Next Article in Journal
Characteristics of Antisense Transcript Promoters and the Regulation of Their Activity
Next Article in Special Issue
Induction of ROS Overload by Alantolactone Prompts Oxidative DNA Damage and Apoptosis in Colorectal Cancer Cells
Previous Article in Journal
Cell Density-Dependent Upregulation of PDCD4 in Keratinocytes and Its Implications for Epidermal Homeostasis and Repair
Previous Article in Special Issue
FLIP the Switch: Regulation of Apoptosis and Necroptosis by cFLIP
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Research Advances on Pathways of Nickel-Induced Apoptosis

1
Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University, Ya’an 625014, China
2
College of Veterinary Medicine, Sichuan Agricultural University Ya’an, Ya’an 625014, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2016, 17(1), 10; https://doi.org/10.3390/ijms17010010
Submission received: 22 September 2015 / Revised: 15 December 2015 / Accepted: 16 December 2015 / Published: 23 December 2015
(This article belongs to the Collection Programmed Cell Death and Apoptosis)

Abstract

:
High concentrations of nickel (Ni) are harmful to humans and animals. Ni targets a number of organs and produces multiple toxic effects. Apoptosis is important in Ni-induced toxicity of the kidneys, liver, nerves, and immune system. Apoptotic pathways mediated by reactive oxygen species (ROS), mitochondria, endoplasmic reticulum (ER), Fas, and c-Myc participate in Ni-induced cell apoptosis. However, the exact mechanism of apoptosis caused by Ni is still unclear. Understanding the mechanism of Ni-induced apoptosis may help in designing measures to prevent Ni toxicity.

Graphical Abstract

1. Chemical Properties and Toxicity of Nickel

1.1. Chemical Properties of Ni

Nickel (Ni) is the 24th most abundant element in the earth’s crust [1,2]. It is the fifth heaviest element and is a part of group VIII B of the periodic table. In nature, Ni is found in combination with arsenic, antimony, and sulfur [3]. Elemental Ni is a silver-white solid metal, with high thermal conductivity, electrical conductivity, and melting point. Ni metal is highly stable [4] and is used for electroplating and protective coating. Ordinary Ni has an oxidation state of +2; higher oxidation states (such as Ni+3 and Ni+4) occur rarely in certain oxide systems [5]. Ni compounds are classified as soluble and insoluble [5,6]. Strong acid and organic acid Ni salts are soluble, whereas weak inorganic acid Ni salts are insoluble. Due to its chemical properties, gloss, and low price, Ni is used in jewelry, alloys, stainless steel, food processing industries, catalysts, and pigments [7,8,9]. Ni chloride, nitrate, sulphate, hydroxide, acetate, carbonate, and oxide are the most commercially important Ni compounds [10].

1.2. Biological Properties of Ni

Ni is essential for many microorganisms, plants, and animals [9,11,12]. Some studies have shown that very low levels of Ni are also essential for humans [13]. In rats, Ni deficiency reduces iron content in organs, haemoglobin, and hematocrit [14]. Ni has several biological functions, including activation of calcineurin; action and formation of cGMP [15]; transmission of genetic code (DNA, RNA) [16]; acting as a cofactor of albumin, proteins, and amino acids; transport of oxygen; stimulation of metabolism through interaction with iron in hemoglobin [17,18]; and formation of urease, carbon monoxide dehydrogenase, and methyl-S-coenzyme M reductase [15].

1.3. Ni Toxicity

The widespread use of Ni increases its concentration in biogeochemical cycles and increases human exposure by environmental contamination and occupational exposure [8]. Exposure to Ni commonly occurs by ingestion of contaminated water and food [9,19]. Workers in Ni-producing and processing industries are exposed by inhalation and, to a lesser extent, dermal contact [20]. People may also be exposed through contact with stainless steel, jewelry, and coins. Ni is toxic at high doses to both humans and animals [21]. Exposure to Ni can cause allergy, contact dermatitis, and toxicity of organ systems [22]. Li et al. [23] reported that NiCl2 increases the secretion of a pro-inflammatory cytokine, interleukin-1β (IL-1β), in bone marrow-derived macrophages and bone marrow dendritic cells. Ni may cause neurotoxicity, hepatotoxicity, nephrotoxicity, gene toxicity, reproductive toxicity, and increased risk of cancer [20,24,25,26,27,28,29,30,31,32]. Bones, kidneys, lungs, liver, and heart are the main organs of Ni accumulation [32,33,34].
Humans, especially Ni metallurgy workers, are exposed to Ni by inhalation and ingestion, with plants being the primary source of Ni for humans [34,35,36]. Ni may damage multiple organs and cause lung and nasal cancer [19,22,37]. Immediate and delayed hypersensitivity and allergic skin reactions are common adverse effects of Ni. Ni is both an allergen and a potential immunomodulatory and immunotoxic agent [38]. With the exception of metallic Ni, all Ni compounds have been classified as human carcinogens by the International Agency for Research on Cancer, based on studies on Ni workers and laboratory animals [39].
Numerous in vitro, in vivo, and epidemiological studies have documented the carcinogenic quality of Ni [40,41,42,43,44,45]. The LD50 of oral nickel acetate in rats and mice are 350 and 420 mg/kg, respectively [39]. Dietary NiCl2 at ≥300 mg/kg can cause reduced growth rate, and at ≥1100 mg/kg it can cause anemia or death in chickens [12]. Growth inhibition occurs at ≥700 mg/kg nickel sulfate (NiSO4) and nickel acetate in chicks [46]. Chicks fed a diet containing ≥250–300 mg/kg Ni can show inhibition of growth and decreased feed intake [47]. Oral NiCl2 decreases body and liver weight in mice [25]. Amudha et al. [48] have suggested that intraperitoneal NiCl2 causes significant kidney damage and reduced activities of enzymatic and non-enzymatic antioxidants in rats [48]. Dietary NiCl2 at ≥300 mg/kg damages the intestines and kidney, and decreases the immune function of the spleen, thymus, and bursa of Fabricius in chickens [49,50,51,52,53,54,55,56,57].

2. Biological Characteristics of Cell Apoptosis

Apoptosis is required for homeostasis of the cell population and defense during injury [58]. Failure to undergo apoptosis can cause several diseases such as cancer and autoimmune diseases, whereas excessive cell death is responsible for several neurodegenerative diseases [59]. So far, the main focus of research on the mechanism of apoptosis has been on the extrinsic and intrinsic apoptosis pathways (Figure 1) [60,61,62,63,64].

2.1. Bcl-2 Family Protein in Apoptosis

B-cell lymphoma-2 (Bcl-2) family proteins are involved in apoptosis [65], partly through the control and modulation of outer mitochondrial membrane integrity [66,67]. Based on their functions, Bcl-2 family proteins are classified into anti- and pro-apoptotic proteins (Figure 2). A loss of balance between anti- and pro-apoptotic proteins may cause either inhibition or promotion of apoptosis. Bcl-2 family members have single or multiple homology domains, such as Bcl-2 homology (BH1, -2, -3, and -4) that are important in the heterodimeric interaction among Bcl-2 family proteins [68]. Anti-apoptotic Bcl-2 family multi-domain proteins contain BH-(1-4) domains, such as Bcl-2, Bcl-2 homolog of ovary, Bcl-extra long (Bcl-xL), Bcl-w, and A1. Myeloid cell leukemia factor-1 (Mcl-1) is the only anti-apoptotic Bcl-2 protein with three BH domains: BH-1, -2, and -3 [69]. Based on the number of BH domains, pro-apoptotic Bcl-2 family proteins are classified into two subgroups. Bcl-2 antagonistic killer (Bak), Bcl-2 associated X protein (Bax), Bcl-2 related ovarian killer, and Bcl-extra short have multiple BH domains [68]. There are eight BH3 domain-only members: BH3 interacting domain death agonist (Bid), Bcl-2 interacting mediator of cell death (Bim), hara-kiri, p53-upregulated modulator of apoptosis (Puma), Bcl-2 modifying factor (Bmf), Bcl-2 antagonist of cell death (Bad), Noxa (named for “damage”), and Bcl-2 interacting killer [Bik, also known as natural born killer (Nbk)] [66,70]. In general, BH3 domain-only proteins suppress Bcl-2 anti-apoptotic proteins and induce apoptosis after lethal stress [71]. Bcl-2 maintains Bax and Bak in an inactive state. Furthermore, Bcl-2 titration away from Bax and Bak by BH3-only Bcl-2 family members allows them to oligomerize and form a channel through which cytochrome c (cyt c) can translocate to cytoplasm. Cyt c combines in a very specific stoichiometric fashion with apaf1 to form the apoptosome and activate caspase-9 [68,72].
Figure 1. Summary of the extrinsic and intrinsic pathway in apoptosis. Extrinsic apoptosis: The combination of a ligand (FasL/TNFR1) and a death receptor (Fas/TNF) starts the extrinsic apoptosis pathway. This binding can attract the combination of FADD, TRADD and procaspase 8. Activated caspase 8 then activates downstream effector caspases or truncates the Bid. tBid can disrupt the mitochondria, and then induces mitochondria-mediated apoptosis. Intrinsic apoptosis: Mitochondria-mediated apoptosis pathway: The MMP disruption results in the release of cyt c, Smac, HtrA2, AIF and Endo G from mitochondrial intermembrane space to cytoplasm. Cytoplasmic cyt c promotes the aggregation of procaspase 9 and apaf1, and then activates caspase-9. Activated caspase-9 cleaves and activates caspase-3, 6, 7, which then induces apoptosis. Concurrently, Smac and HtrA2 inhibit XIAP expression for also contributing to the apoptosis. AIF and Endo G activate the caspase-independent mitochondria-mediated pathway. ER-mediate apoptosis pathway: Under prolonged ER stress, protein kinase RNA (PKR)-like ER kinase (PERK) pathway can induce apoptosis. Activated transcription factor 4 (ATF4) is an important transcription factor that accumulates via an un-conventional mechanism following PERK activation. ATF4 accumulation up-regulates the pro-apoptotic transcriptional factor, e.g., C/EBP homologous protein (CHOP), which induces apoptosis. The IRE1α pathway is also the important mechanism of ER stress. IRE1α mediates apoptosis through activation of c-Jun amino terminal kinase (JNK) and mitogen-activated protein kinase (MAPK) pathways. After the ER damage, acute translocation of Ca2+ from ER to mitochondria promotes Ca2+-mediated mitochondrial cell death. In addition, caspases-4 and -12 are also involved in ER stress-induced apoptosis.
Figure 1. Summary of the extrinsic and intrinsic pathway in apoptosis. Extrinsic apoptosis: The combination of a ligand (FasL/TNFR1) and a death receptor (Fas/TNF) starts the extrinsic apoptosis pathway. This binding can attract the combination of FADD, TRADD and procaspase 8. Activated caspase 8 then activates downstream effector caspases or truncates the Bid. tBid can disrupt the mitochondria, and then induces mitochondria-mediated apoptosis. Intrinsic apoptosis: Mitochondria-mediated apoptosis pathway: The MMP disruption results in the release of cyt c, Smac, HtrA2, AIF and Endo G from mitochondrial intermembrane space to cytoplasm. Cytoplasmic cyt c promotes the aggregation of procaspase 9 and apaf1, and then activates caspase-9. Activated caspase-9 cleaves and activates caspase-3, 6, 7, which then induces apoptosis. Concurrently, Smac and HtrA2 inhibit XIAP expression for also contributing to the apoptosis. AIF and Endo G activate the caspase-independent mitochondria-mediated pathway. ER-mediate apoptosis pathway: Under prolonged ER stress, protein kinase RNA (PKR)-like ER kinase (PERK) pathway can induce apoptosis. Activated transcription factor 4 (ATF4) is an important transcription factor that accumulates via an un-conventional mechanism following PERK activation. ATF4 accumulation up-regulates the pro-apoptotic transcriptional factor, e.g., C/EBP homologous protein (CHOP), which induces apoptosis. The IRE1α pathway is also the important mechanism of ER stress. IRE1α mediates apoptosis through activation of c-Jun amino terminal kinase (JNK) and mitogen-activated protein kinase (MAPK) pathways. After the ER damage, acute translocation of Ca2+ from ER to mitochondria promotes Ca2+-mediated mitochondrial cell death. In addition, caspases-4 and -12 are also involved in ER stress-induced apoptosis.
Ijms 17 00010 g001
Figure 2. Bcl-2 Proteins. The Bcl-2 proteins can be subdivided into pro-survival and pro-apoptotic members, and have single or mutiple conserved functional Bcl-2 homology domains.
Figure 2. Bcl-2 Proteins. The Bcl-2 proteins can be subdivided into pro-survival and pro-apoptotic members, and have single or mutiple conserved functional Bcl-2 homology domains.
Ijms 17 00010 g002
Some studies have shown that p53 can regulate Bcl-2 proteins, but the specific mechanism is not clear [73]. The phosphoinositide-3-kinase/serine-threonine kinase (PI3K/Akt) pathway inhibits apoptosis via the modulation of Bcl-2 proteins [74,75,76]. This pathway increases anti-apoptotic proteins (Bcl-2 and Bcl-xL) and decreases pro-apoptotic proteins (Bad and Bax) [77,78,79,80].

2.2. Caspases in Apoptosis

Caspases are cysteine proteases that are extremely important for intracellular apoptotic pathways [81,82,83]. After various intracellular and extracellular stimuli have occurred, caspases can be activated to execute apoptosis [84]. Pro-caspases are omnipresent in the cell, so apoptosis can occur rapidly without the need of transcription and translation. The removal of sick, damaged, and senescent cells provides a distinct survival advantage [85]. Caspases are divided into initiators and executioners, based on their physiological functions (Figure 3) [72,86,87,88,89].
Figure 3. Caspase classification.
Figure 3. Caspase classification.
Ijms 17 00010 g003
Caspases-2, -8, -9 and -10 are initiators, as they are closely coupled to pro-apoptotic signals [90]. The initiator caspases exist in normal cells as inactive pro-caspase monomers and are activated by dimerization rather than cleavage. [91,92,93]. Dimerization promotes autocatalytic cleavage of caspase monomers into a large and a small subunit that results in dimer stabilization [94].
Caspases-3, -6, and -7 are executioners and exist as inactive pro-caspase dimers [95]. The initiator caspases activate executioner caspases by cleavage [94,96], which causes a conformational change that brings the two active sites of the executioner caspase dimer together and creates a functional mature protease [97]. Caspases-3 and -7 have identical cleavage sites. On being activated, an accelerated feedback loop of caspase activation occurs, ultimately causing cell death [97].
Murine caspase-12 and human caspase-4 belong to the interleukin-1-converting enzyme subfamily. Murine caspase-12 and human caspase-4 have 48% homology at the amino acid level and have structures similar to initiator caspases [98,99]. Caspase-12 and -4 are involved in endoplasmic reticulum (ER) stress-induced apoptosis [98,99,100].

2.3. Extrinsic Pathway in Apoptosis

The extrinsic apoptosis pathway is activated by the binding of extracellular death ligands to cell-surface death receptors [101]. The tumor necrosis factor (TNF) receptor gene super family is a member of the death receptors, which have a death domain (DD) that is important for apoptotic signal transduction [102,103]. FasL/FasR, TNF-α/TNFR1, Apo3L/DR3, Apo2L/DR4, and Apo2L/DR5 are the notable ligands and their corresponding death receptors [104,105,106].
FasL/FasR and TNF-α/TNFR1 are the characteristic models in the extrinsic apoptotic pathway. The extrinsic pathway is initiated by the binding of an extracellular death ligand to its cell surface transmembrane death receptor, causing oligomerization of the receptor [107]. This binding recruits the intracellular domain of the receptor (e.g., Fas associated death domain (FADD), TNF receptor associated death domain (TRADD)) and initiator caspases (e.g., procaspase-8 or -10) [108]. These complexes are regarded as death-inducing signaling complexes (DISC) [102,109,110,111]. The formation of these complexes promotes the oligomerization of procaspase-8 and -10 and auto-activation through self-cleavage [112].
Activated caspase-8 cleaves and activates downstream effector caspases such as caspase-1, -3, -6, and -7 [113]. As an important executioner of apoptosis, caspase-3 can activate many proteins, including the nuclear enzyme poly ADP-ribose polymerase (PARP) through proteolytic cleavage [114]. These downstream cleavage events are the morphological characteristics of apoptosis.

2.4. Intrinsic Pathway in Apoptosis

The intrinsic signaling pathways are known as mitochondria- and ER-initiated apoptosis [115,116].

2.4.1. Mitochondria in the Intrinsic Pathway

The apoptotic stimuli cause changes in the Bcl-2 proteins. Bcl-2 titration away from Bax and Bak by BH3-only Bcl-2 family members allows them to oligomerize. This step opens the mitochondrial permeability transition pore (MPT) and disrupts the mitochondrial membrane potential (MMP), which translocates some pro-apoptotic proteins from the intermembrane space to the cytosol [117]. These pro-apoptotic proteins include cyt c, second mitochondrial activator of caspases/direct inhibitors of apoptosis binding protein with low pI (Smac/Diablo), the serine protease high-temperature-requirement protein A2/Omi (HtrA2/Omi), apoptosis inducing factor (AIF), and endonuclease G (Endo G) [61,118,119,120,121].
Apoptosis induced by cyt c, Smac/Diablo, and HtrA2/Omi requires the participation of caspases. Cyt c binds and activates apoptotic peptidase activating factor 1 (Apaf-1) and procaspase-9 to form an apoptosome [61]. The clustering of procaspase-9 in this apoptosome leads to caspase-9 activation. Caspase-9 activates downstream caspases, including caspase-3, -6, and -7, causing apoptosis. Inhibitors of apoptosis proteins (IAP) suppress apoptosis by directly inhibiting caspases [122,123]. Smac/DIABLO and HtrA2/Omi can promote apoptosis by inhibiting IAP proteins activity [124].
AIF and Endo G activate the caspase-independent mitochondrial pathway. After disruption of the mitochondrial membrane potential, AIF and Endo G translocate to the nucleus and cause DNA fragmentation and apoptosis [125,126,127].

2.4.2. ER in the Intrinsic Pathway

Recent studies have shown that the ER also plays an important role in the intrinsic pathway [128,129]. The main function of ER is synthesis, folding, and translocation of proteins [130]. ER stress is activated by the disruption of ER function, which leads to the accumulation of misfolded and unfolded proteins in the ER lumen. ER stress induces an adaptive signal, Unfolded Protein Response (UPR), which restores ER homeostasis and protects the cell [116]. In the absence of this response, apoptosis occurs [116].
Under ER stress, protein kinase RNA (PKR)-like ER kinase (PERK) activates and phosphorylates eukaryotic translation initiation factor 2α (eIF2α), causing a translational arrest. However, under prolonged ER stress, it can promote apoptosis. Activated transcription factor 4 (ATF4) is an important transcription factor that accumulates via an unconventional mechanism following activation of the protein PERK [131]. Prolonged accumulation of ATF4 leads to apoptosis by a variety of mechanisms. ATF4 can upregulate the pro-apoptotic transcriptional factors, e.g., CHOP (C/EBP homologous protein, also called the growth arrest and DNA-damage-inducible 153, GADD153) [132]. CHOP promotes apoptosis through activation of downstream factors such as GADD34, a caspase-activating cell-surface death receptor of the tumor necrosis factor death receptor 5 (DR5), and endoplasmic reticulum oxidoreductase 1α (ERO1α) [133]. GADD34 inhibits the phosphorylation of elF2α [131]. This inhibition causes the accumulation of unfolded proteins in the ER lumen and translation of pro-apoptotic proteins. CHOP suppresses Bcl-2 transcription [134] and increases Bim expression [135], thereby promoting apoptosis. ERO1α activates the inositol triphosphate receptor (IP3R), which stimulates the translocation of excessive Ca2+ to the mitochondria and triggers the mitochondria-mediated apoptotic pathway [136]. ATF4 also regulates Noxa (a BH3-only protein) [137,138]. Noxa activates the effector proteins Bax and/or Bak [139] and induces mitochondria-mediated apoptosis.
The inositol-requiring enzyme/endonuclease 1α (IRE1α) pathway also plays an important part in ER stress. The IRE1α pathway has a pro-survival function, but it may cause apoptosis under prolonged ER stress [140]. IRE1α stimulates the activation of TNF receptor-associated factor 2 (TRAF2), which activates the apoptosis signal-regulating kinase 1/c-Jun amino terminal kinase (ASK1/JNK) cascade [141,142]. In addition, ER stress induces apoptosis through p38 mitogen-activated protein kinase (MAPK), which is activated by ASK1. p38 MAPK phosphorylates and activates CHOP and induces apoptosis [143]. Additionally, both p38 MAPK and JNK promote apoptosis through an increase in Bax [144].
The ER is the reservoir of Ca2+ in cells [145]. Ca2+ translocation from the ER to mitochondria triggers mitochondria-mediated apoptosis [146,147]. Excessive Ca2+ can disrupt the balance between Bcl-2 proteins and induce the translocation of cyt c [148].
In addition, some studies have suggested that caspase-12 is a specific marker of ER stress-induced apoptosis [128]. Mouse caspase-12 cleaves and activates caapase-9 and -3 and induces apoptosis [98,149]. However, human caspase-12 has no parallel function due to a frameshift disruption of its gene, resulting in a premature stop codon. In addition, human caspase-12 also contains amino acid substitutions in the caspase activity area [150]. Instead, human caspase-4 is specifically cleaved under ER stress, suggesting that it may be a functional ortholog of mouse caspase-12 in ER stress-induced apoptosis [99,151]. So far, little is known about how caspase-4 is activated in ER stress-triggered apoptosis.

3. Apoptosis Induced by Ni

Studies have demonstrated that Ni can increase apoptosis. Su et al. and Liu et al. [152,153] reported that NiSO4 induces DNA damage, apoptosis and oxidative damage in the liver and testes of mouse. Also, NiSO4 induces JNK-mediated oxidative damage and apoptosis in the liver of Carassius auratus [154]. NiCl2 has been proved to induce apoptosis in the liver of Kunming mice [153]. Our previous studies indicated that NiCl2 ≥ 300 mg/kg (dietary) can increase apoptosis percentages thymus, spleen, and cecal tonsil of broiler chickens [49,155,156].
In vitro research of Ni-induced apoptosis, Ni compounds can promote apoptosis in B cells [157], human T hybridoma cells [158], human hepatoma cells [159], keratinocytes [160], human airway epithelial (HEp-2) and human breast cancer (MCF-7) cells [161], human liver cells (HepG2) [162], and normal rat kidney cells [27], as well as human neutrophils and lymphocytes [163,164]. Shiao et al. [165] have reported that DNA fragmentation is detected in Chinese hamster ovary (CHO) cells treated with ≥160 μM Ni(II) and its intensity increases with increasing Ni(II) concentration.

4. Pathways or Mechanisms of Ni-Induced Cell Apoptosis

4.1. ROS-Mediated Apoptosis

Reactive oxygen species (ROS) acts as a crucial factor in the early stages of apoptosis. Mitochondria are both the source and target of ROS. When excess ROS is generated, it induces MMP depolarization and cyt c release, which triggers caspase activation [166,167,168,169]. The ROS generation can also induce DNA damage, which then promotes apoptosis [170].
Ni can induce oxidative stress in vitro and in vivo [171,172]. Ma et al. [173] demonstrated that Ni NWs induce apoptosis through ROS generation, and that ROS induces apoptosis through mitochondrial damage or activation of cell cycle checkpoints in HeLa cells. Nickel subsulfide (Ni3S2) induces ROS-mediated apoptosis in human bronchial epithelial cells (BEAS-2B) [174]. Ahamed [175] suggests that nickel nanoparticle (NiNPs) induces oxidative damage, which decreases glutathione (GSH), and induces ROS and lipid peroxidation (LPO) in human lung epithelial A549 cells.
Excess generation of ROS will result in oxidative stress that mediates apoptosis. The reduction of antioxidant defense is the main reason for ROS generation. Superoxide dismutase (SOD), catalase (CAT), glutathione reductase (GR), glutathione peroxidase (GSH-Px) and glutathione-S-transferase (GST) and GSH are the important antioxidant molecules. Some studies have reported that NiCl2 can decrease the antioxidative system in the Tigriopus japonicas, goldfish and broiler [48,50,53,155,156,176,177,178]. NiSO4 can decrease the GSH levels and activities of SOD and GSH-Px [154]. An amount of 13 uM Ni decreases CAT and GST activities in M. galloprovincialis (Lam) [179]. After exposure to NiNPs, ROS-induced apoptosis is observed in human skin epidermal cells [180]. However, Tyagi et al. [11] reported that renal SOD activity and GSH content are significantly increased after a high dose of NiCl2 has been used in rat.
Yet, Ni (II) at concentrations of 10–50 μM does not increase the intracellular ROS generation, but can activate the Nrf2 (NF-E2-related factor 2) signaling that is an antioxidant pathway [181]. The GSH depletion is due to the Ni-GSH complexes or Ni-mediated ROS formation [182].

4.2. The Extrinsic Pathway of Ni-Induced Apoptosis

At present, there are few studies on the extrinsic pathway of Ni-induced apoptosis. The results of Zhao et al. [183] suggest that metallic nickel particles can increase Fas, FADD, DR 3, and caspase-8 expression, but not DR6 and TNF-R2. Also, this study detects the formation of DISC. These data indicate that the extrinsic apoptotic pathway is involved in the apoptotic process caused by metallic nickel particles in JB6 cells. Bonin et al. [184] reported that higher expression levels of caspase-8 mRNA are detected in the Ni-exposed workers.

4.3. The Intrinsic Pathway of Ni-Induced Apoptosis

The mitochondria play a dominant role in the intrinsic apoptotic pathway [115,185]. Wang et al. [186] suggested that Nickel acetate induces MMP loss and apoptosis. Ni NWs-induced MMP disruption and apoptosis, and nickel ferrite nanoparticle-induced MMP loss are also found in HeLa cells [173] and in HepG2 and MCF-7 cells [187]. After the MMP loss, there are several apoptotic factor releases from the mitochondria. There is evidence that cyt c and caspases-9, -3 and -6 protein expression is increased after nickel acetate has been added in human proximal tubule cells [186]. Nickel nanoparticles and nickel fine particles can increase cyt c and AIF translocation from mitochondria to the cytoplasm [184]. AIF can induce apoptosis through caspase-independent pathway. However, cyt c can activate the caspase-dependent pathway. It has been demonstrated that fine nickel particles and nickel nanoparticles increase the protein expression and activation of caspase-3, -6, and -9 [183]. Additionally, NiSO4 increases caspase-3 activity in Carassius auratus liver [154]. Patel et al. [188] suggested that NiCl2 increases caspae-3, caspase-7 protein expression in human lung epithelial cells. Caspase-3 activity is obviously up-regulated with the increase in time and Ni NPs dose [175]. It has been also demonstrated that NiO nanoparticles (NiONPs) increases the numbers of apoptotic cells and the activaty of caspase-3 [189]. p53-defective human leukemic cells (U937) exposed to bis (S-citronellalthiosemicarbazonato) nickel(II) ([Ni(tcitr)2]) cause MMP disruption and caspase-3 activation [190]. Also, nickel nanoparticles (NiNPs) increases caspase-3 activity and apoptosis in A431 cells [180]. After exposure to nickel ferrite nanoparticles, the activation and gene expression of caspase-3 and caspase-9 are increased in HepG2 and MFC-7 cancer cells [187]. Dietary NiCl2 can increase caspase-3 expression in the thymus, spleen, and cecal tonsil in broiler chickens [49,155,156]. Abovementioned results show that the intrinsic signal pathway takes part in the apoptotic process induced by Ni and Ni compounds.
The Bcl-2 family of proteins is important in the modulation of the outer mitochondrial membrane integrity. In studies of Ni-induced apoptosis, Ni can alter the expression of Bcl-2 family proteins. Liu et al. [153] reported that NiCl2 can decrease the Bcl-2 protein expression and increase the Bax protein expression through suppression of PI3K/Akt pathway in the liver of Kunming mice. Ahamed et al. [187] suggest that nickel ferrite nanoparticles increase Bax mRNA expression through the p53 pathway, and decrease Bcl-2 mRNA expression. NiSO4 increases Bax protein expression and decreases Bcl-2 protein expression in Carassius auratus liver [154]. Reduction of Bcl-2 and Bcl-xL protein expression and enhancement of Bad, Bcl-Xs, Bax protein expression are observed after human proximal tubule cells and human bronchial epithelial cells have been cultured with nickel acetate and NiONPs [186,189]. Human BEAS-2B cells exposed to Ni3S2 show down-regulation of several antiapoptotic proteins (Bcl-2 and Bcl-xL) [174]. Dietary NiCl2 can increase Bax expression and Bax/Bcl-2 ratio and decrease Bcl-2 expression in the thymus, spleen, and cecal tonsil of broiler chickens [49,155,156]. However, the results of Zhao et al. [183] show that nickel nanoparticles and nickel fine particles can increase Bcl-2 expression and decrease Bax expression.
Up to now, there is only one report that nickel acetate can induce ER stress and increase CHOP protein expression in NRK52E cells and Hepa-1c1c7 cells [191].

4.4. Others

It has been shown that c-Myc amplifies the mitochondrial pathway and promotes and/or amplifies the death receptor pathway [192]. c-Myc can increase FasL expression, provoke combination of tBid and mitochondria, affect DISC components, inhibit Bcl-X expression and disrupt nuclear factor-κB (NF-κB) activation [193]. Besides, c-Myc activation provokes cyt c release [194]. c-Myc can selectively suppress Bcl-2 family proteins [193,195].
NiSO4 increases c-Myc protein and mRNA expression in non-tumorigenic Beas-2B and human keratinocyte HaCaT cells. It has been proved that the c-Myc is important in Ni-induced apoptosis through knockout and restoration technology. ERK/MEK (extracellular regulated protein kinases/mitogen-activated protein kinase) inhibitors (U0126 and PD98059) attenuate c-Myc expression. These results indicate that the ERK-dependent c-Myc pathway takes part in Ni-induced Beas-2B cell apoptosis [196].

5. Conclusions and Future Perspectives

A number of studies have explored the molecular mechanism of Ni and Ni compounds-induced apoptosis. However, the precise mechanisms of Ni-induced apoptosis are inconclusive up to now. As shown in Figure 4, previous studies demonstrated that ROS-, mitochondria-, ER-, Fas- and c-Myc-mediated apoptotis are all involved in Ni-induced cell apoptosis. Most of these studies focus on the ROS- and mitochondria-mediated apoptosis. Only one study presents the ER-, Fas- and c-Myc-mediated apoptosis pathway. Therefore, more research should be conducted to explore whether ER-, Fas- and c-Myc-mediate apoptotic pathways are involved in Ni-induced apoptosis.
Figure 4. Pathways of Ni-induced apoptosis. The ROS-, mitochondria-, ER-, Fas- and c-Myc-mediated apoptotic pathway are all involved in Ni-induced cell apoptosis.
Figure 4. Pathways of Ni-induced apoptosis. The ROS-, mitochondria-, ER-, Fas- and c-Myc-mediated apoptotic pathway are all involved in Ni-induced cell apoptosis.
Ijms 17 00010 g004
In addition to the conventional apoptotic proteins, another molecule has been found to regulate apoptosis. MicroRNAs (miRNAs) negatively regulate gene expression. Recent studies have reported that miRNAs participate in development, differentiation, and cell apoptosis [197,198,199].
Disorders of apoptosis may be important in the Ni or/and Ni compound toxicology, which includes neurotoxicity, hepatotoxicity, renaltoxicity, toxicity of the autoimmune system, and carcinogenicity. Revealing the mechanisms of Ni-induced apoptosis may contribute to the prevention of Ni toxicity.

Acknowledgments

The study was supported by the program for Changjiang scholars and innovative research team in university (IRT 0848) and the Shuangzhi project of Sichuan Agricultural University (03570327; 03571198).

Author Contributions

Hongrui Guo, Lian Chen and Hengmin Cui designed the study, reviewed the literature, wrote the first draft and prepared the schemes; Hongrui Guo, Hengmin Cui, Xi Peng, Jing Fang, Zhicai Zuo, Junliang Deng, Xun Wang and Bangyuan Wu reviewed the paper; Hengmin Cui and Hongrui Guo critically reviewed the paper and revised the study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Greenwood, N.N.; Earnshaw, A.; Earnshaw, A. Chemistry of the Elements; Butterworth-Heinemann: Oxford, UK, 1997. [Google Scholar]
  2. Doll, R. Nickel exposure: A human health hazard. IARC Sci. Publ. 1984, 53, 3–21. [Google Scholar] [PubMed]
  3. Gurley, L.R.; Tobey, R.A.; Valdez, J.G.; Halleck, M.S.; Barham, S.S. Biological availability of nickel arsenides: Toxic effects of particulate Ni5As2. Sci. Total Environ. 1983, 28, 415–432. [Google Scholar] [CrossRef]
  4. Rando, D.; Steglitz, U.; Morsdorf, G.; Kaltwasser, H. Nickel availability and urease expression in Proteus mirabilis. Arch. Microbiol. 1990, 154, 428–432. [Google Scholar] [CrossRef] [PubMed]
  5. Schaumlöffel, D. Nickel species: Analysis and toxic effects. J. Trace Elem. Med. Biol. 2012, 26, 1–6. [Google Scholar] [CrossRef] [PubMed]
  6. Munoz, A.; Costa, M. Elucidating the mechanisms of nickel compound uptake: A review of particulate and nano-nickel endocytosis and toxicity. Toxicol. Appl. Pharmacol. 2012, 260, 1–16. [Google Scholar] [CrossRef] [PubMed]
  7. Holbrook, J.; Minocha, J.; Laumann, A. Body piercing: Complications and prevention of health risks. Am. J. Clin. Dermatol. 2012, 13, 1–17. [Google Scholar] [CrossRef] [PubMed]
  8. Henderson, R.G.; Durando, J.; Oller, A.R.; Merkel, D.J.; Marone, P.A.; Bates, H.K. Acute oral toxicity of nickel compounds. Regul. Toxicol. Pharm. 2012, 62, 425–432. [Google Scholar] [CrossRef] [PubMed]
  9. Cempel, M.; Nikel, G. Nickel: A review of its sources and environmental toxicology. Pol. J. Environ. Stud. 2006, 15, 375–382. [Google Scholar]
  10. Grandjean, P. Human exposure to nickel. IARC Sci. Publ. 1984, 469–485. [Google Scholar]
  11. Tyagi, R.; Rana, P.; Gupta, M.; Khan, A.R.; Bhatnagar, D.; Bhalla, P.J.; Chaturvedi, S.; Tripathi, R.P.; Khushu, S. Differential biochemical response of rat kidney towards low and high doses of NiCl2 as revealed by NMR spectroscopy. J. Appl. Toxicol. 2013, 33, 134–141. [Google Scholar] [CrossRef] [PubMed]
  12. Ling, J.; Leach, R. Studies on nickel metabolism: Interaction with other mineral elements. Poultry Sci. 1979, 58, 591–596. [Google Scholar] [CrossRef]
  13. Wen, Z.T.; Burne, R.A. Functional genomics approach to identifying genes required for biofilm development by Streptococcus mutans. Appl. Environ. Microbiol. 2002, 68, 1196–1203. [Google Scholar] [CrossRef] [PubMed]
  14. Kirchgessner, M.; Perth, J.; Schnegg, A. Deficient nickel supply and the content of calcium, magnesium and phosphorus inthe bone of growing rats. Arch. Tierernahr. 1980, 30, 805–810. [Google Scholar] [CrossRef] [PubMed]
  15. Poonkothai, M.V.B.S. Nickel as an essential element and a toxicant. Int. J. Environ. Sci. 2012, 1, 285–288. [Google Scholar]
  16. Coogan, T.P.; Latta, D.M.; Imbra, R.J.; Costa, M. Effect of nickel(II) on DNA-protein interactions. Biol. Trace Elem. Res. 1989, 21, 13–21. [Google Scholar] [CrossRef] [PubMed]
  17. Schnegg, A.; Kirchgessner, M. Absorption and metabolic efficiency of iron in nickel deficiency. Int. J. Vitam. Nutr. Res. 1976, 46, 96–99. [Google Scholar] [PubMed]
  18. Tallkvist, J.; Tjalve, H. Effect of dietary iron-deficiency on the disposition of nickel in rats. Toxicol. Lett. 1997, 92, 131–138. [Google Scholar] [CrossRef]
  19. Haber, L.; Erdreicht, L.; Diamond, G.; Maier, A.; Ratney, R.; Zhao, Q.; Dourson, M. Hazard identification and dose response of inhaled nickel-soluble salts. Regul. Toxicol. Pharmacol. 2000, 31, 210–230. [Google Scholar] [CrossRef] [PubMed]
  20. Oller, A.R.; Costa, M.; Oberdorster, G. Carcinogenicity assessment of selected nickel compounds. Toxicol. Appl. Pharmacol. 1997, 143, 152–166. [Google Scholar] [CrossRef] [PubMed]
  21. Wataha, J.C.; Lockwood, P.E.; Schedle, A.; Noda, M.; Bouillaguet, S. Ag, Cu, Hg and Ni ions alter the metabolism of human monocytes during extended low-dose exposures. J. Oral Rehabil. 2002, 29, 133–139. [Google Scholar] [CrossRef] [PubMed]
  22. Das, K.K.; Das, S.N.; Dhundasi, S.A. Nickel, its adverse health effects & oxidative stress. Indian J. Med. Res. 2008, 128, 412–425. [Google Scholar] [PubMed]
  23. Li, X.; Zhong, F. Nickel induces interleukin-1β secretion via the NLRP3-ASC-caspase-1 pathway. Inflammation 2014, 37, 457–466. [Google Scholar] [CrossRef] [PubMed]
  24. Xu, S.; He, M.; Zhong, M.; Li, L.; Lu, Y.; Zhang, Y.; Zhang, L.; Yu, Z.; Zhou, Z. The neuroprotective effects of taurine against nickel by reducing oxidative stress and maintaining mitochondrial function in cortical neurons. Neurosci. Lett. 2015, 590, 52–57. [Google Scholar] [CrossRef] [PubMed]
  25. Gathwan, K.H.; Al-Karkhi, I.H.T.; Jaffar Al-Mulla, E.A. Hepatic toxicity of nickel chloride in mice. Res. Chem. Intermed. 2012, 39, 2537–2542. [Google Scholar] [CrossRef]
  26. Scutariu, M.D.; Ciupilan, C. Nickel and magnesium effects in the rat kidney, treated with acid retinoic. Comparative study. Rev. Med. Chir. Soc. Med. Nat. Iasi. 2007, 111, 744–747. [Google Scholar] [PubMed]
  27. Chen, C.Y.; Lin, T.K.; Chang, Y.C.; Wang, Y.F.; Shyu, H.W.; Lin, K.H.; Chou, M.C. Nickel (II)-induced oxidative stress, apoptosis, G2/M arrest, and genotoxicity in normal rat kidney cells. J. Toxicol. Environ. Health A 2010, 73, 529–539. [Google Scholar] [CrossRef] [PubMed]
  28. Goodman, J.E.; Prueitt, R.L.; Dodge, D.G.; Thakali, S. Carcinogenicity assessment of water-soluble nickel compounds. Crit. Rev. Toxicol. 2009, 39, 365–417. [Google Scholar] [CrossRef] [PubMed]
  29. Forgacs, Z.; Massanyi, P.; Lukac, N.; Somosy, Z. Reproductive toxicology of nickel—Review. J. Environ. Sci. Health A 2012, 47, 1249–1260. [Google Scholar] [CrossRef] [PubMed]
  30. Duman, F.; Ozturk, F. Nickel accumulation and its effect on biomass, protein content and antioxidative enzymes in roots and leaves of watercress (Nasturtium officinale R. Br.). J. Environ. Sci. 2010, 22, 526–532. [Google Scholar] [CrossRef]
  31. Kasprzak, K.S.; Sunderman, F.W., Jr.; Salnikow, K. Nickel carcinogenesis. Mutat. Res. 2003, 533, 67–97. [Google Scholar] [CrossRef] [PubMed]
  32. Spears, J.W.; Harvey, R.W.; Samsell, L.J. Effects of dietary nickel and protein on growth, nitrogen metabolism and tissue concentrations of nickel, iron, zinc, manganese and copper in calves. J. Nutr. 1986, 116, 1873–1882. [Google Scholar] [PubMed]
  33. Das, K.K.; Dasgupta, S. Effect of nickel on testicular nucleic acid concentrations of rats on protein restriction. Biol. Trace Elem. Res. 2000, 73, 175–180. [Google Scholar] [CrossRef]
  34. Ragsdale, S.W. Nickel biochemistry. Curr. Opin. Chem. Biol. 1998, 2, 208–215. [Google Scholar] [CrossRef]
  35. Denkhausa, E.S.K. Nickel essentiality, toxicity, and carcinogenicity. Crit. Rev. Oncol. Hemat. 2002, 42, 35–36. [Google Scholar] [CrossRef]
  36. Krockova, J.Z.; Massanyi, P.; Sirotkin, A.V.; Pivko, J.; Makarevich, A.V.; Lukac, N.; Capcarova, M.; Toman, R.; Polakova, Z. Nickel induced structural and functional alterations in mouse Leydig cells in vitro. J. Trace Elem. Med. Biol. 2011, 25, 14–18. [Google Scholar] [CrossRef] [PubMed]
  37. Shi, Z. Nickel carbonyl: Toxicity and human health. Sci. Total Environ. 1994, 148, 293–298. [Google Scholar] [CrossRef]
  38. Das, K.K.; Buchner, V. Effect of nickel exposure on peripheral tissues: Role of oxidative stress in toxicity and possible protection by ascorbic acid. Rev. Environ. Health 2007, 22, 157–173. [Google Scholar] [CrossRef] [PubMed]
  39. IARC. IARC Monograph on the Evaluation of Carcinogenic Risks to Humans; International Agency for Research on Cancer: Lyon, France, 1990; Volume 49, pp. 318–411. [Google Scholar]
  40. Cavallo, D.; Ursini, C.; Setini, A.; Chianese, C.; Piegari, P.; Perniconi, B.; Iavicoli, S. Evaluation of oxidative damage and inhibition of DNA repair in an in vitro study of nickel exposure. Toxicol. Vitro 2003, 17, 603–607. [Google Scholar] [CrossRef]
  41. Salnikow, K.; Zhitkovich, A. Genetic and epigenetic mechanisms in metal carcinogenesis and cocarcinogenesis: Nickel, arsenic, and chromium. Chem. Res. Toxicol. 2008, 21, 28–44. [Google Scholar] [CrossRef] [PubMed]
  42. Xu, Z.; Ren, T.; Xiao, C.; Li, H.; Wu, T. Nickel promotes the invasive potential of human lung cancer cells via TLR4/MyD88 signaling. Toxicology 2011, 285, 25–30. [Google Scholar] [CrossRef] [PubMed]
  43. Beyersmann, D.; Hartwig, A. Carcinogenic metal compounds: Recent insight into molecular and cellular mechanisms. Arch. Toxicol. 2008, 82, 493–512. [Google Scholar] [CrossRef] [PubMed]
  44. Chakrabarti, S.K.; Bai, C.; Subramanian, K.S. DNA-protein crosslinks induced by nickel compounds in isolated rat lymphocytes: Role of reactive oxygen species and specific amino acids. Toxicol. Appl. Pharmacol. 2001, 170, 153–165. [Google Scholar] [CrossRef] [PubMed]
  45. Lei, Y.X.; Chen, J.K.; Wu, Z.L. Detection of DNA strand breaks, DNA-protein crosslinks, and telomerase activity in nickel-transformed BALB/c-3T3 cells. Teratog. Carcinog. Mutagen. 2001, 21, 463–471. [Google Scholar] [CrossRef] [PubMed]
  46. Weber, C.W.; Reid, B.L. Nickel toxicity in growing chicks. J. Nutr. 1968, 95, 612–616. [Google Scholar] [PubMed]
  47. Szilagyi, M.; Szentmihalyi, S.; Anke, M. Changes in Some of the Biochemical Parameters in Ni and Mo Deficient Animals (Goat, Sheep, Pig, Chicken, Rat). Available online: http://agris.fao.org/agris-search/search.do?recordID=HU8200908 (accessed on 9 April 2015).
  48. Amudha, K.; Pari, L. Beneficial role of naringin, a flavanoid on nickel induced nephrotoxicity in rats. Chem. Biol. Interact. 2011, 193, 57–64. [Google Scholar] [CrossRef] [PubMed]
  49. Tang, K.; Guo, H.; Deng, J.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Wang, X.; Wu, B.; Li, J.; et al. Inhibitive Effects of Nickel Chloride (NiCl2) on Thymocytes. Biol. Trace Elem. Res. 2015, 164, 242–252. [Google Scholar] [CrossRef] [PubMed]
  50. Tang, K.; Li, J.; Yin, S.; Guo, H.; Deng, J.; Cui, H. Effects of nickel chloride on histopathological lesions and oxidative damage in the thymus. Health 2014, 6, 2875. [Google Scholar] [CrossRef]
  51. Huang, J.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Wang, X.; Wu, B. Effect of dietary nickel chloride on splenic immune function in broilers. Biol. Trace Elem. Res. 2014, 159, 183–191. [Google Scholar] [CrossRef] [PubMed]
  52. Huang, J.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Wang, X.; Wu, B. Downregulation of TLR4 and 7 mRNA expression levels in broiler’s spleen caused by diets supplemented with nickel chloride. Biol. Trace Elem. Res. 2014, 158, 353–358. [Google Scholar] [CrossRef] [PubMed]
  53. Guo, H.; Wu, B.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Wang, X.; Deng, J.; Yin, S.; et al. NiCl2-down-regulated antioxidant enzyme mRNA expression causes oxidative damage in the broiler’s kidney. Biol. Trace Elem. Res. 2014, 162, 288–295. [Google Scholar] [CrossRef] [PubMed]
  54. Wu, B.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Huang, J. Dietary nickel chloride restrains the development of small intestine in broilers. Biol. Trace Elem. Res. 2013, 155, 236–246. [Google Scholar] [CrossRef] [PubMed]
  55. Yin, S.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Wang, X.; Wu, B.; Guo, H. Toxic effect of NiCl2 on development of the bursa of Fabricius in broiler chickens. Oncotarget 2015. [Google Scholar] [CrossRef]
  56. Guo, H.; Deng, H.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Wang, X.; Wu, B.; Chen, K. Nickel chloride (NiCl2)-caused inflammatory responses via activation of NF-κB pathway and reduction of anti-inflammatory mediator expression in the kidney. Oncotarget 2015, 6, 28607–28620. [Google Scholar] [PubMed]
  57. Guo, H.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Wang, X.; Wu, B.; Chen, K.; Deng, J. Dietary NiCl2 causes G2/M cell cycle arrest in the broiler’s kidney. Oncotarget 2015, 6, 35964–35977. [Google Scholar] [PubMed]
  58. Hengartner, M.O. The biochemistry of apoptosis. Nature 2000, 407, 770–776. [Google Scholar] [CrossRef] [PubMed]
  59. Lin, M.T.; Beal, M.F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 2006, 443, 787–795. [Google Scholar] [CrossRef] [PubMed]
  60. Pulido, M. Metal-induced apoptosis: Mechanisms. Mutat. Res. 2003, 533, 227–241. [Google Scholar] [CrossRef] [PubMed]
  61. Jiang, X.; Wang, X. Cytochrome C-mediated apoptosis. Annu. Rev. Biochem. 2004, 73, 87–106. [Google Scholar] [CrossRef] [PubMed]
  62. Schulze-Osthoff, K.; Ferrari, D.; Los, M.; Wesselborg, S.; Peter, M.E. Apoptosis signaling by death receptors. Eur. J. Biochem. 1998, 254, 439–459. [Google Scholar] [CrossRef] [PubMed]
  63. Green, D.R.; Reed, J.C. Mitochondria and apoptosis. Science 1998, 281, 1309–1312. [Google Scholar] [CrossRef] [PubMed]
  64. Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef] [PubMed]
  65. Hockenbery, D.M.; Oltvai, Z.N.; Yin, X.M.; Milliman, C.L.; Korsmeyer, S.J. Bcl-2 functions in an antioxidant pathway to prevent apoptosis. Cell 1993, 75, 241–251. [Google Scholar] [CrossRef]
  66. Chao, D.T.; Korsmeyer, S.J. Bcl-2 family: Regulators of cell death. Annu. Rev. Immunol. 1998, 16, 395–419. [Google Scholar] [CrossRef] [PubMed]
  67. Besbes, S.; Mirshahi, M.; Pocard, M.; Billard, C. New dimension in therapeutic targeting of Bcl-2 family proteins. Oncotarget 2015, 6, 12862–12871. [Google Scholar] [CrossRef] [PubMed]
  68. Martinou, J.C.; Youle, R.J. Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Dev. Cell 2011, 21, 92–101. [Google Scholar] [CrossRef] [PubMed]
  69. Hetz, C. Bcl-2 protein family. Essential regulators of cell death. Preface. Adv. Exp. Med. Biol. 2010, 687, 7–8. [Google Scholar]
  70. Lavik, A.R.; Zhong, F.; Chang, M.J.; Greenberg, E.; Choudhary, Y.; Smith, M.R.; McColl, K.S.; Pink, J.; Reu, F.J.; Matsuyama, S.; et al. A synthetic peptide targeting the BH4 domain of Bcl-2 induces apoptosis in multiple myeloma and follicular lymphoma cells alone or in combination with agents targeting the BH3-binding pocket of Bcl-2. Oncotarget 2015, 6, 27388–27402. [Google Scholar] [CrossRef] [PubMed]
  71. Kelly, P.N.; White, M.J.; Goschnick, M.W.; Fairfax, K.A.; Tarlinton, D.M.; Kinkel, S.A.; Bouillet, P.; Adams, J.M.; Kile, B.T.; Strasser, A. Individual and overlapping roles of BH3-only proteins Bim and Bad in apoptosis of lymphocytes and platelets and in suppression of thymic lymphoma development. Cell Death Differ. 2010, 17, 1655–1664. [Google Scholar] [CrossRef] [PubMed]
  72. Ola, M.S.; Nawaz, M.; Ahsan, H. Role of Bcl-2 family proteins and caspases in the regulation of apoptosis. Mol. Cell. Biochem. 2011, 351, 41–58. [Google Scholar] [CrossRef] [PubMed]
  73. Schuler, M.; Green, D.R. Mechanisms of p53-dependent apoptosis. Biochem. Soc. Trans. 2001, 29, 684–688. [Google Scholar] [CrossRef] [PubMed]
  74. Osaki, M.; Oshimura, M.A.; Ito, H. PI3K-Akt pathway: Its functions and alterations in human cancer. Apoptosis Int. J. Program. Cell Death 2004, 9, 667–676. [Google Scholar] [CrossRef] [PubMed]
  75. Porta, C.; Figlin, R.A. Phosphatidylinositol-3-kinase/Akt signaling pathway and kidney cancer, and the therapeutic potential of phosphatidylinositol-3-kinase/Akt inhibitors. J. Urol. 2009, 182, 2569–2577. [Google Scholar] [CrossRef] [PubMed]
  76. Chen, J.; Yang, H.; Wen, J.; Luo, K.; Liu, Q.; Huang, Y.; Zheng, Y.; Tan, Z.; Huang, Q.; Fu, J. NHE9 induces chemoradiotherapy resistance in esophageal squamous cell carcinoma by upregulating the Src/Akt/β-catenin pathway and Bcl-2 expression. Oncotarget 2015, 6, 12405–12420. [Google Scholar] [CrossRef] [PubMed]
  77. Franke, T.F.; Hornik, C.P.; Segev, L.; Shostak, G.A.; Sugimoto, C. PI3K/Akt and apoptosis: Size matters. Oncogene 2003, 22, 8983–8998. [Google Scholar] [CrossRef] [PubMed]
  78. Aziz, M.H.; Nihal, M.; Fu, V.X.; Jarrard, D.F.; Ahmad, N. Resveratrol-caused apoptosis of human prostate carcinoma LNCaP cells is mediated via modulation of phosphatidylinositol 3′-kinase/Akt pathway and Bcl-2 family proteins. Mol. Cancer Ther. 2006, 5, 1335–1341. [Google Scholar] [CrossRef] [PubMed]
  79. Hu, L.; Sun, Y.; Hu, J. Catalpol inhibits apoptosis in hydrogen peroxide-induced endothelium by activating the PI3K/Akt signaling pathway and modulating expression of Bcl-2 and Bax. Eur. J. Pharmacol. 2010, 628, 155–163. [Google Scholar] [CrossRef] [PubMed]
  80. Pan, J.J.; Chang, Q.S.; Wang, X.; Son, Y.O.; Liu, J.; Zhang, Z.; Bi, Y.Y.; Shi, X. Activation of Akt/GSK3β and Akt/Bcl-2 signaling pathways in nickel-transformed BEAS-2B cells. Int. J. Oncol. 2011, 39, 1285–1294. [Google Scholar] [PubMed]
  81. Bratton, S.B.; Salvesen, G.S. Regulation of the Apaf-1 caspase-9 apoptosome. J. Cell Sci. 2010, 123, 3209–3214. [Google Scholar] [CrossRef] [PubMed]
  82. Martin, S.J.; Green, D.R. Protease activation during apoptosis: Death by a thousand cuts? Cell 1995, 82, 349. [Google Scholar] [CrossRef]
  83. Fan, T.J.; Han, L.H.; Cong, R.S.; Liang, J. Caspase family proteases and apoptosis. Acta Biochim. Biophys. Sin. 2005, 37, 719–727. [Google Scholar] [CrossRef] [PubMed]
  84. Lam, M.; Bhat, M.B.; Nunez, G.; Ma, J.; Distelhorst, C.W. Regulation of Bcl-xL channel activity by calcium. J. Biol. Chem. 1998, 273, 17307–17310. [Google Scholar] [PubMed]
  85. Parrish, A.B.; Freel, C.D.; Kornbluth, S. Cellular mechanisms controlling caspase activation and function. CSH Perspect. Biol. 2013, 5. [Google Scholar] [CrossRef]
  86. McIlwain, D.R.; Berger, T.; Mak, T.W. Caspase functions in cell death and disease. CSH. Perspect. Biol. 2013, 5, a008656. [Google Scholar]
  87. Brentnall, M.; Rodriguez-Menocal, L.; de Guevara, R.L.; Cepero, E.; Boise, L.H. Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol. 2013, 14, 32. [Google Scholar] [PubMed]
  88. Xu, G.; Shi, Y. Apoptosis signaling pathways and lymphocyte homeostasis. Cell Res. 2007, 17, 759–771. [Google Scholar] [PubMed]
  89. Salvesen, G.S. Caspases: Opening the boxes and interpreting the arrows. Cell Death Differ. 2002, 9, 3–5. [Google Scholar] [CrossRef] [PubMed]
  90. De Calignon, A.; Fox, L.M.; Pitstick, R.; Carlson, G.A.; Bacskai, B.J.; Spires-Jones, T.L.; Hyman, B.T. Caspase activation precedes and leads to tangles. Nature 2010, 464, 1201–1204. [Google Scholar] [CrossRef] [PubMed]
  91. Chang, D.W.; Xing, Z.; Capacio, V.L.; Peter, M.E.; Yang, X. Interdimer processing mechanism of procaspase-8 activation. EMBO J. 2003, 22, 4132–4142. [Google Scholar] [CrossRef] [PubMed]
  92. Boatright, K.M.; Deis, C.; Denault, J.B.; Sutherlin, D.P.; Salvesen, G.S. Activation of caspases-8 and -10 by FLIPL. Biochem. J. 2004, 382, 651–657. [Google Scholar] [CrossRef] [PubMed]
  93. Muzio, M.; Stockwell, B.R.; Stennicke, H.R.; Salvesen, G.S.; Dixit, V.M. An induced proximity model for caspase-8 activation. J. Biol. Chem. 1998, 273, 2926–3290. [Google Scholar] [CrossRef] [PubMed]
  94. Shi, Y. Mechanisms of caspase activation and inhibition during apoptosis. Mol. Cell 2002, 9, 459–470. [Google Scholar] [CrossRef]
  95. Tait, S.W.; Green, D.R. Mitochondria and cell death: Outer membrane permeabilization and beyond. Nat. Rev. Mol. Cell Biol. 2010, 11, 621–632. [Google Scholar] [CrossRef] [PubMed]
  96. Earnshaw, W.C.; Martins, L.M.; Kaufmann, S.H. Mammalian caspases: Structure, activation, substrates, and functions during apoptosis. Annu. Rev. Biochem. 1999, 68, 383–424. [Google Scholar] [CrossRef] [PubMed]
  97. Cohen, G. Caspases: The executioners of apoptosis. Biochem. J. 1997, 326, 1–16. [Google Scholar] [CrossRef] [PubMed]
  98. Nakagawa, T.; Zhu, H.; Morishima, N.; Li, E.; Xu, J.; Yankner, B.A.; Yuan, J. Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-β. Nature 2000, 403, 98–103. [Google Scholar] [CrossRef] [PubMed]
  99. Hitomi, J.; Katayama, T.; Eguchi, Y.; Kudo, T.; Taniguchi, M.; Koyama, Y.; Manabe, T.; Yamagishi, S.; Bando, Y.; Imaizumi, K.; et al. Involvement of caspase-4 in endoplasmic reticulum stress-induced apoptosis and Aβ-induced cell death. J. Cell Biol. 2004, 165, 347–356. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, Z.; Liu, Z.; Cao, Z.; Li, L. Allicin induces apoptosis in EL-4 cells in vitro by activation of expression of caspase-3 and -12 and up-regulation of the ratio of Bax/Bcl-2. Nat. Prod. Res. 2012, 26, 1033–1037. [Google Scholar] [CrossRef] [PubMed]
  101. Schmitz, I.; Kirchhoff, S.; Krammer, P.H. Regulation of death receptor-mediated apoptosis pathways. Int. J. Biochem. 2000, 32, 1123–1136. [Google Scholar] [CrossRef]
  102. Locksley, R.M.; Killeen, N.; Lenardo, M.J. The TNF and TNF receptor superfamilies: Integrating mammalian biology. Cell 2001, 104, 487–501. [Google Scholar] [CrossRef]
  103. Ashkenazi, A.; Dixit, V.M. Death receptors: Signaling and modulation. Science 1998, 281, 1305–1308. [Google Scholar] [CrossRef] [PubMed]
  104. Suliman, A.; Lam, A.; Datta, R.; Srivastava, R.K. Intracellular mechanisms of TRAIL: Apoptosis through mitochondrial-dependent and -independent pathways. Oncogene 2001, 20, 2122–2133. [Google Scholar] [CrossRef] [PubMed]
  105. Krammer, P.H. CD95(APO-1/Fas)-mediated apoptosis: Live and let die. Adv. Immunol. 1999, 71, 163–210. [Google Scholar] [PubMed]
  106. Rubio-Moscardo, F.; Blesa, D.; Mestre, C.; Siebert, R.; Balasas, T.; Benito, A.; Rosenwald, A.; Climent, J.; Martinez, J.I.; Schilhabel, M.; et al. Characterization of 8p21.3 chromosomal deletions in B-cell lymphoma: TRAIL-R1 and TRAIL-R2 as candidate dosage-dependent tumor suppressor genes. Blood 2005, 106, 3214–3222. [Google Scholar] [CrossRef] [PubMed]
  107. Danial, N.N.; Korsmeyer, S.J. Cell death: Critical control points. Cell 2004, 116, 205–219. [Google Scholar] [CrossRef]
  108. Marino, G.; Niso-Santano, M.; Baehrecke, E.H.; Kroemer, G. Self-consumption: The interplay of autophagy and apoptosis. Nat. Rev. Mol. Cell Biol. 2014, 15, 81–94. [Google Scholar] [CrossRef] [PubMed]
  109. Scott, F.L.; Stec, B.; Pop, C.; Dobaczewska, M.K.; Lee, J.J.; Monosov, E.; Robinson, H.; Salvesen, G.S.; Schwarzenbacher, R.; Riedl, S.J. The Fas-FADD death domain complex structure unravels signalling by receptor clustering. Nature 2009, 457, 1019–1022. [Google Scholar] [CrossRef] [PubMed]
  110. Kichev, A.; Rousset, C.I.; Baburamani, A.A.; Levison, S.W.; Wood, T.L.; Gressens, P.; Thornton, C.; Hagberg, H. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling and cell death in the immature central nervous system after hypoxia-ischemia and inflammation. J. Biol. Chem. 2014, 289, 9430–9439. [Google Scholar] [CrossRef] [PubMed]
  111. Curtin, J.F.; Cotter, T.G. Live and let die: Regulatory mechanisms in Fas-mediated apoptosis. Cell Signal. 2003, 15, 983–992. [Google Scholar] [CrossRef]
  112. Dickens, L.S.; Powley, I.R.; Hughes, M.A.; MacFarlane, M. The “complexities” of life and death: Death receptor signalling platforms. Exp. Cell. Res. 2012, 318, 1269–1277. [Google Scholar] [CrossRef] [PubMed]
  113. Lavrik, I.N.; Krammer, P.H. Regulation of CD95/Fas signaling at the DISC. Cell Death Differ. 2012, 19, 36–41. [Google Scholar] [CrossRef] [PubMed]
  114. Nicholson, D.W.; Ali, A.; Thornberry, N.A.; Vaillancourt, J.P.; Ding, C.K.; Gallant, M.; Gareau, Y.; Griffin, P.R.; Labelle, M.; Lazebnik, Y.A.; et al. Identification and inhibition of the ICE/CED-3 protease necessary for mammalian apoptosis. Nature 1995, 376, 37–43. [Google Scholar] [CrossRef] [PubMed]
  115. Mishra, N.C.; Kumar, S. Apoptosis: A mitochondrial perspective on cell death. Indian J. Exp. Biol. 2005, 43, 25–34. [Google Scholar] [PubMed]
  116. Logue, S.E.; Cleary, P.; Saveljeva, S.; Samali, A. New directions in ER stress-induced cell death. Apoptosis Int. J. Program. Cell Death 2013, 18, 537–546. [Google Scholar] [CrossRef] [PubMed]
  117. Vaux, D.L. Apoptogenic factors released from mitochondria. Biochim. Biophys. Acta 2011, 1813, 546–550. [Google Scholar] [CrossRef] [PubMed]
  118. Du, C.; Fang, M.; Li, Y.; Li, L.; Wang, X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000, 102, 33–42. [Google Scholar] [CrossRef]
  119. Duckett, C.S. IAP proteins: Sticking it to Smac. Biochem. J. 2005, 385, e1–e2. [Google Scholar] [CrossRef] [PubMed]
  120. Garrido, C.; Galluzzi, L.; Brunet, M.; Puig, P.E.; Didelot, C.; Kroemer, G. Mechanisms of cytochrome c release from mitochondria. Cell Death Differ. 2006, 13, 1423–1433. [Google Scholar] [CrossRef] [PubMed]
  121. Vande Walle, L.; Lamkanfi, M.; Vandenabeele, P. The mitochondrial serine protease HtrA2/Omi: An overview. Cell Death Differ. 2008, 15, 453–460. [Google Scholar] [CrossRef] [PubMed]
  122. Deveraux, Q.L.; Reed, J.C. IAP family proteins—Suppressors of apoptosis. Genes Dev. 1999, 13, 239–252. [Google Scholar] [CrossRef] [PubMed]
  123. Hasegawa, T.; Suzuki, K.; Sakamoto, C.; Ohta, K.; Nishiki, S.; Hino, M.; Tatsumi, N.; Kitagawa, S. Expression of the inhibitor of apoptosis (IAP) family members in human neutrophils: Up-regulation of cIAP2 by granulocyte colony-stimulating factor and overexpression of cIAP2 in chronic neutrophilic leukemia. Blood 2003, 101, 1164–1171. [Google Scholar] [CrossRef] [PubMed]
  124. Wu, G.; Chai, J.; Suber, T.L.; Wu, J.W.; Du, C.; Wang, X.; Shi, Y. Structural basis of IAP recognition by Smac/DIABLO. Nature 2000, 408, 1008–1012. [Google Scholar] [PubMed]
  125. Norberg, E.; Orrenius, S.; Zhivotovsky, B. Mitochondrial regulation of cell death: Processing of apoptosis-inducing factor (AIF). Biochem. Biophys Res. Commun. 2010, 396, 95–100. [Google Scholar] [CrossRef] [PubMed]
  126. Buttner, S.; Eisenberg, T.; Carmona-Gutierrez, D.; Ruli, D.; Knauer, H.; Ruckenstuhl, C.; Sigrist, C.; Wissing, S.; Kollroser, M.; Frohlich, K.U.; et al. Endonuclease G regulates budding yeast life and death. Mol. Cell 2007, 25, 233–246. [Google Scholar] [CrossRef] [PubMed]
  127. Van Loo, G.; Schotte, P.; van Gurp, M.; Demol, H.; Hoorelbeke, B.; Gevaert, K.; Rodriguez, I.; Ruiz-Carrillo, A.; Vandekerckhove, J.; Declercq, W.; et al. Endonuclease G: A mitochondrial protein released in apoptosis and involved in caspase-independent DNA degradation. Cell Death Differ. 2001, 8, 1136–1142. [Google Scholar] [CrossRef] [PubMed]
  128. Urra, H.; Dufey, E.; Lisbona, F.; Rojas-Rivera, D.; Hetz, C. When ER stress reaches a dead end. Biochim. Biophys. Acta 2013, 1833, 3507–3517. [Google Scholar] [CrossRef] [PubMed]
  129. Sano, R.; Reed, J.C. ER stress-induced cell death mechanisms. Biochim. Biophys. Acta 2013, 1833, 3460–3470. [Google Scholar] [CrossRef] [PubMed]
  130. Yoshida, H. ER stress and diseases. FEBS J. 2007, 274, 630–658. [Google Scholar] [CrossRef] [PubMed]
  131. Iwasaki, N.; Sugiyama, Y.; Miyazaki, S.; Nakagawa, H.; Nishimura, K.; Matsuo, S. An ATF4-signal-modulating machine other than GADD34 acts in ATF4-to-CHOP signaling to block CHOP expression in ER-stress-related autophagy. J. Cell. Biochem. 2015, 116, 1300–1309. [Google Scholar] [CrossRef] [PubMed]
  132. Ohoka, N.; Yoshii, S.; Hattori, T.; Onozaki, K.; Hayashi, H. TRB3, a novel ER stress-inducible gene, is induced via ATF4-CHOP pathway and is involved in cell death. EMBO J. 2005, 24, 1243–1255. [Google Scholar] [CrossRef] [PubMed]
  133. Verfaillie, T.; Rubio, N.; Garg, A.D.; Bultynck, G.; Rizzuto, R.; Decuypere, J.P.; Piette, J.; Linehan, C.; Gupta, S.; Samali, A.; et al. PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death Differ. 2012, 19, 1880–1891. [Google Scholar] [CrossRef] [PubMed]
  134. Hetz, C.A. ER stress signaling and the Bcl-2 family of proteins: From adaptation to irreversible cellular damage. Antioxid. Redox Signal. 2007, 9, 2345–2355. [Google Scholar] [CrossRef] [PubMed]
  135. Huang, H.L.; Wu, J.L.; Chen, M.H.; Hong, J.R. Aquatic birnavirus-induced ER stress-mediated death signaling contribute to downregulation of Bcl-2 family proteins in salmon embryo cells. PLoS ONE 2011, 6, e22935. [Google Scholar] [CrossRef] [PubMed]
  136. Li, G.; Mongillo, M.; Chin, K.T.; Harding, H.; Ron, D.; Marks, A.R.; Tabas, I. Role of ERO1-α-mediated stimulation of inositol 1,4,5-triphosphate receptor activity in endoplasmic reticulum stress-induced apoptosis. J. Cell Biol. 2009, 186, 783–792. [Google Scholar] [CrossRef] [PubMed]
  137. Zhu, H.; Yang, W.; He, L.J.; Ding, W.J.; Zheng, L.; Liao, S.D.; Huang, P.; Lu, W.; He, Q.J.; Yang, B. Upregulating Noxa by ER stress, celastrol exerts synergistic anti-cancer activity in combination with ABT-737 in human hepatocellular carcinoma cells. PLoS ONE 2012, 7, e52333. [Google Scholar] [CrossRef] [PubMed]
  138. Wang, Q.; Mora-Jensen, H.; Weniger, M.A.; Perez-Galan, P.; Wolford, C.; Hai, T.; Ron, D.; Chen, W.; Trenkle, W.; Wiestner, A.; et al. ERAD inhibitors integrate ER stress with an epigenetic mechanism to activate BH3-only protein NOXA in cancer cells. Proc. Natl. Acad. Sci. USA 2009, 106, 2200–2205. [Google Scholar] [CrossRef] [PubMed]
  139. Gautam, S.; Kirschnek, S.; Wiesmeier, M.; Vier, J.; Hacker, G. Roscovitine-induced apoptosis in neutrophils and neutrophil progenitors is regulated by the Bcl-2-family members Bim, Puma, Noxa and Mcl-1. PLoS ONE 2013, 8, e79352. [Google Scholar] [CrossRef] [PubMed]
  140. Han, D.; Lerner, A.G.; Vande Walle, L.; Upton, J.P.; Xu, W.; Hagen, A.; Backes, B.J.; Oakes, S.A.; Papa, F.R. IRE1α kinase activation modes control alternate endoribonuclease outputs to determine divergent cell fates. Cell 2009, 138, 562–575. [Google Scholar] [CrossRef] [PubMed]
  141. Jurczak, M.J.; Lee, A.H.; Jornayvaz, F.R.; Lee, H.Y.; Birkenfeld, A.L.; Guigni, B.A.; Kahn, M.; Samuel, V.T.; Glimcher, L.H.; Shulman, G.I. Dissociation of inositol-requiring enzyme (IRE1α)-mediated c-Jun N-terminal kinase activation from hepatic insulin resistance in conditional X-box-binding protein-1 (XBP1) knock-out mice. J. Biol. Chem. 2012, 287, 2558–2567. [Google Scholar] [CrossRef] [PubMed]
  142. Urano, F.; Wang, X.; Bertolotti, A.; Zhang, Y.; Chung, P.; Harding, H.P.; Ron, D. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 2000, 287, 664–666. [Google Scholar] [CrossRef] [PubMed]
  143. Furuhata, M.; Takada, E.; Noguchi, T.; Ichijo, H.; Mizuguchi, J. Apoptosis signal-regulating kinase (ASK)-1 mediates apoptosis through activation of JNK1 following engagement of membrane immunoglobulin. Exp. Cell Res. 2009, 315, 3467–3476. [Google Scholar] [CrossRef] [PubMed]
  144. Newton, V.L.; Ali, S.; Duddy, G.; Whitmarsh, A.J.; Gardiner, N.J. Targeting apoptosis signalling kinase-1 (ASK-1) does not prevent the development of neuropathy in streptozotocin-induced diabetic mice. PLoS ONE 2014, 9, e107437. [Google Scholar] [CrossRef] [PubMed]
  145. Michalak, M.; Robert Parker, J.M.; Opas, M. Ca2+ signaling and calcium binding chaperones of the endoplasmic reticulum. Cell Calcium 2002, 32, 269–278. [Google Scholar] [CrossRef] [PubMed]
  146. Timmins, J.M.; Ozcan, L.; Seimon, T.A.; Li, G.; Malagelada, C.; Backs, J.; Backs, T.; Bassel-Duby, R.; Olson, E.N.; Anderson, M.E.; et al. Calcium/calmodulin-dependent protein kinase II links ER stress with Fas and mitochondrial apoptosis pathways. J. Clin. Investig. 2009, 119, 2925–2941. [Google Scholar] [CrossRef] [PubMed]
  147. Rizzuto, R.; Pinton, P.; Carrington, W.; Fay, F.S.; Fogarty, K.E.; Lifshitz, L.M.; Tuft, R.A.; Pozzan, T. Close contacts with the endoplasmic reticulum as determinants of mitochondrial Ca2+ responses. Science 1998, 280, 1763–1766. [Google Scholar] [CrossRef] [PubMed]
  148. Bravo, R.; Vicencio, J.M.; Parra, V.; Troncoso, R.; Munoz, J.P.; Bui, M.; Quiroga, C.; Rodriguez, A.E.; Verdejo, H.E.; Ferreira, J.; et al. Increased ER-mitochondrial coupling promotes mitochondrial respiration and bioenergetics during early phases of ER stress. J. Cell Sci. 2011, 124, 2143–2152. [Google Scholar] [CrossRef] [PubMed]
  149. Nakagawa, T.; Yuan, J. Cross-talk between two cysteine protease families: Activation of caspase-12 by calpain in apoptosis. J. Cell Biol. 2000, 150, 887–894. [Google Scholar] [CrossRef] [PubMed]
  150. Fischer, H.; Koenig, U.; Eckhart, L.; Tschachler, E. Human caspase 12 has acquired deleterious mutations. Biochem. Biophys. Res. Commun. 2002, 293, 722–726. [Google Scholar] [CrossRef]
  151. Bian, Z.M.; Elner, S.G.; Elner, V.M. Dual involvement of caspase-4 in inflammatory and ER stress-induced apoptotic responses in human retinal pigment epithelial cells. Investig. Ophthalmol. Vis. Sci. 2009, 50, 6006–6014. [Google Scholar] [CrossRef] [PubMed]
  152. Su, L.; Deng, Y.; Zhang, Y.; Li, C.; Zhang, R.; Sun, Y.; Zhang, K.; Li, J.; Yao, S. Protective effects of grape seed procyanidin extract against nickel sulfate-induced apoptosis and oxidative stress in rat testes. Toxicol. Mech. Methods 2011, 21, 487–494. [Google Scholar] [CrossRef] [PubMed]
  153. Liu, C.M.; Zheng, G.H.; Ming, Q.L.; Chao, C.; Sun, J.M. Sesamin protects mouse liver against nickel-induced oxidative DNA damage and apoptosis by the PI3K-Akt pathway. J. Agric. Food Chem. 2013, 61, 1146–1154. [Google Scholar] [CrossRef] [PubMed]
  154. Zheng, G.H.; Liu, C.M.; Sun, J.M.; Feng, Z.J.; Cheng, C. Nickel-induced oxidative stress and apoptosis in Carassius auratus liver by JNK pathway. Aquat. Toxicol. 2014, 147, 105–111. [Google Scholar] [CrossRef] [PubMed]
  155. Wu, B.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Huang, J. Dietary nickel chloride induces oxidative stress, apoptosis and alters Bax/Bcl-2 and caspase-3 mRNA expression in the cecal tonsil of broilers. Food Chem. Toxicol. 2014, 63, 18–29. [Google Scholar] [CrossRef] [PubMed]
  156. Huang, J.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Wu, B. The association between splenocyte apoptosis and alterations of Bax, Bcl-2 and caspase-3 mRNA expression, and oxidative stress induced by dietary nickel chloride in broilers. Int. J. Environ. Res. Public Health 2013, 10, 7310–7326. [Google Scholar] [CrossRef] [PubMed]
  157. Nowak, M.; Kopp, F.; Roelofs-Haarhuis, K.; Wu, X.; Gleichmann, E. Oral nickel tolerance: Fas ligand-expressing invariant NK T cells promote tolerance induction by eliciting apoptotic death of antigen-carrying, effete B cells. J. Immunol. 2006, 176, 4581–4589. [Google Scholar] [CrossRef] [PubMed]
  158. Guan, F.; Zhang, D.; Wang, X.; Chen, J. Nitric oxide and Bcl-2 mediated the apoptosis induced by nickel(II) in human T hybridoma cells. Toxicol. Appl. Pharmacol. 2007, 221, 86–94. [Google Scholar] [CrossRef] [PubMed]
  159. Kang, J.; Zhang, D.; Chen, J.; Lin, C.; Liu, Q. Involvement of histone hypoacetylation in Ni2+-induced Bcl-2 down-regulation and human hepatoma cell apoptosis. J. Biol. Inorg. Chem. 2004, 9, 713–723. [Google Scholar] [CrossRef] [PubMed]
  160. Cavani, A. Breaking tolerance to nickel. Toxicology 2005, 209, 119–121. [Google Scholar] [CrossRef] [PubMed]
  161. Siddiqui, M.A.; Ahamed, M.; Ahmad, J.; Majeed Khan, M.A.; Musarrat, J.; Al-Khedhairy, A.A.; Alrokayan, S.A. Nickel oxide nanoparticles induce cytotoxicity, oxidative stress and apoptosis in cultured human cells that is abrogated by the dietary antioxidant curcumin. Food Chem. Toxicol. 2012, 50, 641–647. [Google Scholar] [CrossRef] [PubMed]
  162. Ahamed, M.; Ali, D.; Alhadlaq, H.A.; Akhtar, M.J. Nickel oxide nanoparticles exert cytotoxicity via oxidative stress and induce apoptotic response in human liver cells (HepG2). Chemosphere 2013, 93, 2514–2522. [Google Scholar] [CrossRef] [PubMed]
  163. Freitas, M.; Barcellos-de-Souza, P.; Barja-Fidalgo, C.; Fernandes, E. Nickel induces apoptosis in human neutrophils. Biometals Int. J. Role Metal Ions Biol. Biochem. Med. 2013, 26, 13–21. [Google Scholar] [CrossRef] [PubMed]
  164. Chen, C.Y.; Wang, Y.F.; Huang, W.R.; Huang, Y.T. Nickel induces oxidative stress and genotoxicity in human lymphocytes. Toxicol. Appl. Pharmacol. 2003, 189, 153–159. [Google Scholar] [CrossRef]
  165. Shiao, Y.H.; Lee, S.H.; Kasprzak, K.S. Cell cycle arrest, apoptosis and p53 expression in nickel(II) acetate-treated Chinese hamster ovary cells. Carcinogenesis 1998, 19, 1203–1207. [Google Scholar] [CrossRef] [PubMed]
  166. Venditti, P.; Di Stefano, L.; Di Meo, S. Mitochondrial metabolism of reactive oxygen species. Mitochondrion 2013, 13, 71–82. [Google Scholar] [CrossRef] [PubMed]
  167. Dallas, L.J.; Bean, T.P.; Turner, A.; Lyons, B.P.; Jha, A.N. Oxidative DNA damage may not mediate Ni-induced genotoxicity in marine mussels: Assessment of genotoxic biomarkers and transcriptional responses of key stress genes. Mutat. Res. 2013, 754, 22–31. [Google Scholar] [CrossRef] [PubMed]
  168. Mates, J.M.; Segura, J.A.; Alonso, F.J.; Marquez, J. Oxidative stress in apoptosis and cancer: An update. Arch. Toxicol. 2012, 86, 1649–1665. [Google Scholar] [CrossRef] [PubMed]
  169. Avery, S.V. Molecular targets of oxidative stress. Biochem. J. 2011, 434, 201–210. [Google Scholar] [CrossRef] [PubMed]
  170. Simon, H.U.; Haj-Yehia, A.; Levi-Schaffer, F. Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis Int. J. Program. Cell Death 2000, 5, 415–418. [Google Scholar] [CrossRef]
  171. Jomova, K.; Valko, M. Advances in metal-induced oxidative stress and human disease. Toxicology 2011, 283, 65–87. [Google Scholar] [CrossRef] [PubMed]
  172. Valko, M.; Morris, H.; Cronin, M. Metals, toxicity and oxidative stress. Curr. Med. Chem. 2005, 12, 1161–1208. [Google Scholar] [CrossRef] [PubMed]
  173. Ma, C.; Song, M.; Zhang, Y.; Yan, M.; Zhang, M.; Bi, H. Nickel nanowires induce cell cycle arrest and apoptosis by generation of reactive oxygen species in HeLa cells. Toxicol. Rep. 2014, 1, 114–121. [Google Scholar] [CrossRef]
  174. Pan, J.; Chang, Q.; Wang, X.; Son, Y.; Zhang, Z.; Chen, G.; Luo, J.; Bi, Y.; Chen, F.; Shi, X. Reactive oxygen species-activated Akt/ASK1/p38 signaling pathway in nickel compound-induced apoptosis in BEAS 2B cells. Chem. Res. Toxicol. 2010, 23, 568–577. [Google Scholar] [CrossRef] [PubMed]
  175. Ahamed, M. Toxic response of nickel nanoparticles in human lung epithelial A549 cells. Toxicol. Vitro 2011, 25, 930–936. [Google Scholar] [CrossRef] [PubMed]
  176. Kubrak, O.I.; Husak, V.V.; Rovenko, B.M.; Poigner, H.; Mazepa, M.A.; Kriews, M.; Abele, D.; Lushchak, V.I. Tissue specificity in nickel uptake and induction of oxidative stress in kidney and spleen of goldfish Carassius auratus, exposed to waterborne nickel. Aquat. Toxicol. 2012, 118–119, 88–96. [Google Scholar] [CrossRef] [PubMed]
  177. Wu, B.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Huang, J. Dietary nickel chloride induces oxidative intestinal damage in broilers. Int. J. Environ. Res. Publ. Health 2013, 10, 2109–2119. [Google Scholar] [CrossRef] [PubMed]
  178. Wang, M.; Wang, G. Oxidative damage effects in the copepod Tigriopus japonicus Mori experimentally exposed to nickel. Ecotoxicology 2010, 19, 273–284. [Google Scholar] [CrossRef] [PubMed]
  179. Attig, H.; Kamel, N.; Sforzini, S.; Dagnino, A.; Jamel, J.; Boussetta, H.; Viarengo, A.; Banni, M. Effects of thermal stress and nickel exposure on biomarkers responses in Mytilus galloprovincialis (Lam). Mar. Environ. Res. 2014, 94, 65–71. [Google Scholar] [CrossRef] [PubMed]
  180. Alarifi, S.; Ali, D.; Alakhtani, S.; Al Suhaibani, E.S.; Al-Qahtani, A.A. Reactive oxygen species-mediated DNA damage and apoptosis in human skin epidermal cells after exposure to nickel nanoparticles. Biol. Trace Elem. Res. 2014, 157, 84–93. [Google Scholar] [CrossRef] [PubMed]
  181. Lewis, J.B.; Messer, R.L.; McCloud, V.V.; Lockwood, P.E.; Hsu, S.D.; Wataha, J.C. Ni(II) activates the Nrf2 signaling pathway in human monocytic cells. Biomaterials 2006, 27, 5348–5356. [Google Scholar] [CrossRef] [PubMed]
  182. Krezel, A.; Szczepanik, W.; Sokolowska, M.; Jezowska-Bojczuk, M.; Bal, W. Correlations between complexation modes and redox activities of Ni(II)-GSH complexes. Chem. Res. Toxicol. 2003, 16, 855–864. [Google Scholar] [CrossRef] [PubMed]
  183. Zhao, J.; Bowman, L.; Zhang, X.; Shi, X.; Jiang, B.; Castranova, V.; Ding, M. Metallic nickel nano- and fine particles induce JB6 cell apoptosis through a caspase-8/AIF mediated cytochrome c-independent pathway. J. Nanobiotechnol. 2009, 7, 2. [Google Scholar] [CrossRef] [PubMed]
  184. Bonin, S.; Larese, F.F.; Trevisan, G.; Avian, A.; Rui, F.; Stanta, G.; Bovenzi, M. Gene expression changes in peripheral blood mononuclear cells in occupational exposure to nickel. Exp. Dermatol. 2011, 20, 147–148. [Google Scholar] [CrossRef] [PubMed]
  185. Susin, S.A.; Zamzami, N.; Castedo, M.; Hirsch, T.; Marchetti, P.; Macho, A.; Daugas, E.; Geuskens, M.; Kroemer, G. Bcl-2 inhibits the mitochondrial release of an apoptogenic protease. J. Exp. Med. 1996, 184, 1331–1341. [Google Scholar] [CrossRef] [PubMed]
  186. Wang, Y.F.; Shyu, H.W.; Chang, Y.C.; Tseng, W.C.; Huang, Y.L.; Lin, K.H.; Chou, M.C.; Liu, H.L.; Chen, C.Y. Nickel (II)-induced cytotoxicity and apoptosis in human proximal tubule cells through a ROS- and mitochondria-mediated pathway. Toxicol. Appl. Pharmacol. 2012, 259, 177–186. [Google Scholar] [CrossRef] [PubMed]
  187. Ahamed, M.; Akhtar, M.J.; Alhadlaq, H.A.; Khan, M.A.; Alrokayan, S.A. Comparative cytotoxic response of nickel ferrite nanoparticles in human liver HepG2 and breast MFC-7 cancer cells. Chemosphere 2015, 135, 278–288. [Google Scholar] [CrossRef] [PubMed]
  188. Patel, E.; Lynch, C.; Ruff, V.; Reynolds, M. Co-exposure to nickel and cobalt chloride enhances cytotoxicity and oxidative stress in human lung epithelial cells. Toxicol. Appl. Pharmacol. 2012, 258, 367–375. [Google Scholar] [CrossRef] [PubMed]
  189. Duan, W.X.; He, M.D.; Mao, L.; Qian, F.H.; Li, Y.M.; Pi, H.F.; Liu, C.; Chen, C.H.; Lu, Y.H.; Cao, Z.W.; et al. NiO nanoparticles induce apoptosis through repressing SIRT1 in human bronchial epithelial cells. Toxicol. Appl. Pharmacol. 2015, 286, 80–91. [Google Scholar] [CrossRef] [PubMed]
  190. Buschini, A.; Pinelli, S.; Pellacani, C.; Giordani, F.; Ferrari, M.B.; Bisceglie, F.; Giannetto, M.; Pelosi, G.; Tarasconi, P. Synthesis, characterization and deepening in the comprehension of the biological action mechanisms of a new nickel complex with antiproliferative activity. J. Inorg. Biochem. 2009, 103, 666–677. [Google Scholar] [CrossRef] [PubMed]
  191. Hiramatsu, N.; Kasai, A.; Du, S.; Takeda, M.; Hayakawa, K.; Okamura, M.; Yao, J.; Kitamura, M. Rapid, transient induction of ER stress in the liver and kidney after acute exposure to heavy metal: Evidence from transgenic sensor mice. FEBS Lett. 2007, 581, 2055–2059. [Google Scholar] [CrossRef] [PubMed]
  192. Hoffman, B.; Liebermann, D.A. Apoptotic signaling by c-Myc. Oncogene 2008, 27, 6462–6472. [Google Scholar] [CrossRef] [PubMed]
  193. Nilsson, J.A.; Cleveland, J.L. Myc pathways provoking cell suicide and cancer. Oncogene 2003, 22, 9007–9021. [Google Scholar] [CrossRef] [PubMed]
  194. Juin, P.; Hueber, A.O.; Littlewood, T.; Evan, G. c-Myc-induced sensitization to apoptosis is mediated through cytochrome c release. Genes Dev. 1999, 13, 1367–1381. [Google Scholar] [CrossRef] [PubMed]
  195. Eischen, C.M.; Woo, D.; Roussel, M.F.; Cleveland, J.L. Apoptosis triggered by Myc-induced suppression of Bcl-X(L) or Bcl-2 is bypassed during lymphomagenesis. Mol. Cell. Biol. 2001, 21, 5063–5070. [Google Scholar] [CrossRef] [PubMed]
  196. Li, Q.; Suen, T.C.; Sun, H.; Arita, A.; Costa, M. Nickel compounds induce apoptosis in human bronchial epithelial Beas-2B cells by activation of c-Myc through ERK pathway. Toxicol. Appl. Pharmacol. 2009, 235, 191–198. [Google Scholar] [CrossRef] [PubMed]
  197. Cimmino, A.; Calin, G.A.; Fabbri, M.; Iorio, M.V.; Ferracin, M.; Shimizu, M.; Wojcik, S.E.; Aqeilan, R.I.; Zupo, S.; Dono, M.; et al. miR-15 and miR-16 induce apoptosis by targeting Bcl2. Proc. Natl. Acad. Sci. USA 2005, 102, 13944–13949. [Google Scholar] [CrossRef] [PubMed]
  198. Jovanovic, M.; Hengartner, M.O. miRNAs and apoptosis: RNAs to die for. Oncogene 2006, 25, 6176–6187. [Google Scholar] [CrossRef] [PubMed]
  199. Lima, R.T.; Busacca, S.; Almeida, G.M.; Gaudino, G.; Fennell, D.A.; Vasconcelos, M.H. MicroRNA regulation of core apoptosis pathways in cancer. Eur. J. Cancer 2011, 47, 163–174. [Google Scholar] [CrossRef] [PubMed]

Share and Cite

MDPI and ACS Style

Guo, H.; Chen, L.; Cui, H.; Peng, X.; Fang, J.; Zuo, Z.; Deng, J.; Wang, X.; Wu, B. Research Advances on Pathways of Nickel-Induced Apoptosis. Int. J. Mol. Sci. 2016, 17, 10. https://doi.org/10.3390/ijms17010010

AMA Style

Guo H, Chen L, Cui H, Peng X, Fang J, Zuo Z, Deng J, Wang X, Wu B. Research Advances on Pathways of Nickel-Induced Apoptosis. International Journal of Molecular Sciences. 2016; 17(1):10. https://doi.org/10.3390/ijms17010010

Chicago/Turabian Style

Guo, Hongrui, Lian Chen, Hengmin Cui, Xi Peng, Jing Fang, Zhicai Zuo, Junliang Deng, Xun Wang, and Bangyuan Wu. 2016. "Research Advances on Pathways of Nickel-Induced Apoptosis" International Journal of Molecular Sciences 17, no. 1: 10. https://doi.org/10.3390/ijms17010010

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop