Abstract
Environmental and/or occupational exposure to metals such as Arsenic (As), Cadmium (Cd), and Chromium (Cr) have been shown to induce carcinogenesis in various organs, including the urogenital system. However, the mechanisms responsible for metal-induced carcinogenesis remain elusive. We and others have shown that metals are potent inducers of autophagy, which has been suggested to be an adaptive stress response to allow metal-exposed cells to survive in hostile environments. Albeit few, recent experimental studies have shown that As and Cd promote tumorigenesis via autophagy and that inhibition of autophagic signaling suppressed metal-induced carcinogenesis. In light of the newly emerging role of autophagic involvement in metal-induced carcinogenesis, the present review focuses explicitly on the mechanistic role of autophagy and potential signaling pathways involved in As-, Cd-, and Cr-induced urogenital carcinogenesis.
Keywords: Molecular signaling, impaired autophagy, Cadmium, Arsenic, Chromium
1. Introduction:
Metal-induced carcinogenesis caused by environmental and/or occupational exposure to heavy metals is rapidly becoming a global health problem due to their increased use in industrial, agricultural, and pharmaceutical applications [1]. Unlike organic contaminants, metals cannot be broken down by microorganisms and tend to bioaccumulate in living organisms [2, 3]; this chronic exposure, in turn, has been linked to various malignancies in humans [1, 4–6]. Heavy metals such as Arsenic (As), Cadmium (Cd), and Chromium (Cr) are well-known carcinogens [2, 7, 8] that have been associated with an increased risk of urogenital cancers [3, 5, 9–11]. While the exact mechanism of heavy metal-induced carcinogenesis remains elusive, studies have shown that metals may drive carcinogenesis via several mechanisms, including by causing DNA damage, uncontrolled oxidative stress, impaired autophagy, and epigenetic modifications such as the silencing of DNA repair and tumor-suppressor genes [1, 9, 12]. In the present review, we focus specifically on the role of autophagy and discuss possible signaling pathways responsible for metal-induced urogenital carcinogenesis.
2. Molecular machinery of autophagy
Mammalian autophagy is an essential catabolic process involving the lysosomal degradation and recycling of cellular proteins and organelles. Essentially a survival mechanism, autophagy is dramatically upregulated in conditions of metabolic stress (such as nutrient deprivation and bioenergetics failure) to increase the availability of critical metabolic intermediates [13, 14]. On the other hand, autophagy has also been shown to accompany type II programmed cell death, as characterized by the appearance of large intracellular vesicles and the engagement of the autophagy machinery; however, its function as an active cell death mechanism remains controversial [15].
Three types of autophagy are known to occur in eukaryotic cells, namely, macroautophagy, microautophagy, and chaperone-mediated autophagy [16], with macroautophagy (referred to as autophagy) providing the basis for the majority of our knowledge of this process. Autophagy is initiated by forming an isolating membrane or phagophore, which gradually closes to form a double-membrane vesicle or autophagosome. The autophagosome then fuses with a lysosome to form an autolysosome, which enables its contents to be degraded and recycled. This multi-step process is tightly regulated by autophagy-related genes (Atg) and several downstream signaling pathways (Figure 1), which have been discussed below.
Figure 1:

Molecular pathways involved in the regulation of autophagy in metal-induced carcinogenesis. Metal induces uncontrolled oxidative stress and excessive ROS production, which triggers ER stress, activation of antioxidant response (p62/Nrf2), and DNA damage. Metal exposure also induces epigenetic changes that affect DNA repair pathways (ERCC1, XRCC), inflammation (NFκB), cell proliferation (Ras/MAPK/PI3K/mTORC1), and survival (Bcl-2). Cellular stress conditions like oxidative stress, hypoxia and ER stress activate autophagy to enable cells to adapt to unfavorable conditions.
2.1. Phagophore formation:
Autophagy is initiated by the activation of the Unc-51-like Autophagy Activating kinase 1/2 (ULK 1/2) complex (comprising ULK 1/2 kinases, Atg13, Atg101, and FIP200 proteins) [17]. In low nutrient conditions, the energy senor kinase of 5’ AMP-activated protein kinase (AMPK) senses alterations in intracellular AMP/ATP ratios and activates the ULK 1/2 complex by phosphorylating the serine residues (467, 555, and 638) of ULK-1 while simultaneously inhibiting the autophagy repressor mammalian Target of Rapamycin (mTOR) [18–22]. The phosphorylation of ULK1 triggers the translocation of a multi-protein complex comprising Beclin 1 (or Atg6), Activating molecule in Beclin 1-regulated autophagy (AMBRA), Atg14L, Vps15, and class III phosphoinositide 3-kinase (also known as vacuolar protein sorting 34 (Vps34)) from the cytoplasm to the nascent phagophore [23–25]. Beclin 1 plays a key role in the complex by forming an assembly platform.
2.2. Elongation:
Two ubiquitin-like conjugation systems (Atg5-Atg12 and microtubule-associated protein 1A/1B-light chain 3 [LC3B]) and several soluble NSF attachment protein receptor (SNARE) proteins are required for the subsequent elongation of the phagophore membrane [26, 27]. The Atg5-Atg12 complex is first formed by the action of Atg7 (ubiquitin-E1-like enzyme) and Atg10 (ubiquitin-E2-like enzyme) and then noncovalently conjugated to Atg16 to form the Atg12-Atg5-Atg16 complex, which displays E3 ligase activity [28, 29]. This large complex then forms the autophagosome membrane by recruiting the second conjugation system, the LC3B [30, 31]. Prior to its recruitment, the LC3B protein is cleaved by Atg4 and then lapidated by the addition of a phosphatidyl ethanolamine group through the action of Atg7 and Atg3 [14, 30, 31]. Subsequent phagophore closure is regulated by members of the endosomal sorting complex required for transport (ESCRT), CHMP2A, and VPS4 [32]. Proteins such as p62 (sequestosome-1), NBR1, and NIX that harbor an LC3-interacting region (LIR) also facilitate the recognition of ubiquitylated proteins or specific organelle membranes to be selectively delivered as cargo to the autophagosomes [33].
2.3. Maturation:
The autophagic process is completed with the maturation stage, in which the autophagosomes fuse with the lysosomes to form autolysosomes, leading to their degradation by acid hydrolases. This final step is regulated by members of the SNARE family, including Qa-SNARE, syntaxin 17, Qbc-SNARE, and lysosomal R-SNARE [34]. Autophagy degradation products are then exported from lysosomes into the cytoplasm, where they are recycled for cellular metabolism and repair mechanisms [35].
3. Molecular mechanism of autophagy
Several signaling pathways are known to regulate autophagy in response to cellular and environmental cues. Depletion of nutrients is the most potent physiological inducer of autophagy, in which critical energy sensor pathways such as mTOR and AMPK have been shown to regulate starvation-induced autophagy [36–38]. Additionally, autophagy is induced by extra- and intracellular stresses such as hypoxia, endoplasmic reticulum (ER), and oxidative stress, enabling cells to adapt to or overcome unfavorable conditions [39].
3.1. mTOR pathway:
As an essential signaling molecule that regulates diverse cellular functions, including cell growth and proliferation, mTOR functions as a convergent point for multiple diverse extracellular stimuli (such as growth factors, amino acids, glucose, energy status) and upstream signaling cascades involved in the regulation of autophagy [40, 41]. The three main signaling pathways that regulate mTOR activity are the class I PtdIns3K-protein kinase B (PI3K/AKT), the RAS-proto-oncogene, and the liver kinase B1-AMPK (LKB1/AMPK) pathways [42]. Activation of mTOR negatively regulates autophagy via the action of its two distinct complexes: complex 1 (mTORC1) and 2 (mTORC2), with the former playing a direct role in inhibiting autophagy by phosphorylating the ULK 1/2 complex [19–22, 42, 43]. mTORC2, on the other hand, inhibits autophagy through the phosphorylation and activation of PKB/AKT [44, 45]. Activation of growth factor receptors in nutrient-rich environments stimulates the PI3K complex and small GTPase Ras, leading to the subsequent activation of PI3K/AKT/mTORC1 and Raf-1-MEK1/2-ERK1/2 pathways, respectively. Both PKB/AKT and ERK1/2 phosphorylate and inhibit the GTPase-activating protein complex TSC1/TSC2, resulting in the stabilization of Rheb-GTPase and the activation of mTORC1, which in turn inhibits autophagy [46–49]. In contrast, nutrient depletion triggers autophagy by activating the AMPK pathway, which inhibits mTORC1 activity, causing it to dissociate from the ULK 1/2 complex [19–21, 50].
3.2. AMPK signaling:
Like the mTOR pathway, AMPK also functions as a sensor of cellular bioenergetics and is a well-known autophagy regulator [51, 52]. Studies have shown that at least three independent signaling pathways (LKB1, Ca (2+)/calmodulin-dependent kinase kinase-β [CaMKKβ], and transforming growth factor-β-activating kinase 1 [TAK1]) induce autophagy in an AMPK-dependent manner [53, 54]. AMPK, in turn, initiates autophagy directly by phosphorylating the ULK 1/2 complex and does so indirectly by suppressing mTORC1 activity [50]. Hypoxia is also a potent inducer of AMPK activation, an event independent of HIF activity and that can occur even in the absence of any detectable fluctuations of intracellular ATP levels [55–57]. AMPK’s role in carcinogenesis is controversial, as it can function as either a tumor suppressor or promoter in a context-dependent manner. On one hand, AMPK signaling has been shown to inhibit carcinogenesis by either triggering p53 and FOXO pathways and/or inhibiting AKT/mTOR and nuclear factor-κB (NFκB) pathways [58–60]. It has also been shown to support cancer cell survival by promoting autophagy by stabilizing cytosolic p27 levels or in response to starvation or hypoxic conditions [18, 61].
3.3. Other autophagy signaling pathways:
Autophagy is also regulated by other pathways such as Ras, a membrane-anchored protein, which once activated, stimulates diverse downstream effectors and cellular signaling networks, including class I PI3K/AKT/mTOR1, Raf-1/MEK/ERK, and Rac1/JNK pathways [62]. Ras seemingly plays complex but opposing roles in autophagy regulation. While Ras inhibits autophagy by activating the class I PI3K/AKT/mTOR1 pathway [47, 48], it also promotes autophagy via the activation of Raf-1-MEK1/2-ERK1/2 [49, 50] and Rac1/MKK7/JNK [54, 61] pathways. The former pathway inhibits Bcl-2 binding of Beclin 1, allowing for the formation of the class III PI3K complex, while the latter pathway promotes autophagy by upregulating Atg5/Atg7. Interestingly, Ras-driven tumors have been shown to induce autophagy as an adaptive response to oxidative and metabolic stress while promoting survival [63]. Thus, autophagy induced by Ras can affect tumor progression by modulating cell death, cell proliferation, mitochondrial integrity, and sensitivity to matrix detachment and metabolic stress. Another important pathway that regulates autophagy is the NFκB cascade, which plays a critical role in stress-related cellular inflammation and survival. Both NFκB and autophagy have been shown to regulate one another’s activity in a context-dependent manner. While NFκB can promote autophagy by inducing the expressions of various autophagy-associated proteins such as Beclin 1, it’s signaling can also suppress the expression of autophagic inducers like BNIP3, Jun‐N‐terminal kinase (JNK), and reactive oxygen species (ROS). In contrast, increased autophagy has been shown to inhibit NFκB signaling by degrading IKK subunits (detailed in [64, 65]). Similar to NFκB, the B-cell lymphoma 2 (Bcl-2) protein family also plays a dual role in autophagic regulation. While the oncogenic potential of the Bcl-2 family is attributed mainly to its inhibition of apoptotic signals [detailed in [66]), its emerging role as an autophagy regulator suggests that members such as Bcl-2/Bcl-XL promote carcinogenesis by impairing autophagic function, leading to increased genomic instability and improved survival of cancer cells.
3.4. Oxidative stress:
Excessive ROS production in tissues or cells can induce oxidative stress and lead to oxidative damage such as DNA hydroxylation, protein degeneration, and tissue damage [67]. While the mitochondria are generally accepted as being the main sources of ROS, various intracellular stresses, including nutrition starvation, ER stress, and hypoxia can also induce oxidative stress and damage [68–72]. ROS induces autophagy through various signaling pathways including inhibiting PI3K-AKT-mTOR [67, 73] whilst activating AMPK [74, 75], mitogen activated-protein kinase (MAPK) [76], extracellular signal-regulated kinase (ERK) [77, 78], and JNK [79] signaling. Aside from eliminating ROS via autophagy, cells protect themselves from oxidative stress by promoting the transcription of antioxidant-defense genes. A key transcriptional factor regulating this response is nuclear factor erythroid 2-related factor 2 (Nrf2) [80, 81]. During oxidative stress, Nrf2 is released either through its dissociation from Kelch-like ECH-associated protein 1 (Keap1) or by the upregulation of SQSTM1 (the gene encoding p62). The dissociated Nrf2 then translocates to the nucleus, where it activates the expression of anti-oxidant genes and promotes cell survival [82, 83]. While p62 is degraded during the autophagic process [33], its accumulation in autophagy-impaired cells results in the constitutive activation of Nrf2 and increased production of antioxidant proteins to maintain the redox balance. Moreover, upregulation of p62 activates other pro-survival oncogenic pathways, including NF-κB and mTOR [84]. Oxidative stress and DNA damage have been suggested to play a central role in metal-induced carcinogenesis [85, 86].
3.5. ER stress:
In mammalian cells, the ER is the key compartment that facilitates the folding of newly synthesized proteins and the initiation of vesicular movement of the membrane and proteins to the various organelles and the cell surface. A major ER stress pathway is the unfolded protein response (UPR), which is a potent stimulus of autophagy. ER stress promotes autophagic response via the induction Ca2+ as well as UPR downstream effectors RNA-dependent protein kinase-like ER kinase (PERK) and the inositol-requiring protein‐1α (IRE1α) pathways [51, 87, 88]. Ca2+ released from the ER lumen induces autophagy through activation of the CaMKKβ-AMPK pathways [53, 89] as well as by the death‐associated protein kinase (DAPK)-mediated dissociation of Beclin 1 from the Beclin 1/Bcl‐2 complex [90, 91]. Similarly, IRE1α also promotes autophagy by JNK-mediated phosphorylation of Bcl‐2, resulting in the dissociation of Beclin 1 from Bcl‐2 [92, 93]. PERK, on the other hand, drives autophagy via Activating Transcription Factor 4 (ATF4)’s transcriptional regulation of Atg12, while ATF4-mediated activation of C/EBP‐homologous protein (CHOP) transcriptionally induces Atg5 [88, 94]. ER-stress-induced autophagy can be either pro-survival or pro-death. While many studies indicate that autophagy has a cytoprotective pro‐survival function following ER stress [95, 96], prolonged stress can induce autophagy‐dependent cell death mechanisms and cell damage [97].
3.6. Hypoxia:
Upregulation of autophagy has been observed in hypoxic regions of tumors [43]. Accumulating data show that hypoxia induces autophagy in mammalian cells [98, 99] by both hypoxia-inducible factor-1 (HIF-1)-dependent and independent mechanisms (like AMPK-mTOR, UPR). Considering that hypoxia induces ER stress through UPR and that mitochondria have reduced function in oxidative phosphorylation in hypoxic conditions, the induction of autophagy could allow cells to eliminate portions of compacted ER and reduce mitochondrial mass in periods when no oxygen is available to accept free electrons from the respiratory chain [99]. Hypoxia also increases the transcription of essential autophagy genes LC3 and Atg5 through transcription factors ATF4 and CHOP, respectively, which are in turn regulated by PERK [100].
Carcinogenic metals have been shown to be potent inducers of autophagy; an event likely triggered as a self-defense mechanism against metal-induced oxidative stress [10, 101]. Moreover, cancer cells are known to take advantage of the autophagic process to survive hostile environments. Concurrently, we and others have demonstrated that heavy metals like As and Cd promote tumorigenesis via impaired autophagy [102–106], whereas the subsequent inhibition of autophagy suppressed metal-induced carcinogenesis [105]. Thus, considering the newly emerging role of autophagic response as a key event in metal-induced carcinogenesis, this review will discuss autophagy’s mechanistic role in As-, Cd-, and Cr-induced urogenital carcinogenesis.
4. Metal-induced prostate carcinogenesis
Prostate cancer (CaP) is the most commonly diagnosed cancer and the second leading cause of cancer-related death among men in western countries [107]. Despite identifying several risk factors for CaP, including age, diet, ethnicity, and genetic/family history [108], CaP pathogenesis remains unclear. Several epidemiological studies have suggested a possible link between heavy metal exposure and increased risk of CaP [109–117] (Figure 2)
Figure 2:

Induction of autophagy in metal-induced prostate carcinogenesis. Cd and As have been shown to mediate effects using the same molecular pathways. Both promote carcinogenesis by directly triggering proteins involved in cell proliferation (ERK/MAPK), and by increasing oxidative stress through epigenetic changes or disruption of antioxidant signaling. Increased ROS triggers ER stress and autophagic responses that lead to CaP. Cr also causes excessive ROS production and NFκB activation resulting in cell survival. Albeit limited, studies have shown that autophagy is a key event during the malignant transformation of metal-exposed prostate cells. Dotted lines represent plausible mechanisms involved.
4.1. Cadmium:
Cd is a toxic heavy metal whose presence in nature has become magnified due to human activities. This persistent environmental pollutant is ranked #7 on the 2017 Agency for Toxic Substances and Disease Registry Substance Priority List and has been listed as a significant public health concern by the World Health Organization [118, 119]. Cd is a confirmed carcinogen [120] with a biological half-life >20 years [2, 121, 122]. It is widely found in metal coatings, food, and cigarette smoke [123–129]. The average intake of Cd from food varies between 9 and 25 μg/day in US and Europe and between 19.7 and 35.4 μg/day in Asia [130]. Cigarettes contain approximately 1–2μg of Cd, with about 50% of the inhaled Cd being absorbed by the body [131]. The prostate is one of the organs with the highest levels of Cd deposition [132, 133], and several epidemiological studies have suggested a link between Cd exposure and increased CaP risk/mortality [111, 134–138]. Risk assessment studies suggest that the likelihood of developing CaP is 1.1–2.76 times greater in Cd-exposed cases than in non-exposed cases [135], suggesting Cd as a risk factor for CaP.
Several studies have shown that chronic exposure to low concentrations of Cd-induced malignant transformation of normal human prostate epithelial cells (RWPE-1) [139–141]. These malignant Cd transformed cells, termed Cd-induced Prostate Epithelial cells (CTPE), were, in turn, shown to form aggressive but poorly differentiated adenocarcinomas when inoculated into nude mice [142, 143]. However, the precise molecular mechanisms involved in Cd-mediated carcinogenesis have been only partially elucidated. The complex network of mechanisms involved in Cd-mediated prostate carcinogenesis includes the induction of oxidative stress [144, 145], DNA damage [146, 147], autophagy [105, 106, 148–150], inhibition of antioxidant enzymes such as superoxide dismutase and GSH peroxidase [151, 152], suppression of DNA repair [146, 147, 153], epigenetic changes resulting in hypermethylation of tumor suppressor and apoptosis gene activities [141, 154–156], and enhanced proliferation [157, 158].
Acute or chronic Cd exposure increases ROS levels [120, 159], which, if persistent, induces alterations to the redox signaling pathway, DNA mutations, and changes in methylation and chromatin remodeling patterns [160]. A finding supported by studies demonstrating global hypermethylation and silencing of tumor suppressor genes such as p16 promoter and DNA repair genes ERCC1 and XRCC1 in CTPE cells via the overexpression of DNA methyltransferase 3b (DNMT3b) [141, 161–163]. Moreover, the activation of several signaling cascades, including mitogen-activated PI3K/AKT, NFκB, and p53 inactivation, has been shown to be involved in Cd-induced CaP [139, 164–167]. More recently, Dasgupta et al., [168] reported that enhanced ERK/MAPK signaling is a significant event in the malignant transformation of Cd-induced prostate carcinogenesis.
Our lab group was the first to report that autophagy plays a significant role in Cd-induced prostate carcinogenesis. While attempting to delineate the mechanism of Cd-mediated malignant transformation of prostate epithelial cells, we performed a comparative analysis of global gene arrays of RWPE-1 and CTPE cells and discovered that autophagy-regulated genes (Plac8, LC3B, STX8, STX17, and Lamp-1), particularly placental-specific 8 (Plac8), a precursor of autophagosome-lysosome fusion, were differentially expressed in both cell types [149]. Further examination showed a concomitant increase of both pro-survival proteins (pAKTS473, p65, Bcl-2) and autophagosome/lysosomal proteins (Plac8, Lamp-1, STX8, STX17, and LC3B) in CTPE cells, suggesting that the induction of the pro-survival function of autophagy protected these cells from Cd-induced toxicity. Our finding of the oncogenic role of Plac8 is supported by other studies that have reported a positive correlation between its increased expression and the activation of the pro-survival function of autophagy in other cancers [169–171]. We also found that the inhibition of Plac8-mediated autophagy and pro-survival signaling of NFκB/Bcl2 suppressed CTPE growth [105]. Subsequent delineation of the upstream events responsible for Cd-induced autophagy revealed that Cd-induced ROS, which in turn triggered ER stress and subsequent phosphorylation of downstream stress transducers (PERK, eIF2-α, and ATF4) [106]. Together, these results suggest that induction of ROS and subsequent ER stress are responsible for defective autophagy in Cd-induced prostate carcinogenesis.
4.2. Arsenic:
A naturally occurring metalloid found in food, soil, and water, As inorganic compounds such as As trioxide, sodium arsenite, As trichloride, and arsenates (e.g., calcium arsenate) are classified as class 1 human carcinogen due to their high toxicity and ability to cause multiple cancers including urogenital cancers [172]. As is a known pollutant, and its concentrations ranging from 1 ng/m3 to 2–3.6 mg/L have been reported in groundwater worldwide [160]. Drinking water, crops irrigated with contaminated water, and food are the main sources of As exposure [160]. A causal link between As and CaP has been described in several epidemiological studies in human populations exposed to high concentrations of As (reported range between>10 ppb and 2500 ppb) in drinking water [109, 112, 173–175].
Studies have shown that chronic exposure to low doses of As induced malignant transformation of RWPE-1 cells [176, 177], and subsequent in vivo experiments found that these transformed cells formed aggressive carcinomas exhibiting characteristics commonly found in human prostatic cancers [178]. As has been shown to induce oxidative stress by increasing intracellular ROS levels [179] and inhibiting glutathione (GSH) antioxidant activity [180], resulting in oxidative DNA damage [181–184]. Moreover, epigenetic changes and altered signaling that play important roles in cell proliferation, differentiation, and transformation have been reported in As-mediated malignant transformation of prostate cells [141, 185–188]. Although the role of autophagy in As-mediated prostate carcinogenesis has not yet been studied, autophagy has been shown to confer therapeutic resistance and enhance the invasive ability of CaP cells [189]. Moreover, the use of autophagy inhibitors in combination with therapeutic agents was found to suppress the proliferation of resistant CaP cells and promote cell death [190–192]. Based on this evidence, it is clear that autophagy plays a significant role in CaP pathogenesis and that more studies are needed to explore its role in metal-induced prostate carcinogenesis.
4.3. Chromium:
Cr toxicity is caused primarily by its hexavalent form, Cr(VI), and is classified as a Group 1 carcinogen by the IARC. Cr(VI)’s environmental presence is almost entirely industrial in origin [1], and many epidemiological studies have reported its carcinogenic effect in several organs, including the prostate [193]. Cr(VI)-induced epigenetic changes, genotoxicity, and oxidative DNA damage have been implicated as mechanisms through which it mediates its carcinogenicity [7, 116, 194]. Cr(VI) exposure has also been shown to induce ROS production, which, in turn, has been shown to activate NFκB and AP-1 signaling pathways [186, 187, 195, 196]. In CaP, Cr(VI) exposure was reported to induce ROS and increase HIF-1α and VEGF levels via activation of the p38 MAP kinase pathway [197]. However, despite being well studied in other cancers such as that of the lung, Cr’s carcinogenic role in CaP remains underexplored. Considering that studies have shown the activity of many upstream effectors of autophagy, it is possible that Cr(VI) may mediate some of its carcinogenic effects via defective autophagy. However, more studies are needed in this area.
5. Metal-induced bladder carcinogenesis
Bladder cancer is the fourth most prevalent cancer among men; while it can occur in women, its overall incidence is atleast three to four times lower [107]. Among the heavy metals discussed in this review, As is considered the most potent bladder carcinogen, followed by Cd and hexavalent Cr. Despite metal-induced carcinogenesis being documented in several epidemiological studies, the exact mechanisms underlying metal-induced bladder carcinogenesis have yet to be explored. Mechanistic insights suggest that autophagy may play an important role in bladder carcinogenesis (Figure 3).
Figure 3:

Metal-induced autophagy-mediated bladder carcinogenesis. As exposure induces GSH mediated superoxide formation that affects several pathways, including Nrf2-Keap1 and ATF4-mediated UPR. Inhibition of mTORC1 or activation of ULK1 and Atg induced autophagy resulting in the malignant transformation of bladder cells. Cd exposure increased Nrf2 binding to the ARE promoter in p62, causing an increase in antioxidants and concomitant ROS inhibition ultimately leading to carcinogenesis of bladder epithelial cells. Similarly, Cr-exposure dysregulated the p62 mediated Nrf2-Keap1 pathway and promoted constitutive activation of Nrf2.
5.1. Arsenic:
Although As carcinogenicity is associated mainly with skin and lung cancers [198, 199], its accumulation in the bladder makes it a potent carcinogen for the onset of bladder carcinogenesis [200]. The trivalent form of As (iAs3+) is known to induce ROS (O2−, H2O2) [201] and, consequently, the Nrf2-Keap1 pathway. In fact, As exposure in UROtsa cells has been shown to activate Nrf2 signaling [202], with the subsequent dysregulation of the Nrf2-Keap1 axis being shown to induce malignant transformation of normal bladder epithelial cells and increased cell proliferation [203]. As exposure also induces UPR and the activation of ATF4 signaling. While oxidative stress has been implicated as the major driver of malignant cellular transformation [204], it should be noted that the activation of autophagy in response to persistent oxidative stress maybe how the transforming cells survive and proliferate in these stressed conditions. As-induced ROS activation has been shown to activate AMPK signaling [205] resulting in the dephosphorylation of mTORC1 and activation of ULK1-induced protective autophagy in bladder cancer cells [206]. As has also been shown to bind to cysteine 299 of AMPKα directly [207]. Key autophagic proteins such as Beclin 1 and Atg7 protein have also been shown to be predominately expressed in high-grade bladder cancers. Moreover, modest alterations of autophagic proteins such as Beclin 1, Atg-5, −7, −12 and LC3B have been shown in As-exposed UROtsa cells [208]. Taken together, these studies suggest that ROS-mediated autophagy activation may be a key event cell survival during As-induced malignant transformation of normal bladder epithelial cells.
5.2. Cadmium:
Epidemiologic studies suggest that Cd is also a potent carcinogen for bladder cancer [209–212]. Chronic exposure to low doses of Cd has been shown to cause UROtsa cells to acquire clonogenic abilities and develop tumors in mice models [213]. Moreover, another study demonstrated that Cd exposure induces autophagy proteins’ expression, including Beclin 1 in Cd-induced malignant transformed bladder epithelial cells [208]. Similar to As-induced carcinogenesis, Cd-induced oncogenic transformation increases ROS production in cells, which in turn causes the constitutive activation of Nrf2 [214]. Cd exposure enhances the binding of Nrf2 to the antioxidant response element (ARE) promoter regions of p62/Bcl-2/Bcl-XL. Cd exposure similar to that of As, causes epigenetic alterations resulting in impaired DNA repair mechanisms and subsequent carcinogenesis. Cd-induced activation of transcription factors such as Metal Regulatory Transcription Factor 1, upstream regulator of autophagy, has also been implicated as another mechanism through which this metal may drive bladder carcinogenesis [215]. Although new information pertaining Cd-induced carcinogenicity is rapidly evolving, more comprehensive studies are still needed to elucidate the roles of the various molecular mechanisms especially those related to autophagic activation in Cd-mediated bladder carcinogenesis.
5.3. Chromium:
Epidemiological evidence suggests that occupational exposure to Cr causes bladder cancer [216–219]. Kutze et al., [220] in their study, found a significantly increased probability of lower urinary tract urothelial tumors in patients occupationally exposed to Cr/chromate. Similarly, another study reported significantly higher Cr concentration in the bladder cancer tissues (99.632 ng/g) as compared to non-cancer tissue (33.144ng/g) [216–218]. Chronic Cr(VI) exposure was shown to induce chromosomal damage in bladder cells, which in turn was suggested as a possible mechanism for Cr-induced bladder carcinogenesis [221]. Cr has been linked to the reduced expressions of AMPK and peroxisome proliferator-activated receptor γ coactivator 1α, which resulted in the disruption of mitochondrial dynamics through the dysregulation of the Nrf2 axis [222]. Cr exposure has also been shown to induce autophagy through the activation of ROS-AKT-mTOR pathway [223, 224]. In another study, Yang et al., showed that trivalent Cr, Cr(III), induced autophagy by activating sphingomyelin phosphodiesterase 2 (SMPD2). SMPD2 increases levels of ceramide and ceramide signals overlapped with LC3, suggesting that ceramide might play an important role in the formation of autophagosomes. The study concluded that Cr induces autophagy via structural aberration of the organelle membrane, in particular by the increase of lipid compositions in addition to autophagy-associated proteins [103]. Although studies have shown potential Cr-induced oncogenic mechanisms involved in its promotion of bladder carcinogenesis, there is still a dearth of knowledge regarding the role of autophagy in Cr-mediated bladder carcinogenesis that requires further elucidation.
6. Metal-induced kidney carcinogenesis
Kidney cancer is the sixth and eighth most common cancer among men and women, respectively [107]. The kidney has long been recognized as one of the organs most affected by high metal exposure.
6.1. Cadmium:
Approximately 50% of Cd concentrations in the body is accumulated in the kidney [225]. Because of Cd’s low molecular mass (~7Kd), it is filtered through to proximal tubules, which are the parts most affected by Cd-related toxicity [226]. The free and metallothionein-bound Cd (Cd-MT) is taken up by receptors for small protein ligands such as megalin and cubilin [227]. Once internalized, Cd has been shown to induce autophagy signaling by mTORC1 downregulation and upregulating Atg5, Beclin 1, and LC3 in duck renal tubular epithelial cells [228], cultured mesangial cells [229], rat proximal tubule [230], and NRK-52E cells [231]. Studies have shown that an increase in LC3B expression coupled with a concomitant decrease in the expression of tumor suppressor miR-204, placed a critical role in the progression of renal cell carcinoma (RCC) [232]. Furthermore, autophagy defects caused by decreased microtubule-associated protein family 1 signaling [233] and deregulation of Keap1-Nrf2 axis [234] play an important role in promoting tumor cell survival in RCC. Although these studies present a link between Cd-induced autophagy and survival, the molecular interaction between these pathways, especially in the context of renal cancer, remains unclear. Future studies are urgently needed to amplify the current knowledge regarding the role of autophagy in Cd-induced renal carcinogenesis
6.2. Arsenic:
As has been associated with causing structural damage and renal dysfunction, resulting in nephrotoxicity and subsequent carcinogenesis [235–237]. Although kidney cancer generally occurs in older individuals, studies have shown that exposure to inorganic As increases the incidence of renal cancer among young adults [238]. Moreover, increased oxidative stress has been linked to elevated urine As levels and decreased renal function [239]. As exposure has been shown to induce significant ROS production and DNA damage, and autophagy in the kidney [240]. Studies have shown that As exposure induced p62 expression, which activates LC3B in the rat kidney, and the accumulation of autophagosome disrupted kidney ultrastructure [241]. While chronic As exposure has been shown to induce kidney carcinogenesis through sustained oxidative stress and autophagy signaling, more experimental studies are warranted to better establish the molecular mechanisms underlaying As-induced carcinogenesis.
6.3. Chromium:
Cr exposure has been strongly associated with increasing the mortality risk among patients with kidney cancer [242]. Decreased activity of glutathione reductase in the kidneys of rats exposed to Cr, has been suggested to be the result of Cr-induced oxidative stress. Moreover, Cr-transformed cells show constitutive expression of HIF-1α, which is linked to cellular survival by inducing mitophagy in clear cell renal cell carcinoma [243]. Studies have demonstrated that ROS and autophagy in Cr-exposed cells are closely linked and could play an important role in Cr-mediated malignant transformation of kidney cells. Considering that Cr induces activation of HIF-1α, Nrf2, and p62 pathways [10], which each play significant roles in autophagy activation, more studies are needed to delineate autophagy’s role in Cr-induced renal carcinogenesis.
7. Conclusions and future perspectives
Autophagy is considered to function as a double-edged sword in cancer cells due to its elusive switch from tumor suppressor to tumor promoter. Because cancer cells are known to exploit autophagy as an adaptive stress response to survive hostile environments, the recent decade has seen a resurgence of interest in developing autophagy inhibitors as a therapeutic strategy to combat refractory cancers. On the other hand, despite evidence from various experimental and epidemiological studies, the occurrence of metal-induced carcinogenesis is still a controversial topic among researchers. A factor that has greatly hampered progress in understanding the etiology of metal-induced carcinogenesis has been the lack of established in vivo models of the same, despite the availability of established in vitro models. Reasons for this include but are not limited to physiological and anatomical differences between humans and mice, making it hard to accurately mimic the carcinogenic effect of metals. Moreover, in real-world settings, humans are exposed to different concentrations of multiple metals simultaneously via various routes (drinking water, diet, smoking, and occupational), suggesting that metal-induced carcinogenesis may be a result of multiple hits. However, this aspect has yet to be mimicked in mice models where metal exposure is limited to one carcinogen and one exposure route. Moreover, the role of autophagy in metal-induced carcinogenesis is limited in urogenital cancers, though considering that the majority of experimental studies have shown a strong role of autophagy, it is essential to explore this avenue in the quest for therapeutic targets. Aside from establishing in vivo models that enable researchers to observe the carcinogenic effects of multiple metals through multiple exposure routes, there is a need to determine metal carcinogenesis using established autophagy mouse models.
Moving forward, there is a need to extend the current knowledge of metal-induced carcinogenesis from in vitro settings to in vivo settings, and compare these results to results acquired from human clinical specimens and epidemiological studies. Combining evidence from these fronts will enable us to acquire a complete picture of the etiology and pathology of metal-induced carcinogenesis.
Acknowledgements
The authors regret that they were unable to include all references pertaining to metal carcinogenesis in this review due to page limitations, but would nonetheless like to acknowledge contributions of all researchers in the metal carcinogenesis field. This work was supported by NIEHS-R01ES028102, R01ES030019 to CD.
Abbreviations:
- Atg
Autophagy related genes
- ATF4
Activating transcription factor 4
- Cd-MT
metallothionein-bound Cd
- CTPE
Cd-induced prostate epithelial cells
- HIF-1
Hypoxia inducible factor 1
- JNK
c-Jun N-terminal kinase
- Keap1
Kelch-like ECH-associated protein
- LC3B
microtubule associated protein 1A/1B light chain 3
- MTF1
Metal Regulatory Transcription Factor 1
- Nrf2
Nuclear factor erythroid 2- related factor 2
- p62
Sequestosome-1
- ROS
Reactive oxygen species
- SNAREs
Soluble NSF attachment protein receptors
- ULK1/2
Unc-51like autophagy activating kinase 1/2
- UPR
Unfolded protein response
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest Statement: The authors declare that there are no conflicts of interest.
References:
- [1].Zhu Y, Costa M, Metals and molecular carcinogenesis, Carcinogenesis 41(9) (2020) 1161–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Chen QY, DesMarais T, Costa M, Metals and Mechanisms of Carcinogenesis, Annu Rev Pharmacol Toxicol 59 (2019) 537–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Yang M, A current global view of environmental and occupational cancers, J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 29(3) (2011) 223–49. [DOI] [PubMed] [Google Scholar]
- [4].Hopenhayn-Rich C, Biggs ML, Smith AH, Lung and kidney cancer mortality associated with arsenic in drinking water in Cordoba, Argentina, Int J Epidemiol 27(4) (1998) 561–9. [DOI] [PubMed] [Google Scholar]
- [5].Smith AH, Hopenhayn-Rich C, Bates MN, Goeden HM, Hertz-Picciotto I, Duggan HM, Wood R, Kosnett MJ, Smith MT, Cancer risks from arsenic in drinking water, Environ Health Perspect 97 (1992) 259–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Wild P, Bourgkard E, Paris C, Lung cancer and exposure to metals: the epidemiological evidence, Methods Mol Biol 472 (2009) 139–67. [DOI] [PubMed] [Google Scholar]
- [7].Arita A, Costa M, Epigenetics in metal carcinogenesis: nickel, arsenic, chromium and cadmium, Metallomics 1(3) (2009) 222–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Xu J, Wise JTF, Wang L, Schumann K, Zhang Z, Shi X, Dual Roles of Oxidative Stress in Metal Carcinogenesis, J Environ Pathol Toxicol Oncol 36(4) (2017) 345–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Martinez-Zamudio R, Ha HC, Environmental epigenetics in metal exposure, Epigenetics 6(7) (2011) 820–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Wang L, Wise JT, Zhang Z, Shi X, Progress and prospects of reactive oxygen species in metal carcinogenesis, Curr Pharmacol Rep 2(4) (2016) 178–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Yuan Y, Marshall G, Ferreccio C, Steinmaus C, Selvin S, Liaw J, Bates MN, Smith AH, Acute myocardial infarction mortality in comparison with lung and bladder cancer mortality in arsenic-exposed region II of Chile from 1950 to 2000, Am J Epidemiol 166(12) (2007) 1381–91. [DOI] [PubMed] [Google Scholar]
- [12].Galanis A, Karapetsas A, Sandaltzopoulos R, Metal-induced carcinogenesis, oxidative stress and hypoxia signalling, Mutat Res 674(1–2) (2009) 31–5. [DOI] [PubMed] [Google Scholar]
- [13].Chavez-Dominguez R, Perez-Medina M, Lopez-Gonzalez JS, Galicia-Velasco M, Aguilar-Cazares D, The Double-Edge Sword of Autophagy in Cancer: From Tumor Suppression to Pro-tumor Activity, Front Oncol 10 (2020) 578418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Kroemer G, Marino G, Levine B, Autophagy and the integrated stress response, Mol Cell 40(2) (2010) 280–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Green DR, Llambi F, Cell Death Signaling, Cold Spring Harb Perspect Biol 7(12) (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Bishop E, Bradshaw TD, Autophagy modulation: a prudent approach in cancer treatment?, Cancer Chemother Pharmacol 82(6) (2018) 913–922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Mercer CA, Kaliappan A, Dennis PB, A novel, human Atg13 binding protein, Atg101, interacts with ULK1 and is essential for macroautophagy, Autophagy 5(5) (2009) 649–62. [DOI] [PubMed] [Google Scholar]
- [18].Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor R, Asara JM, Fitzpatrick J, Dillin A, Viollet B, Kundu M, Hansen M, Shaw RJ, Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy, Science 331(6016) (2011) 456–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Ganley IG, Lam du H, Wang J, Ding X, Chen S, Jiang X, ULK1.ATG13.FIP200 complex mediates mTOR signaling and is essential for autophagy, J Biol Chem 284(18) (2009) 12297–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Hosokawa N, Hara T, Kaizuka T, Kishi C, Takamura A, Miura Y, Iemura S, Natsume T, Takehana K, Yamada N, Guan JL, Oshiro N, Mizushima N, Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy, Molecular biology of the cell 20(7) (2009) 1981–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Jung CH, Jun CB, Ro SH, Kim YM, Otto NM, Cao J, Kundu M, Kim DH, ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery, Molecular biology of the cell 20(7) (2009) 1992–2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Kim J, Kundu M, Viollet B, Guan KL, AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1, Nat Cell Biol 13(2) (2011) 132–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Kawamata T, Kamada Y, Kabeya Y, Sekito T, Ohsumi Y, Organization of the pre-autophagosomal structure responsible for autophagosome formation, Molecular biology of the cell 19(5) (2008) 2039–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Fimia GM, Di Bartolomeo S, Piacentini M, Cecconi F, Unleashing the Ambra1-Beclin 1 complex from dynein chains: Ulk1 sets Ambra1 free to induce autophagy, Autophagy 7(1) (2011) 115–7. [DOI] [PubMed] [Google Scholar]
- [25].Yuan HX, Russell RC, Guan KL, Regulation of PIK3C3/VPS34 complexes by MTOR in nutrient stress-induced autophagy, Autophagy 9(12) (2013) 1983–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Ohsumi Y, Molecular mechanism of autophagy in yeast, Saccharomyces cerevisiae, Philos Trans R Soc Lond B Biol Sci 354(1389) (1999) 1577–80; discussion 1580–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Moreau K, Ravikumar B, Renna M, Puri C, Rubinsztein DC, Autophagosome precursor maturation requires homotypic fusion, Cell 146(2) (2011) 303–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Romanov J, Walczak M, Ibiricu I, Schuchner S, Ogris E, Kraft C, Martens S, Mechanism and functions of membrane binding by the Atg5-Atg12/Atg16 complex during autophagosome formation, EMBO J 31(22) (2012) 4304–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Camuzard O, Santucci-Darmanin S, Carle GF, Pierrefite-Carle V, Autophagy in the crosstalk between tumor and microenvironment, Cancer Lett 490 (2020) 143–153. [DOI] [PubMed] [Google Scholar]
- [30].Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, Kominami E, Ohsumi Y, Yoshimori T, LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing, EMBO J 19(21) (2000) 5720–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Kabeya Y, Mizushima N, Yamamoto A, Oshitani-Okamoto S, Ohsumi Y, Yoshimori T, LC3, GABARAP and GATE16 localize to autophagosomal membrane depending on form-II formation, J Cell Sci 117(Pt 13) (2004) 2805–12. [DOI] [PubMed] [Google Scholar]
- [32].Takahashi Y, He H, Tang Z, Hattori T, Liu Y, Young MM, Serfass JM, Chen L, Gebru M, Chen C, Wills CA, Atkinson JM, Chen H, Abraham T, Wang HG, An autophagy assay reveals the ESCRT-III component CHMP2A as a regulator of phagophore closure, Nat Commun 9(1) (2018) 2855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Overvatn A, Bjorkoy G, Johansen T, p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy, J Biol Chem 282(33) (2007) 24131–45. [DOI] [PubMed] [Google Scholar]
- [34].Fader CM, Sanchez DG, Mestre MB, Colombo MI, TI-VAMP/VAMP7 and VAMP3/cellubrevin: two v-SNARE proteins involved in specific steps of the autophagy/multivesicular body pathways, Biochim Biophys Acta 1793(12) (2009) 1901–16. [DOI] [PubMed] [Google Scholar]
- [35].Reggiori F, Ungermann C, Autophagosome Maturation and Fusion, J Mol Biol 429(4) (2017) 486–496. [DOI] [PubMed] [Google Scholar]
- [36].Takeshige K, Baba M, Tsuboi S, Noda T, Ohsumi Y, Autophagy in yeast demonstrated with proteinase-deficient mutants and conditions for its induction, J Cell Biol 119(2) (1992) 301–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Srinivas V, Bohensky J, Zahm AM, Shapiro IM, Autophagy in mineralizing tissues: microenvironmental perspectives, Cell Cycle 8(3) (2009) 391–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Neufeld TP, TOR-dependent control of autophagy: biting the hand that feeds, Curr Opin Cell Biol 22(2) (2010) 157–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Kondo Y, Kanzawa T, Sawaya R, Kondo S, The role of autophagy in cancer development and response to therapy, Nature reviews. Cancer 5(9) (2005) 726–34. [DOI] [PubMed] [Google Scholar]
- [40].Wu YT, Tan HL, Huang Q, Ong CN, Shen HM, Activation of the PI3K-Akt-mTOR signaling pathway promotes necrotic cell death via suppression of autophagy, Autophagy 5(6) (2009) 824–34. [DOI] [PubMed] [Google Scholar]
- [41].Wu YT, Tan HL, Shui G, Bauvy C, Huang Q, Wenk MR, Ong CN, Codogno P, Shen HM, Dual role of 3-methyladenine in modulation of autophagy via different temporal patterns of inhibition on class I and III phosphoinositide 3-kinase, J Biol Chem 285(14) (2010) 10850–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Wang RC, Wei Y, An Z, Zou Z, Xiao G, Bhagat G, White M, Reichelt J, Levine B, Akt-mediated regulation of autophagy and tumorigenesis through Beclin 1 phosphorylation, Science 338(6109) (2012) 956–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, Mukherjee C, Shi Y, Gelinas C, Fan Y, Nelson DA, Jin S, White E, Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis, Cancer Cell 10(1) (2006) 51–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Sarbassov DD, Guertin DA, Ali SM, Sabatini DM, Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex, Science 307(5712) (2005) 1098–101. [DOI] [PubMed] [Google Scholar]
- [45].Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Del Piccolo P, Burden SJ, Di Lisi R, Sandri C, Zhao J, Goldberg AL, Schiaffino S, Sandri M, FoxO3 controls autophagy in skeletal muscle in vivo, Cell Metab 6(6) (2007) 458–71. [DOI] [PubMed] [Google Scholar]
- [46].Ma L, Chen Z, Erdjument-Bromage H, Tempst P, Pandolfi PP, Phosphorylation and functional inactivation of TSC2 by Erk implications for tuberous sclerosis and cancer pathogenesis, Cell 121(2) (2005) 179–93. [DOI] [PubMed] [Google Scholar]
- [47].Furuta S, Hidaka E, Ogata A, Yokota S, Kamata T, Ras is involved in the negative control of autophagy through the class I PI3-kinase, Oncogene 23(22) (2004) 3898–904. [DOI] [PubMed] [Google Scholar]
- [48].Inoki K, Li Y, Zhu T, Wu J, Guan KL, TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling, Nat Cell Biol 4(9) (2002) 648–57. [DOI] [PubMed] [Google Scholar]
- [49].Pattingre S, Bauvy C, Codogno P, Amino acids interfere with the ERK1/2-dependent control of macroautophagy by controlling the activation of Raf-1 in human colon cancer HT-29 cells, J Biol Chem 278(19) (2003) 16667–74. [DOI] [PubMed] [Google Scholar]
- [50].Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, Turk BE, Shaw RJ, AMPK phosphorylation of raptor mediates a metabolic checkpoint, Mol Cell 30(2) (2008) 214–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].He C, Klionsky DJ, Regulation mechanisms and signaling pathways of autophagy, Annu Rev Genet 43 (2009) 67–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Yang Z, Klionsky DJ, Mammalian autophagy: core molecular machinery and signaling regulation, Curr Opin Cell Biol 22(2) (2010) 124–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Hoyer-Hansen M, Bastholm L, Szyniarowski P, Campanella M, Szabadkai G, Farkas T, Bianchi K, Fehrenbacher N, Elling F, Rizzuto R, Mathiasen IS, Jaattela M, Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-beta, and Bcl-2, Mol Cell 25(2) (2007) 193–205. [DOI] [PubMed] [Google Scholar]
- [54].Herrero-Martin G, Hoyer-Hansen M, Garcia-Garcia C, Fumarola C, Farkas T, Lopez-Rivas A, Jaattela M, TAK1 activates AMPK-dependent cytoprotective autophagy in TRAIL-treated epithelial cells, EMBO J 28(6) (2009) 677–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Papandreou I, Lim AL, Laderoute K, Denko NC, Hypoxia signals autophagy in tumor cells via AMPK activity, independent of HIF-1, BNIP3, and BNIP3L, Cell Death Differ 15(10) (2008) 1572–81. [DOI] [PubMed] [Google Scholar]
- [56].Liu L, Cash TP, Jones RG, Keith B, Thompson CB, Simon MC, Hypoxia-induced energy stress regulates mRNA translation and cell growth, Mol Cell 21(4) (2006) 521–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Laderoute KR, Amin K, Calaoagan JM, Knapp M, Le T, Orduna J, Foretz M, Viollet B, 5’-AMP-activated protein kinase (AMPK) is induced by low-oxygen and glucose deprivation conditions found in solid-tumor microenvironments, Mol Cell Biol 26(14) (2006) 5336–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Inoki K, Zhu T, Guan KL, TSC2 mediates cellular energy response to control cell growth and survival, Cell 115(5) (2003) 577–90. [DOI] [PubMed] [Google Scholar]
- [59].Jones RG, Plas DR, Kubek S, Buzzai M, Mu J, Xu Y, Birnbaum MJ, Thompson CB, AMP-activated protein kinase induces a p53-dependent metabolic checkpoint, Mol Cell 18(3) (2005) 283–93. [DOI] [PubMed] [Google Scholar]
- [60].Salminen A, Hyttinen JM, Kaarniranta K, AMP-activated protein kinase inhibits NF-kappaB signaling and inflammation: impact on healthspan and lifespan, J Mol Med (Berl) 89(7) (2011) 667–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Liang J, Shao SH, Xu ZX, Hennessy B, Ding Z, Larrea M, Kondo S, Dumont DJ, Gutterman JU, Walker CL, Slingerland JM, Mills GB, The energy sensing LKB1-AMPK pathway regulates p27(kip1) phosphorylation mediating the decision to enter autophagy or apoptosis, Nat Cell Biol 9(2) (2007) 218–24. [DOI] [PubMed] [Google Scholar]
- [62].Downward J, Targeting RAS signalling pathways in cancer therapy, Nature reviews. Cancer 3(1) (2003) 11–22. [DOI] [PubMed] [Google Scholar]
- [63].Kowalski M, [Congenital crural pseudarthrosis in a hemophiliac], Acta Haematol Pol 21(2) (1990) 231–4. [PubMed] [Google Scholar]
- [64].Baldwin AS, Regulation of cell death and autophagy by IKK and NF-kappaB: critical mechanisms in immune function and cancer, Immunol Rev 246(1) (2012) 327–45. [DOI] [PubMed] [Google Scholar]
- [65].Salminen A, Hyttinen JM, Kauppinen A, Kaarniranta K, Context-Dependent Regulation of Autophagy by IKK-NF-kappaB Signaling: Impact on the Aging Process, Int J Cell Biol 2012 (2012) 849541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Adams JM, Cory S, The Bcl-2 apoptotic switch in cancer development and therapy, Oncogene 26(9) (2007) 1324–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Gao Q, Oxidative Stress and Autophagy, Adv Exp Med Biol 1206 (2019) 179–198. [DOI] [PubMed] [Google Scholar]
- [68].Terlecky SR, Koepke JI, Drug delivery to peroxisomes: employing unique trafficking mechanisms to target protein therapeutics, Adv Drug Deliv Rev 59(8) (2007) 739–47. [DOI] [PubMed] [Google Scholar]
- [69].Finkel T, Signal transduction by mitochondrial oxidants, J Biol Chem 287(7) (2012) 4434–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].de Brito OM, Scorrano L, Mitofusin 2 tethers endoplasmic reticulum to mitochondria, Nature 456(7222) (2008) 605–10. [DOI] [PubMed] [Google Scholar]
- [71].Handy DE, Loscalzo J, Redox regulation of mitochondrial function, Antioxid Redox Signal 16(11) (2012) 1323–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Vasko R, Goligorsky MS, Dysfunctional lysosomal autophagy leads to peroxisomal oxidative burnout and damage during endotoxin-induced stress, Autophagy 9(3) (2013) 442–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Portal-Nunez S, Esbrit P, Alcaraz MJ, Largo R, Oxidative stress, autophagy, epigenetic changes and regulation by miRNAs as potential therapeutic targets in osteoarthritis, Biochem Pharmacol 108 (2016) 1–10. [DOI] [PubMed] [Google Scholar]
- [74].Shiomi M, Miyamae M, Takemura G, Kaneda K, Inamura Y, Onishi A, Koshinuma S, Momota Y, Minami T, Figueredo VM, Sevoflurane induces cardioprotection through reactive oxygen species-mediated upregulation of autophagy in isolated guinea pig hearts, J Anesth 28(4) (2014) 593–600. [DOI] [PubMed] [Google Scholar]
- [75].Hart PC, Mao M, de Abreu AL, Ansenberger-Fricano K, Ekoue DN, Ganini D, Kajdacsy-Balla A, Diamond AM, Minshall RD, Consolaro ME, Santos JH, Bonini MG, MnSOD upregulation sustains the Warburg effect via mitochondrial ROS and AMPK-dependent signalling in cancer, Nat Commun 6 (2015) 6053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Ito K, Hirao A, Arai F, Takubo K, Matsuoka S, Miyamoto K, Ohmura M, Naka K, Hosokawa K, Ikeda Y, Suda T, Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells, Nat Med 12(4) (2006) 446–51. [DOI] [PubMed] [Google Scholar]
- [77].Kano G, Almanan M, Bochner BS, Zimmermann N, Mechanism of Siglec-8-mediated cell death in IL-5-activated eosinophils: role for reactive oxygen species-enhanced MEK/ERK activation, J Allergy Clin Immunol 132(2) (2013) 437–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Zhang Y, Choksi S, Chen K, Pobezinskaya Y, Linnoila I, Liu ZG, ROS play a critical role in the differentiation of alternatively activated macrophages and the occurrence of tumor-associated macrophages, Cell Res 23(7) (2013) 898–914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Tang HW, Liao HM, Peng WH, Lin HR, Chen CH, Chen GC, Atg9 interacts with dTRAF2/TRAF6 to regulate oxidative stress-induced JNK activation and autophagy induction, Dev Cell 27(5) (2013) 489–503. [DOI] [PubMed] [Google Scholar]
- [80].Kobayashi M, Yamamoto M, Nrf2-Keap1 regulation of cellular defense mechanisms against electrophiles and reactive oxygen species, Adv Enzyme Regul 46 (2006) 113–40. [DOI] [PubMed] [Google Scholar]
- [81].Chan K, Han XD, Kan YW, An important function of Nrf2 in combating oxidative stress: detoxification of acetaminophen, Proc Natl Acad Sci U S A 98(8) (2001) 4611–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Villeneuve NF, Lau A, Zhang DD, Regulation of the Nrf2-Keap1 antioxidant response by the ubiquitin proteasome system: an insight into cullin-ring ubiquitin ligases, Antioxid Redox Signal 13(11) (2010) 1699–712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Komatsu M, Kurokawa H, Waguri S, Taguchi K, Kobayashi A, Ichimura Y, Sou YS, Ueno I, Sakamoto A, Tong KI, Kim M, Nishito Y, Iemura S, Natsume T, Ueno T, Kominami E, Motohashi H, Tanaka K, Yamamoto M, The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1, Nat Cell Biol 12(3) (2010) 213–23. [DOI] [PubMed] [Google Scholar]
- [84].Moscat J, Diaz-Meco MT, p62: a versatile multitasker takes on cancer, Trends Biochem Sci 37(6) (2012) 230–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Leonard SS, Bower JJ, Shi X, Metal-induced toxicity, carcinogenesis, mechanisms and cellular responses, Mol Cell Biochem 255(1–2) (2004) 3–10. [DOI] [PubMed] [Google Scholar]
- [86].Ercal N, Gurer-Orhan H, Aykin-Burns N, Toxic metals and oxidative stress part I: mechanisms involved in metal-induced oxidative damage, Curr Top Med Chem 1(6) (2001) 529–39. [DOI] [PubMed] [Google Scholar]
- [87].Smith M, Wilkinson S, ER homeostasis and autophagy, Essays Biochem 61(6) (2017) 625–635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Song S, Tan J, Miao Y, Zhang Q, Crosstalk of ER stress-mediated autophagy and ER-phagy: Involvement of UPR and the core autophagy machinery, J Cell Physiol 233(5) (2018) 3867–3874. [DOI] [PubMed] [Google Scholar]
- [89].Borodkina AV, Shatrova AN, Deryabin PI, Griukova AA, Abushik PA, Antonov SM, Nikolsky NN, Burova EB, Calcium alterations signal either to senescence or to autophagy induction in stem cells upon oxidative stress, Aging (Albany NY) 8(12) (2016) 3400–3418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Simon B, Huart AS, Temmerman K, Vahokoski J, Mertens HD, Komadina D, Hoffmann JE, Yumerefendi H, Svergun DI, Kursula P, Schultz C, McCarthy AA, Hart DJ, Wilmanns M, Death-Associated Protein Kinase Activity Is Regulated by Coupled Calcium/Calmodulin Binding to Two Distinct Sites, Structure 24(6) (2016) 851–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Zalckvar E, Berissi H, Eisenstein M, Kimchi A, Phosphorylation of Beclin 1 by DAP-kinase promotes autophagy by weakening its interactions with Bcl-2 and Bcl-XL, Autophagy 5(5) (2009) 720–2. [DOI] [PubMed] [Google Scholar]
- [92].Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD, Levine B, Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy, Cell 122(6) (2005) 927–39. [DOI] [PubMed] [Google Scholar]
- [93].Yamamoto K, Ichijo H, Korsmeyer SJ, BCL-2 is phosphorylated and inactivated by an ASK1/Jun N-terminal protein kinase pathway normally activated at G(2)/M, Mol Cell Biol 19(12) (1999) 8469–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Song S, Tan J, Miao Y, Li M, Zhang Q, Crosstalk of autophagy and apoptosis: Involvement of the dual role of autophagy under ER stress, J Cell Physiol 232(11) (2017) 2977–2984. [DOI] [PubMed] [Google Scholar]
- [95].Cheng YC, Chang JM, Chen CA, Chen HC, Autophagy modulates endoplasmic reticulum stress-induced cell death in podocytes: a protective role, Exp Biol Med (Maywood) 240(4) (2015) 467–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Ma XH, Piao SF, Dey S, McAfee Q, Karakousis G, Villanueva J, Hart LS, Levi S, Hu J, Zhang G, Lazova R, Klump V, Pawelek JM, Xu X, Xu W, Schuchter LM, Davies MA, Herlyn M, Winkler J, Koumenis C, Amaravadi RK, Targeting ER stress-induced autophagy overcomes BRAF inhibitor resistance in melanoma, J Clin Invest 124(3) (2014) 1406–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Rubiolo JA, Lopez-Alonso H, Martinez P, Millan A, Cagide E, Vieytes MR, Vega FV, Botana LM, Yessotoxin induces ER-stress followed by autophagic cell death in glioma cells mediated by mTOR and BNIP3, Cell Signal 26(2) (2014) 419–32. [PubMed] [Google Scholar]
- [98].Semenza GL, HIF-1: upstream and downstream of cancer metabolism, Curr Opin Genet Dev 20(1) (2010) 51–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Glick D, Barth S, Macleod KF, Autophagy: cellular and molecular mechanisms, J Pathol 221(1) (2010) 3–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Rouschop KM, van den Beucken T, Dubois L, Niessen H, Bussink J, Savelkouls K, Keulers T, Mujcic H, Landuyt W, Voncken JW, Lambin P, van der Kogel AJ, Koritzinsky M, Wouters BG, The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5, J Clin Invest 120(1) (2010) 127–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Zhang T, Qi Y, Liao M, Xu M, Bower KA, Frank JA, Shen HM, Luo J, Shi X, Chen G, Autophagy is a cell self-protective mechanism against arsenic-induced cell transformation, Toxicol Sci 130(2) (2012) 298–308. [DOI] [PubMed] [Google Scholar]
- [102].Liu F, Wang XY, Zhou XP, Liu ZP, Song XB, Wang ZY, Wang L, Cadmium disrupts autophagic flux by inhibiting cytosolic Ca(2+)-dependent autophagosome-lysosome fusion in primary rat proximal tubular cells, Toxicology 383 (2017) 13–23. [DOI] [PubMed] [Google Scholar]
- [103].Yang CL, Chiou SH, Tai WC, Joseph NA, Chow KC, Trivalent chromium induces autophagy by activating sphingomyelin phosphodiesterase 2 and increasing cellular ceramide levels in renal HK2 cells, Mol Carcinog 56(11) (2017) 2424–2433. [DOI] [PubMed] [Google Scholar]
- [104].Kimura A, Ishida Y, Nosaka M, Kuninaka Y, Hama M, Kawaguchi T, Sakamoto S, Shinozaki K, Iwahashi Y, Takayasu T, Kondo T, Exaggerated arsenic nephrotoxicity in female mice through estrogen-dependent impairments in the autophagic flux, Toxicology 339 (2016) 9–18. [DOI] [PubMed] [Google Scholar]
- [105].Pal D, Suman S, Kolluru V, Sears S, Das TP, Alatassi H, Ankem MK, Freedman JH, Damodaran C, Inhibition of autophagy prevents cadmium-induced prostate carcinogenesis, Br J Cancer 117(1) (2017) 56–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Kolluru V, Tyagi A, Chandrasekaran B, Ankem M, Damodaran C, Induction of endoplasmic reticulum stress might be responsible for defective autophagy in cadmium-induced prostate carcinogenesis, Toxicol Appl Pharmacol 373 (2019) 62–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Siegel RL, Miller KD, Jemal A, Cancer statistics, 2020, CA Cancer J Clin 70(1) (2020) 7–30. [DOI] [PubMed] [Google Scholar]
- [108].Guo J, Deng W, Zhang L, Li C, Wu P, Mao P, Prediction of prostate cancer using hair trace element concentration and support vector machine method, Biol Trace Elem Res 116(3) (2007) 257–72. [DOI] [PubMed] [Google Scholar]
- [109].Bulka CM, Jones RM, Turyk ME, Stayner LT, Argos M, Arsenic in drinking water and prostate cancer in Illinois counties: An ecologic study, Environ Res 148 (2016) 450–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Chen C, Xun P, Nishijo M, Carter S, He K, Cadmium exposure and risk of prostate cancer: a meta-analysis of cohort and case-control studies among the general and occupational populations, Sci Rep 6 (2016) 25814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Platz EA, Helzlsouer KJ, Hoffman SC, Morris JS, Baskett CK, Comstock GW, Prediagnostic toenail cadmium and zinc and subsequent prostate cancer risk, Prostate 52(4) (2002) 288–96. [DOI] [PubMed] [Google Scholar]
- [112].Roh T, Lynch CF, Weyer P, Wang K, Kelly KM, Ludewig G, Low-level arsenic exposure from drinking water is associated with prostate cancer in Iowa, Environ Res 159 (2017) 338–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Sahmoun AE, Case LD, Jackson SA, Schwartz GG, Cadmium and prostate cancer: a critical epidemiologic analysis, Cancer Invest 23(3) (2005) 256–63. [DOI] [PubMed] [Google Scholar]
- [114].Chen CJ, Wang CJ, Ecological correlation between arsenic level in well water and age-adjusted mortality from malignant neoplasms, Cancer Res 50(17) (1990) 5470–4. [PubMed] [Google Scholar]
- [115].Wu MM, Kuo TL, Hwang YH, Chen CJ, Dose-response relation between arsenic concentration in well water and mortality from cancers and vascular diseases, Am J Epidemiol 130(6) (1989) 1123–32. [DOI] [PubMed] [Google Scholar]
- [116].Cohen MD, Kargacin B, Klein CB, Costa M, Mechanisms of chromium carcinogenicity and toxicity, Crit Rev Toxicol 23(3) (1993) 255–81. [DOI] [PubMed] [Google Scholar]
- [117].Costa M, Toxicity and carcinogenicity of Cr(VI) in animal models and humans, Crit Rev Toxicol 27(5) (1997) 431–42. [DOI] [PubMed] [Google Scholar]
- [118].Agency for Toxic Substances and Disease Registry, CERCLA Priority List of Hazardous Substances, 2015. http://www.atsdr.cdc.gov/SPL/index.html.
- [119].International Programme on Chemical Safety, Cadmium, 2015. http://www.who.int/ipcs/assessment/public_health/cadmium/en/.
- [120].Zimta AA, Schitcu V, Gurzau E, Stavaru C, Manda G, Szedlacsek S, Berindan-Neagoe I, Biological and molecular modifications induced by cadmium and arsenic during breast and prostate cancer development, Environ Res 178 (2019) 108700. [DOI] [PubMed] [Google Scholar]
- [121].Fox MR, Cadmium bioavailability, Fed. Proc 42 (1983) 1726–1729. [PubMed] [Google Scholar]
- [122].Satarug S, Baker JR, Urbenjapol S, Haswell-Elkins M, Reilly PE, Williams DJ, Moore MR, A global perspective on cadmium pollution and toxicity in non-occupationally exposed population, Toxicol Lett 137(1–2) (2003) 65–83. [DOI] [PubMed] [Google Scholar]
- [123].Agency for Toxic Substances and Disease Registry, Toxicological Profile for Cadmium (update), Agency for Toxic Substances and Disease Registry, Atlanta, GA,, 2012. [PubMed] [Google Scholar]
- [124].Aylett BJ, in: Webb M (Ed.), The Chemistry, Biochemistry and Biology of Cadmium, Elsevier/North-Holland, New York, 1979, pp. 1–62. [Google Scholar]
- [125].Chmielnicka J, Cherian MG, Environmental exposure to cadmium and factors affecting trance-metal metabolism and metal toxicity, Biol. Trance Elements Res 10 (1986) 243–256. [DOI] [PubMed] [Google Scholar]
- [126].Friberg L, Kjellstrom T, Nordberg GF, in: Friberg L, Nordberg GF, Vouk V (Eds.), Handbook of the Toxicology of Metals, Elsevier/North-Holland, Amsterdam, 1986, pp. 130–237. [Google Scholar]
- [127].Lewis GP, Coughlin LL, Jusko WJ, Hartz S, Contribution of cigarette smoking to cadmium accumulation in man, Lancet 1 (1972) 291–292. [DOI] [PubMed] [Google Scholar]
- [128].Watters JL, Park Y, Hollenbeck A, Schatzkin A, Albanes D, Cigarette smoking and prostate cancer in a prospective US cohort study, Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology 18(9) (2009) 2427–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Zu K, Giovannucci E, Smoking and aggressive prostate cancer: a review of the epidemiologic evidence, Cancer causes & control : CCC 20(10) (2009) 1799–810. [DOI] [PubMed] [Google Scholar]
- [130].Jarup L, Akesson A, Current status of cadmium as an environmental health problem, Toxicol Appl Pharmacol 238(3) (2009) 201–8. [DOI] [PubMed] [Google Scholar]
- [131].Alexander A, Cai SL, Kim J, Nanez A, Sahin M, MacLean KH, Inoki K, Guan KL, Shen J, Person MD, Kusewitt D, Mills GB, Kastan MB, Walker CL, ATM signals to TSC2 in the cytoplasm to regulate mTORC1 in response to ROS, Proc Natl Acad Sci U S A 107(9) (2010) 4153–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [132].Brys M, Nawrocka AD, Miekos E, Zydek C, Foksinski M, Barecki A, Krajewska WM, Zinc and cadmium analysis in human prostate neoplasms, Biol Trace Elem Res 59(1–3) (1997) 145–52. [DOI] [PubMed] [Google Scholar]
- [133].Lindegaard PM, Hansen SO, Christensen JE, Andersen BB, Andersen O, The distribution of cadmium within the human prostate, Biol Trace Elem Res 25(2) (1990) 97–104. [DOI] [PubMed] [Google Scholar]
- [134].Eriksen KT, Halkjaer J, Meliker JR, McElroy JA, Sorensen M, Tjonneland A, Raaschou-Nielsen O, Dietary cadmium intake and risk of prostate cancer: a Danish prospective cohort study, BMC Cancer 15 (2015) 177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].van der Gulden JW, Kolk JJ, Verbeek AL, Work environment and prostate cancer risk, Prostate 27(5) (1995) 250–7. [DOI] [PubMed] [Google Scholar]
- [136].Elinder CG, Kjellstrom T, Hogstedt C, Andersson K, Spang G, Cancer mortality of cadmium workers, Br J Ind Med 42(10) (1985) 651–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Lin YS, Caffrey JL, Lin JW, Bayliss D, Faramawi MF, Bateson TF, Sonawane B, Increased risk of cancer mortality associated with cadmium exposures in older Americans with low zinc intake, J Toxicol Environ Health A 76(1) (2013) 1–15. [DOI] [PubMed] [Google Scholar]
- [138].Julin B, Wolk A, Johansson JE, Andersson SO, Andren O, Akesson A, Dietary cadmium exposure and prostate cancer incidence: a population-based prospective cohort study, Br J Cancer 107(5) (2012) 895–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Achanzar WE, Diwan BA, Liu J, Quader ST, Webber MM, Waalkes MP, Cadmium-induced malignant transformation of human prostate epithelial cells, Cancer Res 61(2) (2001) 455–8. [PubMed] [Google Scholar]
- [140].Nakamura K, Yasunaga Y, Ko D, Xu LL, Moul JW, Peehl DM, Srivastava S, Rhim JS, Cadmium-induced neoplastic transformation of human prostate epithelial cells, Int J Oncol 20(3) (2002) 543–7. [PubMed] [Google Scholar]
- [141].Benbrahim-Tallaa L, Waterland RA, Dill AL, Webber MM, Waalkes MP, Tumor suppressor gene inactivation during cadmium-induced malignant transformation of human prostate cells correlates with overexpression of de novo DNA methyltransferase, Environ Health Perspect 115(10) (2007) 1454–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Kissinger PT, Riggin RM, Alcorn RL, Rau LD, Estimation of catecholamines in urine by high performance liquid chromatography with electrochemical detection, Biochem Med 13(4) (1975) 299–306. [DOI] [PubMed] [Google Scholar]
- [143].Waalkes MP, Cadmium carcinogenesis, Mutat Res 533(1–2) (2003) 107–20. [DOI] [PubMed] [Google Scholar]
- [144].Rani A, Kumar A, Lal A, Pant M, Cellular mechanisms of cadmium-induced toxicity: a review, Int J Environ Health Res 24(4) (2014) 378–99. [DOI] [PubMed] [Google Scholar]
- [145].Ju-Kun S, Yuan DB, Rao HF, Chen TF, Luan BS, Xu XM, Jiang FN, Zhong WD, Zhu JG, Association Between Cd Exposure and Risk of Prostate Cancer: A PRISMA-Compliant Systematic Review and Meta-Analysis, Medicine (Baltimore) 95(6) (2016) e2708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Waalkes MP, Poirier LA, In vitro cadmium-DNA interactions: cooperativity of cadmium binding and competitive antagonism by calcium, magnesium, and zinc, Toxicol Appl Pharmacol 75(3) (1984) 539–46. [DOI] [PubMed] [Google Scholar]
- [147].Hartwig A, Carcinogenicity of metal compounds: possible role of DNA repair inhibition, Toxicol Lett 102–103 (1998) 235–9. [DOI] [PubMed] [Google Scholar]
- [148].Chandrasekaran B, Dahiya NR, Tyagi A, Kolluru V, Saran U, Baby BV, States JC, Haddad AQ, Ankem MK, Damodaran C, Chronic exposure to cadmium induces a malignant transformation of benign prostate epithelial cells, Oncogenesis 9(2) (2020) 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Kolluru V, Pal D, Papu John AMS, Ankem MK, Freedman JH, Damodaran C, Induction of Plac8 promotes pro-survival function of autophagy in cadmium-induced prostate carcinogenesis, Cancer Lett (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Kolluru V PD, Papu John AMS, Ankem MK, Freedman JH, Damodaran C, Induction of Plac8 promotes pro-survival function of autophagy in cadmium-induced prostate carcinogenesis., Cancer Lett. 408(Nov 1) (2017) 121–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [151].Liu J, Qu W, Kadiiska MB, Role of oxidative stress in cadmium toxicity and carcinogenesis, Toxicol Appl Pharmacol 238(3) (2009) 209–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Stohs SJ, Bagchi D, Hassoun E, Bagchi M, Oxidative mechanisms in the toxicity of chromium and cadmium ions, J Environ Pathol Toxicol Oncol 19(3) (2000) 201–13. [PubMed] [Google Scholar]
- [153].Lutzen A, Liberti SE, Rasmussen LJ, Cadmium inhibits human DNA mismatch repair in vivo, Biochem Biophys Res Commun 321(1) (2004) 21–5. [DOI] [PubMed] [Google Scholar]
- [154].Pierron F, Baillon L, Sow M, Gotreau S, Gonzalez P, Effect of low-dose cadmium exposure on DNA methylation in the endangered European eel, Environ Sci Technol 48(1) (2014) 797–803. [DOI] [PubMed] [Google Scholar]
- [155].Asara Y, Marchal JA, Carrasco E, Boulaiz H, Solinas G, Bandiera P, Garcia MA, Farace C, Montella A, Madeddu R, Cadmium modifies the cell cycle and apoptotic profiles of human breast cancer cells treated with 5-fluorouracil, Int J Mol Sci 14(8) (2013) 16600–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Hartwig A, Mechanisms in cadmium-induced carcinogenicity: recent insights, Biometals 23(5) (2010) 951–60. [DOI] [PubMed] [Google Scholar]
- [157].Luevano J, Damodaran C, A review of molecular events of cadmium-induced carcinogenesis, J Environ Pathol Toxicol Oncol 33(3) (2014) 183–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Joseph P, Mechanisms of cadmium carcinogenesis, Toxicol Appl Pharmacol 238(3) (2009) 272–9. [DOI] [PubMed] [Google Scholar]
- [159].Khojastehfar A, Aghaei M, Gharagozloo M, Panjehpour M, Cadmium induces reactive oxygen species-dependent apoptosis in MCF-7 human breast cancer cell line, Toxicol Mech Methods 25(1) (2015) 48–55. [DOI] [PubMed] [Google Scholar]
- [160].Vella V, Malaguarnera R, Lappano R, Maggiolini M, Belfiore A, Recent views of heavy metals as possible risk factors and potential preventive and therapeutic agents in prostate cancer, Mol Cell Endocrinol 457 (2017) 57–72. [DOI] [PubMed] [Google Scholar]
- [161].Benbrahim-Tallaa L, Liu J, Webber MM, Waalkes MP, Estrogen signaling and disruption of androgen metabolism in acquired androgen-independence during cadmium carcinogenesis in human prostate epithelial cells, Prostate 67(2) (2007) 135–45. [DOI] [PubMed] [Google Scholar]
- [162].Benbrahim-Tallaa L, Webber MM, Waalkes MP, Mechanisms of acquired androgen independence during arsenic-induced malignant transformation of human prostate epithelial cells, Environ Health Perspect 115(2) (2007) 243–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Aimola P, Carmignani M, Volpe AR, Di Benedetto A, Claudio L, Waalkes MP, van Bokhoven A, Tokar EJ, Claudio PP, Cadmium induces p53-dependent apoptosis in human prostate epithelial cells, PLoS One 7(3) (2012) e33647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Achanzar WE, Achanzar KB, Lewis JG, Webber MM, Waalkes MP, Cadmium induces c-myc, p53, and c-jun expression in normal human prostate epithelial cells as a prelude to apoptosis, Toxicol Appl Pharmacol 164(3) (2000) 291–300. [DOI] [PubMed] [Google Scholar]
- [165].Achanzar WE, Webber MM, Waalkes MP, Altered apoptotic gene expression and acquired apoptotic resistance in cadmium-transformed human prostate epithelial cells, Prostate 52(3) (2002) 236–44. [DOI] [PubMed] [Google Scholar]
- [166].Misra UK, Gawdi G, Pizzo SV, Induction of mitogenic signalling in the 1LN prostate cell line on exposure to submicromolar concentrations of cadmium+, Cell Signal 15(11) (2003) 1059–70. [DOI] [PubMed] [Google Scholar]
- [167].Urani C, Melchioretto P, Fabbri M, Bowe G, Maserati E, Gribaldo L, Cadmium Impairs p53 Activity in HepG2 Cells, ISRN Toxicol 2014 (2014) 976428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Dasgupta P, Kulkarni P, Bhat NS, Majid S, Shiina M, Shahryari V, Yamamura S, Tanaka Y, Gupta RK, Dahiya R, Hashimoto Y, Activation of the Erk/MAPK signaling pathway is a driver for cadmium induced prostate cancer, Toxicol Appl Pharmacol 401 (2020) 115102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Kaistha BP, Lorenz H, Schmidt H, Sipos B, Pawlak M, Gierke B, Kreider R, Lankat-Buttgereit B, Sauer M, Fiedler L, Krattenmacher A, Geisel B, Kraus JM, Frese KK, Kelkenberg S, Giese NA, Kestler HA, Gress TM, Buchholz M, PLAC8 Localizes to the Inner Plasma Membrane of Pancreatic Cancer Cells and Regulates Cell Growth and Disease Progression through Critical Cell-Cycle Regulatory Pathways, Cancer Res 76(1) (2016) 96–107. [DOI] [PubMed] [Google Scholar]
- [170].Li C, Ma H, Wang Y, Cao Z, Graves-Deal R, Powell AE, Starchenko A, Ayers GD, Washington MK, Kamath V, Desai K, Gerdes MJ, Solnica-Krezel L, Coffey RJ, Excess PLAC8 promotes an unconventional ERK2-dependent EMT in colon cancer, J Clin Invest 124(5) (2014) 2172–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [171].Kinsey C, Balakrishnan V, O’Dell MR, Huang JL, Newman L, Whitney-Miller CL, Hezel AF, Land H, Plac8 links oncogenic mutations to regulation of autophagy and is critical to pancreatic cancer progression, Cell reports 7(4) (2014) 1143–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [172].Chen QY, Costa M, PI3K/Akt/mTOR Signaling Pathway and the Biphasic Effect of Arsenic in Carcinogenesis, Mol Pharmacol 94(1) (2018) 784–792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [173].Hong YS, Song KH, Chung JY, Health effects of chronic arsenic exposure, J Prev Med Public Health 47(5) (2014) 245–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Garcia-Esquinas E, Pollan M, Umans JG, Francesconi KA, Goessler W, Guallar E, Howard B, Farley J, Best LG, Navas-Acien A, Arsenic exposure and cancer mortality in a US-based prospective cohort: the strong heart study, Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology 22(11) (2013) 1944–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Yang CY, Chang CC, Chiu HF, Does arsenic exposure increase the risk for prostate cancer?, J Toxicol Environ Health A 71(23) (2008) 1559–63. [DOI] [PubMed] [Google Scholar]
- [176].Achanzar WE, Brambila EM, Diwan BA, Webber MM, Waalkes MP, Inorganic arsenite-induced malignant transformation of human prostate epithelial cells, J Natl Cancer Inst 94(24) (2002) 1888–91. [DOI] [PubMed] [Google Scholar]
- [177].Benbrahim-Tallaa L, Waterland RA, Styblo M, Achanzar WE, Webber MM, Waalkes MP, Molecular events associated with arsenic-induced malignant transformation of human prostatic epithelial cells: aberrant genomic DNA methylation and K-ras oncogene activation, Toxicol Appl Pharmacol 206(3) (2005) 288–98. [DOI] [PubMed] [Google Scholar]
- [178].Benbrahim-Tallaa L, Waalkes MP, Inorganic arsenic and human prostate cancer, Environ Health Perspect 116(2) (2008) 158–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [179].Shi H, Hudson LG, Ding W, Wang S, Cooper KL, Liu S, Chen Y, Shi X, Liu KJ, Arsenite causes DNA damage in keratinocytes via generation of hydroxyl radicals, Chem Res Toxicol 17(7) (2004) 871–8. [DOI] [PubMed] [Google Scholar]
- [180].Shi H, Shi X, Liu KJ, Oxidative mechanism of arsenic toxicity and carcinogenesis, Mol Cell Biochem 255(1–2) (2004) 67–78. [DOI] [PubMed] [Google Scholar]
- [181].Kessel M, Liu SX, Xu A, Santella R, Hei TK, Arsenic induces oxidative DNA damage in mammalian cells, Mol Cell Biochem 234–235(1–2) (2002) 301–8. [PubMed] [Google Scholar]
- [182].Bhadauria S, Flora SJ, Response of arsenic-induced oxidative stress, DNA damage, and metal imbalance to combined administration of DMSA and monoisoamyl-DMSA during chronic arsenic poisoning in rats, Cell Biol Toxicol 23(2) (2007) 91–104. [DOI] [PubMed] [Google Scholar]
- [183].Lee TC, Ho IC, Modulation of cellular antioxidant defense activities by sodium arsenite in human fibroblasts, Arch Toxicol 69(7) (1995) 498–504. [DOI] [PubMed] [Google Scholar]
- [184].Dong JT, Luo XM, Arsenic-induced DNA-strand breaks associated with DNA-protein crosslinks in human fetal lung fibroblasts, Mutat Res 302(2) (1993) 97–102. [DOI] [PubMed] [Google Scholar]
- [185].Ngalame NN, Tokar EJ, Person RJ, Waalkes MP, Silencing KRAS overexpression in arsenic-transformed prostate epithelial and stem cells partially mitigates malignant phenotype, Toxicol Sci 142(2) (2014) 489–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Chen F, Ding M, Lu Y, Leonard SS, Vallyathan V, Castranova V, Shi X, Participation of MAP kinase p38 and IkappaB kinase in chromium (VI)-induced NF-kappaB and AP-1 activation, J Environ Pathol Toxicol Oncol 19(3) (2000) 231–8. [PubMed] [Google Scholar]
- [187].Kaltreider RC, Pesce CA, Ihnat MA, Lariviere JP, Hamilton JW, Differential effects of arsenic(III) and chromium(VI) on nuclear transcription factor binding, Mol Carcinog 25(3) (1999) 219–29. [PubMed] [Google Scholar]
- [188].Ngalame NN, Makia NL, Waalkes MP, Tokar EJ, Mitigation of arsenic-induced acquired cancer phenotype in prostate cancer stem cells by miR-143 restoration, Toxicol Appl Pharmacol 312 (2016) 11–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Zhao R, Bei X, Yang B, Wang X, Jiang C, Shi F, Wang X, Zhu Y, Jing Y, Han B, Xia S, Jiang Q, Endothelial cells promote metastasis of prostate cancer by enhancing autophagy, J Exp Clin Cancer Res 37(1) (2018) 221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Kim KY, Park KI, Kim SH, Yu SN, Park SG, Kim YW, Seo YK, Ma JY, Ahn SC, Inhibition of Autophagy Promotes Salinomycin-Induced Apoptosis via Reactive Oxygen Species-Mediated PI3K/AKT/mTOR and ERK/p38 MAPK-Dependent Signaling in Human Prostate Cancer Cells, Int J Mol Sci 18(5) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [191].Tai S, Xu L, Xu M, Zhang L, Zhang Y, Zhang K, Zhang L, Liang C, Combination of Arsenic trioxide and Everolimus (Rad001) synergistically induces both autophagy and apoptosis in prostate cancer cells, Oncotarget 8(7) (2017) 11206–11218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [192].Chiu HW, Chen YA, Ho SY, Wang YJ, Arsenic trioxide enhances the radiation sensitivity of androgen-dependent and -independent human prostate cancer cells, PLoS One 7(2) (2012) e31579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Costa M, Klein CB, Toxicity and carcinogenicity of chromium compounds in humans, Crit Rev Toxicol 36(2) (2006) 155–63. [DOI] [PubMed] [Google Scholar]
- [194].Brocato J, Costa M, Basic mechanics of DNA methylation and the unique landscape of the DNA methylome in metal-induced carcinogenesis, Crit Rev Toxicol 43(6) (2013) 493–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [195].Shumilla JA, Wetterhahn KE, Barchowsky A, Inhibition of NF-kappa B binding to DNA by chromium, cadmium, mercury, zinc, and arsenite in vitro: evidence of a thiol mechanism, Arch Biochem Biophys 349(2) (1998) 356–62. [DOI] [PubMed] [Google Scholar]
- [196].Ye J, Zhang X, Young HA, Mao Y, Shi X, Chromium(VI)-induced nuclear factor-kappa B activation in intact cells via free radical reactions, Carcinogenesis 16(10) (1995) 2401–5. [DOI] [PubMed] [Google Scholar]
- [197].Gao N, Jiang BH, Leonard SS, Corum L, Zhang Z, Roberts JR, Antonini J, Zheng JZ, Flynn DC, Castranova V, Shi X, p38 Signaling-mediated hypoxia-inducible factor 1alpha and vascular endothelial growth factor induction by Cr(VI) in DU145 human prostate carcinoma cells, J Biol Chem 277(47) (2002) 45041–8. [DOI] [PubMed] [Google Scholar]
- [198].Martinez VD, Vucic EA, Becker-Santos DD, Gil L, Lam WL, Arsenic exposure and the induction of human cancers, J Toxicol 2011 (2011) 431287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [199].Smith AH, Goycolea M, Haque R, Biggs ML, Marked increase in bladder and lung cancer mortality in a region of Northern Chile due to arsenic in drinking water, Am J Epidemiol 147(7) (1998) 660–9. [DOI] [PubMed] [Google Scholar]
- [200].Smith AH, Marshall G, Roh T, Ferreccio C, Liaw J, Steinmaus C, Lung, Bladder, and Kidney Cancer Mortality 40 Years After Arsenic Exposure Reduction, J Natl Cancer Inst 110(3) (2018) 241–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [201].Hughes MF, Beck BD, Chen Y, Lewis AS, Thomas DJ, Arsenic exposure and toxicology: a historical perspective, Toxicol Sci 123(2) (2011) 305–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [202].Wang XJ, Sun Z, Chen W, Li Y, Villeneuve NF, Zhang DD, Activation of Nrf2 by arsenite and monomethylarsonous acid is independent of Keap1-C151: enhanced Keap1-Cul3 interaction, Toxicol Appl Pharmacol 230(3) (2008) 383–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [203].Lau A, Zheng Y, Tao S, Wang H, Whitman SA, White E, Zhang DD, Arsenic inhibits autophagic flux, activating the Nrf2-Keap1 pathway in a p62-dependent manner, Mol Cell Biol 33(12) (2013) 2436–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [204].Li L, Chen F, Oxidative stress, epigenetics, and cancer stem cells in arsenic carcinogenesis and prevention, Curr Pharmacol Rep 2(2) (2016) 57–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [205].Hu Y, Li J, Lou B, Wu R, Wang G, Lu C, Wang H, Pi J, Xu Y, The Role of Reactive Oxygen Species in Arsenic Toxicity, Biomolecules 10(2) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [206].Song Y, Zhang P, Sun Y, Li X, Chen L, Xiao Y, Xing Y, AMPK activation-dependent autophagy compromises oleanolic acid-induced cytotoxicity in human bladder cancer cells, Oncotarget 8(40) (2017) 67942–67954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [207].Beauchamp EM, Kosciuczuk EM, Serrano R, Nanavati D, Swindell EP, Viollet B, O’Halloran TV, Altman JK, Platanias LC, Direct binding of arsenic trioxide to AMPK and generation of inhibitory effects on acute myeloid leukemia precursors, Mol Cancer Ther 14(1) (2015) 202–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [208].Larson JL, Somji S, Zhou XD, Sens MA, Garrett SH, Sens DA, Dunlevy JR, Beclin-1 expression in normal bladder and in Cd2+ and As3+ exposed and transformed human urothelial cells (UROtsa), Toxicol Lett 195(1) (2010) 15–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [209].Wolf C, Strenziok R, Kyriakopoulos A, Elevated metallothionein-bound cadmium concentrations in urine from bladder carcinoma patients, investigated by size exclusion chromatography-inductively coupled plasma mass spectrometry, Anal Chim Acta 631(2) (2009) 218–22. [DOI] [PubMed] [Google Scholar]
- [210].Feki-Tounsi M, Olmedo P, Gil F, Khlifi R, Mhiri MN, Rebai A, Hamza-Chaffai A, Cadmium in blood of Tunisian men and risk of bladder cancer: interactions with arsenic exposure and smoking, Environ Sci Pollut Res Int 20(10) (2013) 7204–13. [DOI] [PubMed] [Google Scholar]
- [211].Kellen E, Zeegers MP, Hond ED, Buntinx F, Blood cadmium may be associated with bladder carcinogenesis: the Belgian case-control study on bladder cancer, Cancer Detect Prev 31(1) (2007) 77–82. [DOI] [PubMed] [Google Scholar]
- [212].Waalkes MP, Rehm S, Cherian MG, Repeated cadmium exposures enhance the malignant progression of ensuing tumors in rats, Toxicol Sci 54(1) (2000) 110–20. [DOI] [PubMed] [Google Scholar]
- [213].Somji S, Bathula CS, Zhou XD, Sens MA, Sens DA, Garrett SH, Transformation of human urothelial cells (UROtsa) by as and cd induces the expression of keratin 6a, Environ Health Perspect 116(4) (2008) 434–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [214].Son YO, Pratheeshkumar P, Roy RV, Hitron JA, Wang L, Zhang Z, Shi X, Nrf2/p62 signaling in apoptosis resistance and its role in cadmium-induced carcinogenesis, J Biol Chem 289(41) (2014) 28660–75. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- [215].Liuzzi JP, Pazos R, Interplay Between Autophagy and Zinc, J Trace Elem Med Biol 62 (2020) 126636. [DOI] [PubMed] [Google Scholar]
- [216].Henry SA, Kennaway NM, Kennaway EL, The Incidence of Cancer of the Bladder and Prostate in Certain Occupations, J Hyg (Lond) 31(2) (1931) 125–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [217].Claude J, Kunze E, Frentzel-Beyme R, Paczkowski K, Schneider J, Schubert H, Life-style and occupational risk factors in cancer of the lower urinary tract, Am J Epidemiol 124(4) (1986) 578–89. [DOI] [PubMed] [Google Scholar]
- [218].Golabek T, Socha K, Kudelski J, Darewicz B, Markiewicz-Zukowska R, Chlosta P, Borawska M, Chromium in urothelial carcinoma of the bladder, Ann Agric Environ Med 24(4) (2017) 602–605. [DOI] [PubMed] [Google Scholar]
- [219].Montanaro F, Ceppi M, Demers PA, Puntoni R, Bonassi S, Mortality in a cohort of tannery workers, Occup Environ Med 54(8) (1997) 588–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [220].Kunze E, Chang-Claude J, Frentzel-Beyme R, [Etiology, pathogenesis and epidemiology or urothelial tumors], Verh Dtsch Ges Pathol 77 (1993) 147–56. [PubMed] [Google Scholar]
- [221].Wise SS, Holmes AL, Liou L, Adam RM, Wise JP Sr., Hexavalent chromium induces chromosome instability in human urothelial cells, Toxicol Appl Pharmacol 296 (2016) 54–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [222].Yang Q, Han B, Xue J, Lv Y, Li S, Liu Y, Wu P, Wang X, Zhang Z, Hexavalent chromium induces mitochondrial dynamics disorder in rat liver by inhibiting AMPK/PGC-1alpha signaling pathway, Environ Pollut 265(Pt A) (2020) 114855. [DOI] [PubMed] [Google Scholar]
- [223].Liang Q, Xiao Y, Liu K, Zhong C, Zeng M, Xiao F, Cr(VI)-Induced Autophagy Protects L-02 Hepatocytes from Apoptosis Through the ROS-AKT-mTOR Pathway, Cell Physiol Biochem 51(4) (2018) 1863–1878. [DOI] [PubMed] [Google Scholar]
- [224].Mahalakshmi R, Priyanga J, Vedha Hari BN, Bhakta-Guha D, Guha G, Hexavalent chromium-induced autophagic death of WRL-68 cells is mitigated by aqueous extract of Cuminum cyminum L. seeds, 3 Biotech 10(5) (2020) 191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [225].Johri N, Jacquillet G, Unwin R, Heavy metal poisoning: the effects of cadmium on the kidney, Biometals 23(5) (2010) 783–92. [DOI] [PubMed] [Google Scholar]
- [226].Zalups RK, Ahmad S, Molecular handling of cadmium in transporting epithelia, Toxicol Appl Pharmacol 186(3) (2003) 163–88. [DOI] [PubMed] [Google Scholar]
- [227].Wolff NA, Abouhamed M, Verroust PJ, Thevenod F, Megalin-dependent internalization of cadmium-metallothionein and cytotoxicity in cultured renal proximal tubule cells, J Pharmacol Exp Ther 318(2) (2006) 782–91. [DOI] [PubMed] [Google Scholar]
- [228].Wang C, Nie G, Zhuang Y, Hu R, Wu H, Xing C, Li G, Hu G, Yang F, Zhang C, Inhibition of autophagy enhances cadmium-induced apoptosis in duck renal tubular epithelial cells, Ecotoxicol Environ Saf 205 (2020) 111188. [DOI] [PubMed] [Google Scholar]
- [229].Wang SH, Shih YL, Ko WC, Wei YH, Shih CM, Cadmium-induced autophagy and apoptosis are mediated by a calcium signaling pathway, Cell Mol Life Sci 65(22) (2008) 3640–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [230].Liu G, Yuan Y, Long M, Luo T, Bian J, Liu X, Gu J, Zou H, Song R, Wang Y, Wang L, Liu Z, Beclin-1-mediated Autophagy Protects Against Cadmium-activated Apoptosis via the Fas/FasL Pathway in Primary Rat Proximal Tubular Cell Culture, Sci Rep 7(1) (2017) 977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [231].Thevenod F, Lee WK, Live and Let Die: Roles of Autophagy in Cadmium Nephrotoxicity, Toxics 3(2) (2015) 130–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [232].Mikhaylova O, Stratton Y, Hall D, Kellner E, Ehmer B, Drew AF, Gallo CA, Plas DR, Biesiada J, Meller J, Czyzyk-Krzeska MF, VHL-regulated MiR-204 suppresses tumor growth through inhibition of LC3B-mediated autophagy in renal clear cell carcinoma, Cancer Cell 21(4) (2012) 532–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [233].Xu G, Jiang Y, Xiao Y, Liu XD, Yue F, Li W, Li X, He Y, Jiang X, Huang H, Chen Q, Jonasch E, Liu L, Fast clearance of lipid droplets through MAP1S-activated autophagy suppresses clear cell renal cell carcinomas and promotes patient survival, Oncotarget 7(5) (2016) 6255–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [234].Fabrizio FP, Costantini M, Copetti M, la Torre A, Sparaneo A, Fontana A, Poeta L, Gallucci M, Sentinelli S, Graziano P, Parente P, Pompeo V, De Salvo L, Simone G, Papalia R, Picardo F, Balsamo T, Flammia GP, Trombetta D, Pantalone A, Kok K, Paranita F, Muscarella LA, Fazio VM, Keap1/Nrf2 pathway in kidney cancer: frequent methylation of KEAP1 gene promoter in clear renal cell carcinoma, Oncotarget 8(7) (2017) 11187–11198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [235].Cheng YY, Huang NC, Chang YT, Sung JM, Shen KH, Tsai CC, Guo HR, Associations between arsenic in drinking water and the progression of chronic kidney disease: A nationwide study in Taiwan, J Hazard Mater 321 (2017) 432–439. [DOI] [PubMed] [Google Scholar]
- [236].Chu BX, Fan RF, Lin SQ, Yang DB, Wang ZY, Wang L, Interplay between autophagy and apoptosis in lead(II)-induced cytotoxicity of primary rat proximal tubular cells, J Inorg Biochem 182 (2018) 184–193. [DOI] [PubMed] [Google Scholar]
- [237].Saint-Jacques N, Brown P, Nauta L, Boxall J, Parker L, Dummer TJB, Estimating the risk of bladder and kidney cancer from exposure to low-levels of arsenic in drinking water, Nova Scotia, Canada, Environ Int 110 (2018) 95–104. [DOI] [PubMed] [Google Scholar]
- [238].Yuan Y, Marshall G, Ferreccio C, Steinmaus C, Liaw J, Bates M, Smith AH, Kidney cancer mortality: fifty-year latency patterns related to arsenic exposure, Epidemiology 21(1) (2010) 103–8. [DOI] [PubMed] [Google Scholar]
- [239].Hsueh YM, Chung CJ, Shiue HS, Chen JB, Chiang SS, Yang MH, Tai CW, Su CT, Urinary arsenic species and CKD in a Taiwanese population: a case-control study, Am J Kidney Dis 54(5) (2009) 859–70. [DOI] [PubMed] [Google Scholar]
- [240].Srivastava RK, Traylor AM, Li C, Feng W, Guo L, Antony VB, Schoeb TR, Agarwal A, Athar M, Cutaneous exposure to lewisite causes acute kidney injury by invoking DNA damage and autophagic response, Am J Physiol Renal Physiol 314(6) (2018) F1166–F1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [241].Tian X, Xie J, Chen X, Dong N, Feng J, Gao Y, Tian F, Zhang W, Qiu Y, Niu R, Ren X, Yan X, Deregulation of autophagy is involved in nephrotoxicity of arsenite and fluoride exposure during gestation to puberty in rat offspring, Arch Toxicol 94(3) (2020) 749–760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [242].Deng Y, Wang M, Tian T, Lin S, Xu P, Zhou L, Dai C, Hao Q, Wu Y, Zhai Z, Zhu Y, Zhuang G, Dai Z, The Effect of Hexavalent Chromium on the Incidence and Mortality of Human Cancers: A Meta-Analysis Based on Published Epidemiological Cohort Studies, Front Oncol 9 (2019) 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [243].Kim D, Dai J, Fai LY, Yao H, Son YO, Wang L, Pratheeshkumar P, Kondo K, Shi X, Zhang Z, Constitutive activation of epidermal growth factor receptor promotes tumorigenesis of Cr(VI)-transformed cells through decreased reactive oxygen species and apoptosis resistance development, J Biol Chem 290(4) (2015) 2213–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
