein kinase B: identification of a convergence point for opposing effects of insulin and amino-acid deficiency on protein translation. J Biochem 1999;344:42731. [43] Sekulic A, Hudson CC, Homme JL, Yin P, Otterness DM, et al. A direct linkage between the phosphoinositide 3-kinaseAKT signaling pathway and the mammalian target of rapamycin in mitogen-stimulated and transformed cells. Cancer Res 2000;60:350413. [44] Reynolds TH, Bodine SC, Lawrence JC. Control of Ser2448 phosphorylation in the mammalian target of rapamycin by insulin and skeletal muscle load. J Biol Chem 2002;277:1765762. [45] Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes Dev 2004;18:192645.

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

78

5. THE ROLE OF mTOR, AUTOPHAGY, APOPTOSIS, AND OXIDATIVE STRESS DURING TOXIC METAL INJURY

[46] Shaw RJ, Cantley LC. Ras, PI(3)K and mTOR signaling controls tumour cell growth. Nature 2006;441:42430. [47] Gardai SJ, Hildeman DA, Frankel SK, Whitlock BB, Frasch SC, et al. Phosphorylation of Bax Ser184 by Akt regulates its activity and apoptosis in neutrophils. J Biol Chem 2004;279: 2108595. [48] Dan HC, Sun M, Kaneko S, Feldman RI, Nicosia SV, et al. Akt phosphorylation and stabilization of X-linked inhibitor of apoptosis protein (XIAP). J Biol Chem 2004;279:540512. [49] Qi XJ, Wildey GM, Howe PH. Evidence that Ser87 of BimEL is phosphorylated by Akt and regulates BimEL apoptotic function. J Biol Chem 2006;281:81323. [50] Yamaguchi H, Wang HG. The protein kinase PKB/Akt regulates cell survival and apoptosis by inhibiting Bax conformational change. Oncogene 2001;20:777986. [51] Maurer U, Charvet C, Wagman AS, Dejardin E, Green DR. Glycogen synthase kinase-3 regulates mitochondrial outer membrane permeabilization and apoptosis by destabilization of MCL-1. Mol Cell 2006;21:74960. [52] Mills JR, Hippo Y, Robert F, Chen SMH, Malina A, et al. mTORC1 promotes survival through translational control of Mcl-1. Proc Natl Acad Sci USA 2008;105:108538. [53] Martelli AM, Evangelisti C, Chiarini F, Blalock WL, Papa V, et al. The Phosphatidylinositol 3-Kinase/Akt/Mammalian target of rapamycin signaling network as a new target for acute myelogenous leukemia therapy. Cancer Ther 2007;5:30930. [54] Martelli AM, Evangelisti C, Chiarini F, Grimaldi C, Cappellini A, et al. The emerging role of the phosphatidylinositol 3-kinase/ Akt/mammalian target of rapamycin signaling network in normal myelopoiesis and leukemogenesis. Biochim Biophys Acta 2010;1803:9911002. [55] Martelli AM, Faenza I, Billi AM, Manzoli L, Evangelisti C, et al. Intranuclear 30 -phosphoinositide metabolism and Akt signaling: new mechanisms for tumorigenesis and protection against apoptosis? Cell Signal 2006;18:11017. [56] Du K, Montminy M. CREB is a regulatory target for the protein kinase Akt/PKB. J Biol Chem 1998;273:323779. [57] Wang L, Piguet AC, Schmidt K, Tordjmann T, Dufour JF. Activation of CREB by tauroursodeoxycholic acid protects cholangiocytes from apoptosis induced by mTOR inhibition. Hepatology 2005;41:124151. [58] Wilson BE, Mochon E, Boxer LM. Induction of bcl-2 expression by phosphorylated CREB proteins during B-cell activation and rescue from apoptosis. Mol Cell Biol 1996;16:554656. [59] Filomeni G, Zio DD, Cecconi F. Oxidative stress and autophagy: the clash between damage and metabolic needs. Cell Death Differ 2015;22:37788. [60] Marnett LJ. Lipid peroxidation—DNA damage by malondialdehyde. Mutat Res 1999;424:8395. [61] Kyle ME, Micaadei S, Nakae D, Farber JL. Superoxide dismutase and catalase protect cultured hepatocytes from the cytotoxicity of acetaminophen. Biochem Biophys Res Commun 1987;149:88996. [62] Kurose I, Higuchi H, Miura S, Saito H, Watanabe N, et al. Oxidative stress mediated apoptosis of hepatocytes exposed to acute ethanol intoxication. Hepatology 1997;25:36878. [63] Lee J, Giordano S, Zhang J. Autophagy, mitochondria and oxidative stress: cross-talk and redox signaling. J Biochem 2012;441:52340. [64] Valko M, Morris H, Cronin MT. Metals, toxicity and oxidative stress. Curr Med Chem 2005;12:1161208. [65] Chatterjee S, Sarkar S, Bhattacharya S. Toxic metals and autophagy. Chem Res Toxicol 2014;27:1887900. [66] Reiling JH, Sabatini DM. Stress and mTOR signaling. Oncogene 2006;25:637383.

[67] Faghiri Z, Bazana NG. PI3K/Akt and mTOR/p70S6K pathways mediate neuroprotectin D1-induced retinal pigment epithelial cell survival during oxidative stress-induced apoptosis. Exp Eye Res 2010;90:71825. [68] Yu L, McPhee CK, Zheng L, Mardones GA, Rong Y, et al. Autophagy termination and lysosome reformation regulated by mTOR. Nature 2010;465:9426. [69] Hayat MA. Autophagy: cancer, other pathologies, inflammation, immunity, infection, and aging. Autophagy 2015;5:148. Available from: ,http://dx.doi.org/10.1016/B978-0-12-801033-4.00001-1.. [70] Jung CH, Jun CB, Ro SH, Kim YM, Otto NM, et al. ULK-Atg13FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol Cell Biol 2009;7:19922003. [71] Chen G, Yu L. Autophagic lysosome reformation. Exp Cell Res 2012;319:1426. [72] Kitchin KT. Recent advances in arsenic carcinogenesis: modes of action, animal model systems, and methylated arsenic metabolites. Toxicol Appl Pharmacol 2001;172:24961. [73] States JC, Reiners JJ, Pounds JG, Kaplan DJ, Beauerle BD, et al. Arsenite disrupts mitosis and induces apoptosis in SV40transformed human skin fibroblasts. Toxicol Appl Pharmacol 2002;180:8391. [74] Datta S, Saha DR, Ghosh D, Majumdar T, Bhattacharya S, et al. Sub-lethal concentration of arsenic interferes with the proliferation of hepatocytes and induces in vivo apoptosis in Clarias batrachus L. Comp Biochem Physiol C 2007;145(Suppl. 3):33949. [75] Kaise T, Yamauchi H, Horiuchi Y, Tani T, Watanabe S, et al. A comparative study on acute toxicity of methylarsonic acid, dimethylarsinic acid and trimethylarsine oxide in mice. Appl Organomet Chem 1989;3:2737. [76] Ma DC, Sun YH, Chang KZ, Ma XF, Huang SL, et al. Selective induction of apoptosis of NB4 cells from G2-M phase by sodium arsenite at lower doses. Eur J Haematol 1998;61:2735. [77] Zeng H. Arsenic suppresses necrosis induced by selenite in human leukemia HL-60 cells. Biol Trace Elem Res 2001;83:115. [78] Bashir S, Sharma Y, Irshad M, Nag TC, Tiwari M, et al. Arsenic induced apoptosis in rat liver following repeated 60 days exposure. Toxicology 2006;217:6370. [79] Chen NY, Ma WY, Yang CS, Dong Z. Inhibition of arseniteinduced apoptosis and AP-1 activity by epigallocatechin-3gallate and theoflavins. J Environ Pathol Toxicol Oncol 2000;19:28795. [80] Milton AG, Zalewski PD, Ratnaike RN. Zinc protects against arsenic-induced apoptosis in a neuronal cell line, measured by DEVD-caspase activity. Biometals 2004;17:70713. [81] Lu T, Liu J, LeCluyse EL, Zhou YS, Cheng ML, et al. Application of cDNA microarray to the study of arsenicinduced liver diseases in the population of Guizhou, China. Toxicol Sci 2001;59:18592. [82] Schuliga M, Chouchane S, Snow ET. Up regulation of glutathione-related genes and enzyme activities in cultured human cells by sublethal concentrations of inorganic arsenic. Toxicol Sci 2002;70:18392. [83] Look AT. Arsenic and apoptosis in the treatment of acute promyelocytic leukemia. J Natl Cancer Inst 1998;90:868. [84] Kann S, Estes C, Reichard JF, Huang MY, Sartor MA, et al. Butylhydroquinone protects cells genetically deficient in glutathione biosynthesis from arsenite-induced apoptosis without significantly changing their prooxidant status. Toxicol Sci 2005;87(Suppl. 2):36584. [85] Liu ZM, Huang HS. As2O3-induced c-Src/EGFR/ERK signaling is via Sp1 binding sites to stimulate p21WAF1/CIP1 expression in human epidermoid carcinoma A431 cells. Cell Signal 2005;18:24455.

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

REFERENCES

[86] Huang SY, Yang CH, Chen YC. As2O3 therapy for acute promyelocytic leukemia: an unusual salvage therapy. Leuk Lymphoma 2000;38:28393. [87] Ray A, Roy S, Agarwal S, Bhattacharya S. As2O3 toxicity in rat hepatocytes: manifestation of caspase mediated apoptosis. Toxicol Ind Health 2008;24:64353. [88] Redondo-Mun˜oz J, Escobar-Dı´az E, Herna´ndez del Cerro M, Pandiella A, Terol MJ, et al. Induction of B-chronic lymphocytic leukemia cell apoptosis by arsenic trioxide involves suppression of the PI3K/Akt survival pathway via JNK activation and PTEN upregulation. Clin Cancer Res 2010;16:438291. [89] Yoon P, Giafis N, Smith J, Mears H, Katsoulidis E, et al. Activation of mammalian target of rapamycin and the p70 S6 kinase by arsenic trioxide in BCR-ABL-expressing cells. Mol Cancer Ther 2006;5:281523. [90] Altman JK, Yoon P, Katsoulidis E, Kroczynska B, Sassano A, et al. Regulatory effects of mammalian target of rapamycinmediated signals in the generation of arsenic trioxide responses. J Biol Chem 2008;283:19922001. [91] Goussetis DJ, Platanias LC. Arsenic trioxide and the PI3K/ AKT pathway in chronic lymphocytic leukemia. Clin Cancer Res 2010. Available from: http://dx.doi.org/10.1158/10780432.CCR-10-0072. [92] Guilbert C, Annis MG, Dong Z, Siegel PM, Miller Jr WH, et al. Arsenic trioxide overcomes rapamycin-induced feedback activation of AKT and ERK signaling to enhance the anti-tumor effects in breast cancer. PLOS One 2013;8:112. [93] Li T, Wang G. Computer-aided targeting of the PI3K/Akt/ mTOR Pathway: toxicity reduction and therapeutic opportunities. Int J Mol Sci 2014;15:1885691. [94] Kanzawa T, Kondo Y, Ito H, Kondo S, Germano I. Induction of autophagic cell death in malignant glioma cells by arsenic trioxide. Cancer Res 2003;63:21038. [95] Kanzawa T, Zhang L, Xiao L, Germano IM, Kondo Y, et al. Arsenic trioxide induces autophagic cell death in malignant glioma cells by upregulation of mitochondrial cell death protein BNIP3. Oncogene 2005;24:98091. [96] Tsai SC, Yang JS, Peng SF, Lu CC, Chiang JH, et al. Bufalin increases sensitivity to AKT/mTOR-induced autophagic cell death in SK-HEP-1 human hepatocellular carcinoma cells. Int J Oncol 2012;41:143142. [97] Bolt M, Byrd RM, Klimecki WT. Autophagy is a biological target of arsenic. In: Jean JS, Bundschuh J, Bhattacharya P, editors. Arsenic in geosphere and human diseases. Boca Raton, FL: CRC Press; 2010. p. 2912. [98] Yang YP, Liang ZQ, Gao B, Jia YL, Qin ZH. Dynamic effects of autophagy on arsenic trioxide-induced death of human leukemia cell line HL60 cells. Acta Pharmacol Sin 2008;29:12334. [99] Qi Y, Zhang M, Li H, Frank JA, Dai L, et al. Autophagy inhibition by sustained overproduction of IL6 contributes to arsenic carcinogenesis. Cancer Res 2014;74:374052. [100] Chen L, Liu L, Luo Y, Huang S. MAPK and mTOR pathways are involved in cadmium-induced neuronal apoptosis. J Neurochem 2008;105:25161. [101] Järup L. Hazards of heavy metal contamination. Br Med Bull 2003;68:16782. [102] Lopez E, Figueroa S, Oset-Gasque M, Gonzalez MP. Apoptosis and necrosis: two distinct events induced by cadmium in cortical neurons in culture. Br J Pharmacol 2003;138:90111. [103] Kim S, Moon C, Eun S, Ryu P, Jo S. Identification of ASK1, MKK4, JNK, c-Jun, and caspase-3 as a signaling cascade involved in cadmium-induced neuronal cell apoptosis. Biochem Biophys Res Commun 2005;328:32634. [104] Johri N, Jacquillet G, Unwin R. Heavy metal poisoning: the effects of cadmium on the kidney. Biometals 2010;23:78392.

79

[105] Koyu A, Gokcimen A, Ozguner F, Bayram DS, Kocak A. Evaluation of the effects of cadmium on rat liver. Mol Cell Biochem 2006;284:815. [106] Jiang G, Xu L, Song S, Zhu C, Wu Q, et al. Effects of long-term low-dose cadmium exposure on genomic DNA methylation in human embryo lung fibroblast cells. Toxicology 2008;244:4955. [107] Thompson J, Bannigan J. Cadmium: toxic effects on the reproductive system and the embryo. Reprod Toxicol 2008;25:30415. [108] Akesson A, Bjellerup P, Lundh T, Lidfeldt J, Nerbrand C, et al. Cadmium-induced effects on bone in a population-based study of women. Environ Health Perspect 2006;114:8304. [109] Kocak M, Akcil E. The effects of chronic cadmium toxicity on the hemostatic system. Pathophysiol Haemost Thromb 2006;35:41116. [110] Pihl R, Parkes M. Hair element content in learning disabled children. Science 1977;198:2046. [111] Wright RO, Amarasiriwardena C, Woolf AD, Jim R, Bellinger DC. Neuropsychological correlates of hair arsenic, manganese and cadmium levels in school-age children residing near a hazardous waste site. Neurotoxicology 2006;27:21016. [112] Ja¨rup L, Persson B, Edling C, Elinder CG. Renal function impairment in workers previously exposed to cadmium. Nephrone 1993;64:7581. [113] Okuda B, Iwamoto Y, Tachibana H, Sugita M. Parkinsonism after acute cadmium poisoning. Clin Neurol Neurosurg 1997;99:2635. [114] Templeton DM, Liu Y. Multiple roles of cadmium in cell death and survival. Chem Biol Interact 2010;188:26775. [115] Bagchi D, Joshi SS, Bagchi M, Balmoori J, Benner EJ, et al. Cadmium- and chromium-induced oxidative stress, DNA damage, and apoptotic cell death in cultured human chronic myelogenous leukemic K562 cells, promyelocytic leukemic HL-60 cells, and normal human peripheral blood mononuclear cells. J Biochem Mol Toxicol 2000;14:3341. [116] Szuster-Ciesielska A, Stachura A, Slotwinska M, Kaminska T, Sniezko R, et al. The inhibitory effect of zinc on cadmiuminduced cell apoptosis and reactive oxygen species (ROS) production in cell cultures. Toxicology 2000;145:15971. [117] Son YO, Jang YS, Heo JS, Chung WT, Choi KC, et al. Apoptosis-inducing factor plays a critical role in caspaseindependent, pyknotic cell death in hydrogen peroxideexposed cells. Apoptosis 2009;14:796808. [118] Shaikh ZA, Vu TT, Zaman K. Oxidative stress as a mechanism of chronic cadmium-induced hepatotoxicity and renal toxicity and protection by antioxidants. Toxicol Appl Pharmacol 1999;154:25663. [119] Wang Y, Fang J, Leonard SS, Rao KM. Cadmium inhibits the electron transfer chain and induces reactive oxygen species. Free Radic Biol Med 2004;36:143443. [120] Wang SH, Shih YL, Kuo TC, Ko WC, Shih CM. Cadmium toxicity toward autophagy through ROS-activated GSK-3beta in mesangial cells. Toxicol Sci 2009;108:12431. [121] Chiarelli R, Agnello M, Roccheri MC. Seaurchin embryos as a model system for studying autophagy induced by cadmium stress. Autophagy 2011;7:102834. [122] Son YO, Wang X, Hitron JA, Zhang Z, Cheng S, et al. Cadmium induces autophagy through ROS-dependent activation of the LKB1-AMPK signaling in skin epidermal cells. Toxicol Appl Pharmacol 2011;255:28796. [123] Hardie DG, Scott JW, Pan DA, Hudson ER. Management of cellular energy by the AMP-activated protein kinase system. FEBS Lett 2003;546:11320. [124] Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, et al. The tumor suppressor LKB1 kinase directly activates

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

80

[125] [126] [127] [128]

[129]

[130]

[131]

[132]

[133]

[134]

[135]

[136] [137]

[138]

[139]

[140]

[141]

[142]

[143]

5. THE ROLE OF mTOR, AUTOPHAGY, APOPTOSIS, AND OXIDATIVE STRESS DURING TOXIC METAL INJURY

AMP-activated kinase and regulates apoptosis in response to energy stress. Proc Natl Acad Sci USA 2004;101:332935. Ruggero D, Pandolfi PP. Does the ribosome translate cancer? Nat Rev Cancer 2003;3:17992. Sabatini DM. mTOR and cancer: insights into a complex relationship. Nat Rev Cancer 2006;6:72934. Fujimaki H, Ishido M, Nohara K. Induction of apoptosis in mouse thymocytes by cadmium. Toxicol Lett 2000;115:99105. Watjen W, Haase H, Biagioli M, Beyersmann D. Induction of apoptosis in mammalian cells by cadmium and zinc. Environ Health Perspect 2002;110:8657. Hossain S, Liu HN, Nguyen M, Shore G, Almazan G. Cadmium exposure induces mitochondria-dependent apoptosis in oligoden-drocytes. Neurotoxicology 2009;30:54454. Wang SH, Shih YL, Ko WC, Wei YH, Shih CM. Cadmiuminduced autophagy and apoptosis are mediated by a calcium signaling pathway. Cell Mol Life Sci 2008;65:364052. Mao WP, Ye JL, Guan ZB, Zhao JM, Zhang C, et al. Cadmium induces apoptosis in human embryonic kidney (HEK) 293 cells by caspase-dependent and—independent pathways acting on mitochondria. Toxicol In Vitro 2007;21:34354. Shih CM, Wu JS, Ko WC, Wang LF, Wei YH, et al. Mitochondria-mediated caspase-independent apoptosis induced by cadmium in normal human lung cells. J Cell Biochem 2003;89:33547. Son YO, Lee JC, Hitron JA, Pan J, Zhang Z, et al. Cadmium induces intracellular Ca21- and H2O2-dependent apoptosis through JNK- and p53-mediated pathways in skin epidermal cell line. Toxicol Sci 2010;113:12737. Yu ZP, Matsuoka M, Wispriyono B, Iryo Y, Igisu H. Activation of mitogen-activated protein kinases by tributyltin in CCRFCEM cells: role of intracellular Ca(2þ). Toxicol Appl Pharmacol 2000;168:2007. Chen S, Gu C, Xu C, Zhang J, Xu Y, et al. Celastrol prevents cadmium-induced cell death via targeting JNK and PTENAkt/mTOR network. J Neurochem 2014;128:25666. Wada T, Penninger JM. Mitogen-activated protein kinases in apoptosis regulation. Oncogene 2004;23:283849. Liu Q, Hofmann PA. Protein phosphatase 2A-mediated crosstalk between p38 MAPK and ERK in apoptosis of cardiac myocytes. Am J Physiol Heart Circ Physiol 2004;286: H220412. Yeh JH, Huang CC, Yeh MY, Wang JS, Lee JK, et al. Cadmium-induced cytosolic Ca 21 elevation and subsequent apoptosis in renal tubular cells. Basic Clin Pharmacol Toxicol 2009;104:34551. Khreiss T, Jozsef L, Hossain S, Chan JSD, Potempa LA, et al. Loss of pentameric pymmetry of C-reactive Protein is associated with delayed apoptosis of human neutrophils. J Biol Chem 2002;277:4077581. Sarkar D, Su ZZ, Lebedeva IV, Sauane M, Gopalkrishnan RV, Valerie K, et al. mda- 7 (IL-24) mediates selective apoptosis in human melanoma cells by inducing the coordinated over expression of the GADD family of genes by means of p38 MAPK. Proc Natl Acad Sci USA 2002;99:100549. Kosnett MJ, Wedeen RP, Rothenberg SJ, Hipkins KL, Materna BL, et al. Recommendations for medical management of adult lead exposure. Environ Health Perspect 2007;115:46371. Zhang J, Cai T, Zhao F, Yao T, Chen Y, et al. The role of α-synuclein and tau hyperphosphorylation-mediated autophagy and apoptosis in lead-induced learning and memory injury. Int J Biol Sci 2012;8:93544. Mudipalli A. Lead hepatotoxicity & potential health effects. Indian J Med Res 2007;126:51827.

[144] Philip AT, Gerson B. Lead poisoning—Part I. Incidence, etiology, and toxicokinetics. Clin Lab Med 1994;14:42344. [145] Patrick L. Lead toxicity, a review of the literature. Part 1: exposure, evaluation and treatment. Altern Med Rev 2006;11: 222. [146] Yedjou CG, Milner JN, Howard CB, Tchounwou PB. Basic apoptotic mechanisms of lead toxicity in human leukemia (Hl-60) cells. Int J Environ Res Public Health 2010;7: 200817. [147] Bressler J, Kim KA, Chakraborti T, Goldstein G. Molecular mechanisms of lead neurotoxicity. Neurochem Res 1999;24:595600. [148] Sui L, Zhang P, Yun KL, Zhang HC, Liu L, et al. Lead toxicity induces autophagy to protect against cell death through mTORC1 pathway in cardiofibroblasts. Biosci Rep 2015. Available from: http://dx.doi.org/10.1042/BSR20140164. [149] Maccioni RB, Cambiazo V. Role of microtubule-associated proteins in the control of microtubule assembly. Physiol Rev 1995;75:83564. [150] Gupta A, Dawson VL, Dawson TM. What causes cell death in Parkinson’s disease? Ann Neurol 2008;2:315. [151] Garza A, Vega R, Soto E. Cellular mechanisms of lead neurotoxicity. Med Sci Monit 2006;12:5765. [152] Braakman I, Bulleid NJ. Protein folding and modification in the mammalian endoplasmic reticulum. Annu Rev Biochem 2011;80:7199. [153] Wirawan E, VandeWalle L, Kersse K, Cornelis S, Claerhout S, et al. Caspase-mediated cleavage of Beclin-1 inactivates Beclin1-induced autophagy and enhances apoptosis by promoting the release of proapoptotic factors from mitochondria. Cell Death Dis 2010. Available from: http://dx.doi.org/10.1038/ cddis.2009.16. [154] Kang R, Zeh HJ, Lotze MT, Tang D. The Beclin 1 network regulates autophagy and apoptosis. Cell Death Differ 2011;18:57180. [155] Broussard LA, Hammett-Stabler CA, Winecker RE, RoperoMiller JD. The toxicology of mercury. Lab Med 2002;33:61425. [156] Bose S, Ghosh P, Ghosh S, Chaudhury S, Bhattacharya S. Time dependent distribution [203Hg] mercuric nitrate in the subcellular fraction of rat and fish liver. Biomed Environ Sci 1993;6:195206. [157] Ghosh N, Bhattacharya S. Acute phase response of rabbit to HgCl2 and CdCl2. Biomed Environ Sci 1993;6:17. [158] Vinaya Kumar S, Maitra S, Bhattacharya S. In vitro binding of inorganic mercury to the plasma membrane of rat platelet affects Na1-K1-ATPase activity and platelet aggregation. BioMetals 2002;15:517. [159] Cantoni O, Christie NT, Robison SH, Costa M. Characterization of DNA lesions produced by HgCl2 in cell culture systems. Chem Biol Interact 1984;49:20924. [160] Zasukhina GD, Vasilyeva IM, Sdirkova NI, Krasovsky GN, Vasyukovich LY, et al. Mutagenic effect of thallium and mercury salts on rodent cells with different repair activities. Mutat Res 1983;124:16373. [161] Patnaik BB, Ray A, Agarwal S, Bhattacharya S. Induction of oxidative stress by non-lethal dose of mercury in rat liver: possible relationships between apoptosis and necrosis. J Environ Biol 2010;31:41316. [162] Ben-Ozer E, Rosenspire A, McCabe M, Worth R, Kindzelskii A, Warra N, et al. Mercuric chloride damages cellular DNA by non-apoptotic mechanism. Mutat Res 2000;470:1927. [163] Chatterjee S, Banerjee PP, Chattopadhyay A, Bhattacharya S. Low concentration of HgCl2 drives rat hepatocytes to

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL

REFERENCES

autophagy/apoptosis/necroptosis in a time-dependent manner. Toxicol Environ Chem 2013;95:1192207. [164] Chatterjee S, Nandi P, Chattopadhyay A, Bhattacharya S. Regulation of autophagy in rat hepatocytes treated in vitro with low concentration of mercury. Toxicol Environ Chem 2013;95:50414. [165] Chatterjee S, Munshi C, Chattopadhyay A, Bhattacharya S. Mercuric chloride effects on adult rat oval cells-induced apoptosis. Toxicol Environ Chem 2014;95:172238. [166] Maiese K, Chong ZZ, Wang S, Shang YC. Oxidant stress and signal transduction in the nervous system with the PI 3-K, Akt, and mTOR cascade. Int J Mol Sci 2012;13:1383066.

81

[167] Qin AP, Liu CF, Qin YY, Hong LZ, Xu M, Yang L, et al. Autophagy was activated in injured astrocytes and mildly decreased cell survival following glucose and oxygen deprivation and focal cerebral ischemia. Autophagy 2010;6:73853. [168] Chong ZZ, Maiese K. Mammalian target of rapamycin signaling in diabetic cardiovascular disease. Cardiovasc Diabetol 2012;11. Available from: http://dx.doi.org/10.1186/1475-284011-45. [169] Wang Y, Hua Z, Liua Z, Chena R, Penga H, Guoa J, et al. MTOR inhibition attenuates DNA damage and apoptosis through autophagy-mediated suppression of CREB1. Autophagy 2013;12:206986.

I. mTOR IN CELLULAR DEVELOPMENT, PROLIFERATION, AND SURVIVAL