Abstract
Iron, the most abundant metal in human brain, is an essential microelement that regulates numerous cellular mechanisms. Some key physiological roles of iron include oxidative phosphorylation and ATP production, embryonic neuronal development, formation of iron-sulfur clusters, and the regulation of enzymes involved in DNA synthesis and repair. Because of its physiological and pathological importance, iron homeostasis must be tightly regulated by balancing its uptake, transport, and storage. Endosomes and lysosomes (endolysosomes) are acidic organelles known to contain readily releasable stores of various cations including iron and other metals. Increased levels of ferrous (Fe2+) iron can generate reactive oxygen species (ROS) via Fenton chemistry reactions and these increases can damage mitochondria and genomic DNA as well as promote carcinogenesis. Accumulation of iron in the brain has been linked with aging, diet, disease, and cerebral hemorrhage. Further, deregulation of brain iron metabolism has been implicated in carcinogenesis and may be a contributing factor to the increased incidence of brain tumors around the world. Here, we provide insight into mechanisms by which iron accumulation in endolysosomes is altered by pH and lysosome membrane permeabilization. Such events generate excess ROS resulting in mitochondrial DNA damage, fission, and dysfunction, as well as DNA oxidative damage in the nucleus; all of which promote carcinogenesis. A better understanding of the roles that endolysosome iron plays in carcinogenesis may help better inform the development of strategic therapeutic options for cancer treatment and prevention.
Keywords: Brain, Iron, Endolysosomes, ROS, Carcinogens
Introduction
Iron is the most abundant transition metal on Earth and the most abundant metal in human brain [1, 2]. Iron is essential for life, is an indispensable nutrient, and helps regulate numerous biological processes and mechanisms. These crucial roles of iron include DNA and RNA synthesis, embryonic neuronal development [3], oxidative phosphorylation and ATP synthesis [4], heme synthesis, oxygen transport, myelin synthesis [5], neurotransmitter synthesis [6, 7], immune metabolism, and the activity of a multitude of enzymes [8, 9]. Iron cycles between ferric (Fe3+) and ferrous (Fe2+) forms and affects redox reactions through the formation of complexes with oxygen. In addition, iron forms the iron-sulfur clusters in mitochondria and plays key roles in the electron transport chain [10]. Because of iron’s importance in regulating cellular functions, organisms have developed highly regulated homeostatic systems that balance the uptake, transport, and storage of this element [11].
Excess iron or a disrupted balance of iron homeostasis easily becomes detrimental to cells and is a source of oxidative damage through the formation of free radicals known as reactive oxygen species (ROS). Because iron is a metal capable of transitioning between Fe3+ and Fe2+ as well as transporting electrons, excess free (labile) iron can produce harmful levels of ROS via Fenton chemistry reactions:
Increased levels of iron can cause cellular damage and increase the risk of cancer, and dysregulated iron has been widely implicated in carcinogenesis [12, 13]. Numerous studies have examined iron regulation pathways and analyzed links between iron levels and enhanced tumor growth [14–16]. For example, high cellular concentrations of iron result in the conversion of hydrogen peroxide and superoxide into the highly reactive hydroxyl radical, which is a specific oxidant responsible for inducing DNA strand breaks and damage [17–19]. Furthermore, hydroxyl radical-induced DNA damage activates the K-ras and C-raf oncogenes via point mutations and deletions, respectively, which suggest that oncogene activation is a potential mechanism of oxidant-induced carcinogenesis [20]. In addition, increased oxygen levels make iron-sulfur clusters susceptible to oxidation and Fenton chemistry reactions that damage DNA [21, 22], and increased iron-sulfur clusters that accumulated the iron-storage complex hemosiderin were detected in macrophage deposits of brain metastatic tumors [23]. These specific mechanisms involving iron, DNA damage, and oxidant-induced carcinogenesis are supported by clinical findings associated with tumor development and progression.
Lymphomas and gliomas are the most common primary brain tumors in adults [24], and the global occurrence of all brain tumors is 10.8 per 100,000 individuals per year [25]. Glioblastoma multiforme (GBM) is the most aggressive of brain tumors, and despite recent advances, survival remains dismal; 2-year survival is 18%, 3-year survival is 11%, and 5-year survival is 4% in patients diagnosed with GBM during or after 2005 [26]. This demonstrates the need to develop novel therapeutic approaches, and studies investigating subcellular mechanisms associated with carcinogenesis in brain have the potential to inform these approaches. Studies have compared the overall survival data from patients with brain tumors to altered gene expression data necessary for iron uptake into the cell, and the results showed that most of the proteins involved in iron uptake were expressed at high levels and were predictive of poor survival [27–29]. These data suggest that iron plays important physiological roles in brain development and helps maintain healthy cellular functions, and excess iron may play a role as a brain carcinogen.
Iron Uptake into the Body
Iron is absorbed (90%) mostly by duodenum enterocytes [30] and comes from two types of dietary iron in its ferric form; heme (mainly from hemoglobin and myoglobin in red meat) and non-heme (from vegetables) [31, 32]. The divalent metal transporter 1 (DMT1) transports iron across enterocyte membranes and out of the endolysosomes into the cytosol as Fe2+ [33]. Ferroportin (FPN) is the primary exporter of intracellular iron that transfers duodenal iron into systemic circulation [34]. Released Fe2+ is then oxidized to Fe3+ by hephaestin or ceruloplasmin in plasma, and it is eventually bound to transferrin (Tf) in blood [35]. Within the body, it is important to note that iron is primarily complexed with the oxygen-carrying heme protein hemoglobin; 75% of the body’s iron is contained in erythrocytes and this is known as heme iron. Non-heme iron forms complexes with Tf, the iron storage protein ferritin [36] as well as amino acids, albumin, and ATP [37, 38]. The average iron concentration in serum ranges from 9 to 30 μM and it is mostly bound to Tf; including hemoglobin the iron concentration of blood is about 8 mM [39, 40].
Transferrin receptors (TfR) are ubiquitously expressed on plasma membranes of most cells and when combined with holo-Tf (Tf containing diferric iron) a complex is formed and then endocytosed [41, 42]. Fe3+ dissociates from Tf in the acidic environment of endolysosomes; apo-Tf (Tf with no iron) still bound to TfR can then be exocytosed back into the systemic circulation [27, 43]. Virtually no iron is excreted; almost all iron is reabsorbed by the kidneys and exported back into circulation [44]. Situations where iron is lost include blood donations, and excessive or abnormal bleeding.
Once iron is endocytosed, the six-transmembrane epithelial antigen of prostate 3 (STEAP3) ferrireductase protein reduces Fe3+ to Fe2+ within endolysosomes. Fe2+ can be released into the cytosol via DMT1 [42, 45, 46] where it can be stored as Fe3+ bound to ferritin, can be taken up into mitochondria, or it can be exported from cells by FPN [47, 48]. However, relatively little is known about the concentration of the labile iron pool in endolysosomes and its key roles in physiological and pathophysiological processes.
Iron Uptake into the Brain
Brain iron is essential for many neurological processes [49] and iron levels are mainly regulated by the blood brain barrier (BBB) [50]. General brain iron transport/homeostasis is illustrated in Figure 1. Iron transport into brain uses holo-Tf complexes and non-transferrin-bound iron (NTBI) that enters into endothelial cells [51, 52]. Iron crosses the BBB first by being taken up by the microvascular endothelial cells at the luminal membrane and second by being released into brain interstitial spaces at the abluminal membrane [53]. The holo-Tf-TfR complex is the main pathway for iron transport across the endothelial luminal membrane [54–57], and iron, likely in the form of Fe2+, crosses the abluminal membrane, enters into brain parenchyma, gets oxidized to Fe3+, and binds Tf [58–61]. The endocytosis of holo-Tf bound to TfR occurs similarly in other cell types in the human body [55]. However, iron transport into brain can bypass the BBB [62] and iron can cross the BBB faster than Tf [63]. The physiological relevance of these phenomena remains unclear [64, 65].
Neurons, astrocytes, and microglia can all uptake iron by means of the TfR-mediated uptake of holo-Tf [66–71]. In contrast, oligodendrocytes appear to uptake iron through processes involving H-ferritin binding to H-ferritin receptors and endocytosis; there is an absence of TfR and DMT1 in these cells [72–75]. Neurons, astrocytes, and microglia can uptake NTBI by means of a Tf-independent pathway; a process not apparent in oligodendrocytes [58, 76–80]. Because the affinity of transferrin for iron is pH-dependent, acidic environments in brain such as occurs in stroke and cancer [81, 82] results in decreased ability of transferrin to bind iron [83], dissociation of iron from Tf [84], and the uptake of NTBI by neurons and other cells [85].
Homeostatic regulation of iron levels also depends on iron efflux out of cell by iron exporters and ferroxidase. The iron exporter, FPN is expressed in neurons [86, 87], oligodendrocytes [87, 88], astrocytes [87, 89], and microglia [70, 89]. Ceruloplasmin, a ferroxidase, is found in neurons [90, 91] and astrocytes [92, 93], while the ferroxidase hephaestin is found in neurons [94, 95], oligodendrocytes [88, 96], and microglia [88].
Regulation of Iron Transport and Storage in the Brain
Iron levels in brain are second only to liver and vary between different brain regions [97, 98]. The majority of brain iron (33 to 75%) resides within glial cells [99] and is bound to ferritin in its soluble form [100]. Ferritin can bind 2,000 to 4,500 iron molecules [101] and recycle iron for heme synthesis as well as sequester labile iron to protect cells from oxidative damage. Ferritins are found in greater quantity in white matter than in gray matter [102]. Hemosiderin, which is a degradation product of ferritin, stores iron in an insoluble state [103]; iron can be mobilized from hemosiderin [104], however the physiological significance of this pool of iron remains unclear [98]. Despite the presence of iron binding proteins and export systems, incubation of cells with Fe3+ results in increases in the intracellular labile iron pool and oxidative damage [105], [106].
While most cells can regulate levels of iron by cell division, neurons are post-mitotic cells and must rely on iron homeostatic mechanisms to avoid iron-induced oxidative stress [105], including downregulating the expression of TfR [107, 108]. Neurons can also control levels of iron at the molecular level via post-transcriptional modification by iron regulatory proteins (IRP1 and IRP2). IRP1 and IRP2 are cytosolic proteins that bind to the iron-responsive elements (IRE) within the untranslated regions of the iron regulatory proteins ferritin, TfR and DMT1 [105].
Iron Content of Different Brain Regions
Iron levels in brain vary greatly between brain regions. The highest iron levels are found in the globus pallidus (GP) followed by the red nucleus, substantia nigra (SN), putamen, and dentate nucleus. The lowest levels of iron are found in regions of the cerebral cortex [97]. GP and SN have the highest levels of stainable iron (3.3–3.8 mM) [97, 98]. The forebrain, midbrain, and cerebellar structures had moderate staining levels of iron, while brain-stem and spinal cord had the lowest intensity of staining [97]. These early findings have been largely confirmed in later studies; high iron concentrations have been noted in GP, dentate gyrus, interpeduncular nucleus, thalamus, ventral palladium, nucleus basalis, and red nucleus [109]. Non-heme iron concentrations in the human parietal cortex were 0.65 mM [97]. Regions of brain associated with motor functions tend to have a higher content of iron [109] and this is likely due to high rates of oxidative metabolism [7], which is important in maintaining ionic membrane gradients, axonal transport, and synaptic transmissions [110].
As humans age, the brain tends to accumulate iron [48], and some regions accumulate more than others. The brain regions that accumulate the most iron with aging are SN, putamen, GP, caudate nucleus, and cortex; other regions such as the locus coeruleus continue to have low iron concentrations throughout life [111]. A number of explanations for this observation include increased BBB permeability, inflammation, redistribution of iron within the brain, and changes in iron homoeostasis [111]. In young, healthy brains, oligodendrocytes within the brain contain the highest iron concentration, while astrocytes have a low cellular iron content [112]. However, with aging, both microglia and astrocytes accumulate iron [113], which could be due to inflammatory processes as both cell types respond to inflammatory stimuli. This is not true for oligodendrocytes, which have a consistent iron content over time despite their higher concentrations of cellular iron to begin with [114].
Conditions Associated with Increased Iron in Brain
Iron accumulates in brain under normal and pathological conditions. Over and above age-induced increases in brain iron, iron accumulation also increases in individuals with certain pathologic conditions [115]. Subsequent to intracerebral hemorrhage (ICH), subarachnoid hemorrhage (SAH), or hypoxia-ischemia, iron is released from Tf or ferritin and accumulates [116]. In addition, following ICH and SAH neurons are exposed to blood components [117] including hemoglobin, which is highly neurotoxic [54, 117–119] and heme oxygenases (HO-1, HO-2) that cleave heme within hemoglobin producing free Fe2+ that induces free radicals and lipid peroxidation [54, 120]. Such insult-induced increases in reactive iron can persist for several days [121] and the observed neurological deficits appear to be due to iron accumulation because the iron chelator deferoxamine (DFO) protects against the brain damage [122].
Our diets also have a key role in iron accumulation, both systemically and in the brain. Heme iron in meat, poultry, and fish is highly absorbed; non-heme iron from plants is less absorbed [123]. Although heme iron is considered an essential nutrient [124], the high heme iron content in red meat has been associated with diseases including brain cancer [125, 126]. Thus, different lifestyles and diseases play key roles in the accumulation of iron in brain may contribute to dysregulation of iron homeostasis (Figure 2).
Heavy Metal and Trace Metal Elements in Carcinogenesis
Heavy metals can have biologic as well as toxic effects depending on their concentrations [127]. Small amounts of metals are necessary for physiological growth, development, and maintenance of cells and organisms [128]. Trace metals can function as micronutrients when they are present in healthy tissues and have known biological functions. Such essential microelements include aluminum, bromine, cobalt, copper, chromium, fluorine, germanium, iron, iodine, lithium, magnesium, manganese, molybdenum, selenium, strontium, zinc, nickel, tin, vanadium, and silicon. Non-essential microelements such as antimony, arsenic, barium, bismuth, cadmium, gold, lead, mercury, rubidium, silver, titanium, and zirconium, are present in animal tissues, but have either unknown or unclear biological functions and/or known neurotoxic impacts [127, 129–134].
Trace metal elements continue to be implicated in cancer progression; their concentrations differ between cancerous and noncancerous tissues, between cancer types, and between different malignancy grades [135, 136]. Numerous studies have focused on trace element levels in cancerous and noncancerous tissue from different human organs including brain [135, 137]. The International Agency for Research on Cancer (IARC) developed a four group classification system to evaluate the carcinogenicity of agents to humans based on scientific evidence from human and animal studies (International Agency for Research on Cancer; IARC) with Group 1 representing those agents labeled as “carcinogenic to humans” and defined as having “sufficient evidence of carcinogenicity”. Group 2 represents likely carcinogenic agents, divided into two subgroups (probable and possible). Groups 3 and 4 represent agents “not able to be classified as carcinogenic” and “probably not carcinogenic”, respectively. Here we will consider how iron works in conjunction with two non-essential heavy metal elements arsenic and cadmium as well as two essential elements chromium and nickel; all four of these heavy metals (arsenic, cadmium, chromium, and nickel) are classified into Group 1 by IARC.
Heavy metals localize to and accumulate in endolysosomes of brain cells (Figure 2) [138, 139]. This organellar store of heavy metals might affect the structure and function of other organelles including mitochondria because cadmium and arsenic are strong uncouplers of oxidative phosphorylation [140, 141], and the nucleus, because nickel can inhibit RNA polymerase activity [142] and cadmium severely inhibits RNA synthesis [143]. Thus, disrupted functions in the nucleus and in mitochondria might originate from heavy metals found in endolysosomes. In contrast to other cancers [135, 137] relatively little is known about trace metals in brain tumors [144], and what information is available is generally highly specific to the tumor type and tumor grade. Astrocytomas (grade I-III) have significantly higher concentrations of arsenic than healthy brain tissue [137]. In contrast, nickel concentrations do not significantly change between glioblastoma (grade IV) and healthy brains [145, 146]. Iron and chromium are significantly increased in astrocytoma (III) and glioblastoma (IV) tumors compared to normal adjacent tissue [147, 148], but the higher the malignancy grade between astrocytoma (I-III) and GBM (IV), the lower are the iron and cadmium concentrations [149, 150].
Distribution of trace metals within individual tumors has also been examined, although studies of tumor tissue distribution was frequently limited by lack of direct within patient tumor-to-normal comparisons, relying instead on comparisons of tumor samples to brain samples from other age-matched individuals [145, 149]; Nevertheless, changes in distribution of trace elements have been observed, with lead, zinc, calcium, and uranium showing homogeneous tumor distribution, while copper and iron are heterogeneously distributed [151, 152].
Mechanisms of Iron and Other Metal Carcinogenesis in Brain Malignancy
Over the last three decades, biochemical studies have contributed to our understanding of metal-induced carcinogenesis. Heavy metals including, but not limited to, arsenic, chromium, cadmium and nickel affect cellular organelles and enzymes involved in metabolism, detoxification, and damage repair [139, 153]. Heavy metal-induced increases in oxidative stress alter mitochondrial permeability and swelling, mutates mtDNA, reduces ATP synthesis, inhibits respiratory chain complexes, and increases apoptotic cell death [127, 154–160]. These metals also affect tumor suppressor genes, DNA repair processes, and enzymatic activities concerned with metabolism and oxidative damage [161]; all can contribute to carcinogenesis [140, 153, 162].
Mutations of genomic DNA capable of activating oncogenes or inactivating tumor suppressor genes are key features of tumor initiation, which is further facilitated by transcription factor activation, amplification of oncogenes, and recombination [163–166]; metals affect the four stages of carcinogenesis - initiation, promotion, progression, and metastasis. Although superoxide and hydroxyl radical formation can be caused by iron, copper, chromium and cobalt, iron appears to be of greatest importance. Free pools of copper are too low to produce free radicals via Fenton chemistry reactions. Cadmium, nickel and mercury cannot directly catalyze Fenton-type reactions and their main role in toxicity is depleting glutathione and binding to sulfhydryl groups of proteins [167–169]. In contrast, iron, chromium and cobalt can undergo redox-cycling reactions leading to oxidative stress [167], but iron must be present for arsenic to be oxidized [167, 170]. Thus, with regard to oxidative stress, iron is critical for excess ROS production and redox cycling. Indeed, the vulnerability of astrocytes to oxidative stress, loss of mitochondrial membrane potential and ATP depletion have been shown to be closely correlated to the iron content [171].
Occupational exposures to different heavy metals can lead to increases in brain cancer [172, 173]. One long term study of iron, lead, cadmium, and chromium that spanned several decades found increased risks of brain tumors to be highest for people with an occupation that exposed them to iron; the standardized incidence ratio was 2.15, which means there was a 115% higher incidence of observed cases than would otherwise be expected [174]. In the same study, cancer incidence was increased by 88% in people working with chromium, 33% in people working with lead, and 47% in people working with cadmium; no differences were found for people working with arsenic and there was a 51% increase for nickel [174]. This study linking iron and brain malignancy is especially strong because the cohort study included 413,877 people, over 183 different job titles, 25 different agents including asbestos and engine exhaust, and the robust finding that the highest brain tumor incidence was in people that had occupational exposures to iron [174].
Toxicity of Iron Accumulation in the Brain
The accumulation of iron in brain can induce neuronal damage. Even after being bound to ferritin, iron in the form of Fe2+ can be released from ferritin; this release is potentiated when the extracellular fluid is acidic [40] and in the presence of superoxide radicals [175] or ascorbate [176]. Iron toxicity occurs when the size of the labile iron pool exceeds the ability of ferritin to sequester iron [177] and is mediated by the increased production of ROS. When Fe3+ is reduced by ascorbate or other reductants, Fe2+ is regenerated and re-enters the oxidation-reduction cycle; a process known as redox cycling. Thus, there is this continuous production of ROS that is sustained and can continuously damage the cell; the quantity of the highly reactive hydroxyl radical is directly proportional to the concentration of Fe2+ [105]; the oxidative capacity of Fe2+ is 5 times greater than that of Fe3+ [178]. Thus, free radical production may overwhelm the cell’s antioxidant mechanisms, and because brain has low antioxidant defenses it is especially vulnerable to metal toxicity [42, 179, 180].
Numerous studies have emphasized the connection between dysregulation of iron metabolism including uptake, storage, and export, and human diseases including cancer [14, 16, 181, 182]. In cancer cells, however, disruption of iron metabolism occurs at the different steps of tumor initiation, progression and metastasis. Iron is very much involved in transformation of malignant glioblastoma cells. The ZIP14 (a metal-ion transporter) and the TfR iron uptake genes are both upregulated, STEAP3 is upregulated leading to an increase in Fe2+, and Dcytb, which is a ferric reductase enzyme that catalyzes the reduction of Fe3+ to Fe2+ required for dietary iron absorption in the duodenum of mammals is upregulated. The upregulated ZIP14 expression in GBM is associated with a significant shortening of patient survival rates [29].
Changes in iron metabolism are now considered by many to be metabolic and immunologic hallmarks of cancer, and further studies are required to determine how alterations of this element might affect tumorigenesis and progression. Clearly, cancer cells require large quantities of iron, and exhibit a strong dependence on iron, a situation that has been termed the “iron addiction” of cancer [183]. Tumor iron demands exist especially for proliferation [184, 185]. Specifically, the increased expression of the TfR has been shown to affect cancer cell proliferation, migration, invasion, and metastasis [186]. These findings suggest that TfR may be a target for cancer therapy. Furthermore, iron reductase and members of STEAP family are important for iron uptake and reduction in endolysosomes [187] and numerous studies have demonstrated altered expressions of STEAP 1–4 in different types of tumor including gliomas [188]. STEAP3 was highly expressed in malignant gliomas, especially in the mesenchymal glioma molecular subtype cell as well as isocitrate dehydrogenase 1/2 (IDH1/2) wild-type gliomas. Expression levels of STEAP3 in gliomas correlated inversely with patient survival [188].
Iron is also essential in forming lipid peroxyl radicals which can become cytotoxic aldehydes that can react with DNA and have damaging effects [189]. Moreover, several iron-dependent enzymes can indirectly produce large amount of ROS including superoxide radicals and peroxides. For example, cytochrome P450 enzymes, nitric oxide synthases, NADPH oxidases, and lipoxygenases are some of the most important enzymes involved in ROS generation [190, 191]. In a normal state, the antioxidant enzymes including glutathione peroxidase, superoxide dismutase and catalases intervene in detoxifying ROS [192], but become overwhelmed in a malignant state.
Astrocytes are more resilient to iron toxicity than neurons
Excess iron and hemoglobin are more toxic to neurons than they are to glial cells [193]. Astrocytes are the most abundant cell type in brain and help maintain an extracellular environment that protects neurons [194, 195]. Among numerous roles, astrocytes are more resistant than neural cells to ROS and iron-accumulated toxicity in brain [193, 196]. Astrocytes have high levels of antioxidants such as metallothionein, reduced glutathione, and manganese superoxide dismutase that all protect against metal-induced toxicity [197–199].
Endolysosome Iron, Mitochondrial Dysfunction, DNA Damage, and Carcinogenesis
Endolysosomes and the labile iron pool
Endolysosomes are acidic organelles containing more than 60 hydrolases, contain regulated stores of hydrogen (H+) ions, and have a vacuolar ATPase (v-ATPase) enzyme, which is the main proton pump responsible for maintaining the acidic lumen. v-ATPase has been identified as a novel therapeutic target for glioblastoma [200, 201]. Endolysosomes contain readily releasable stores of cations, including Fe2+. Although much is known about how Fe2+ is formed in endolysosomes and how that Fe2+ is delivered to the cytosolic pool for storage and use, less is known about the labile pool of iron [202, 203].
The cellular labile iron pool largely resides within the cytosol; however, it may exist in additional subcellular spaces and organelles. In rat liver endothelial cells, chelatable iron concentrations were 7.3 μM in the cytosol, 9.2 μM in mitochondria, 11.8 μM in the nucleus, and 16 μM in endolysosomes [204]. The heterogeneity among the labile iron pools and findings of the highest concentrations of labile iron in endolysosomes has implications for oxidative stress and the actions of iron chelators [204]. Further evidence of the role of labile iron in endolysosomes comes from mineral iron drugs used to treat iron deficiency, such as iron-dextran. Iron-dextran complexes have been shown to induce cancer tumor formation [205]. Because dextran is endocytosed, the administered iron-dextran accumulated in endolysosomes. Following endocytosis, the iron dissociates from the dextran, and release of iron from endolysosomes into the cytosol stimulates the formation of ROS [206, 207]. Of note, iron-dextran is classified as a Group 2B “possible carcinogenic to humans” on the IARC monograph.
Deferoxamine is specific to endolysosomes
Deferoxamine (DFO), specifically chelates the endolysosome labile iron pool because it cannot diffuse across the plasma membrane. DFO instead enters cells via fluid phase-endocytosis, and accumulates and remains in endolysosomes [203, 208–212]. DFO is a strong hydrophilic iron chelator that firmly binds all six coordinates of iron and prevents Fe2+/Fe3+ redox-cycling [203]. Furthermore, DFO is capable of crossing the BBB and has been shown to inhibit glioblastoma proliferation [213, 214]. Because DFO can chelate other metals such as aluminum and chromium [215, 216], DFO may find use against metal toxicity.
Radiation induced lysosomal membrane permeability
Radiation is used clinically for diagnostic and therapeutic reasons [217]. However, repeated exposure to radiation can cause increased lysosomal membrane permeabilization (LMP), increased release of endolysosome iron into the cytosol, and cell death (Figure 2). Drugs that induce LMP can cause similar effects [218, 219]. Radiation-induced apoptosis was largely prevented when endolysosome iron was chelated by DFO [220]. Thus, certain treatments and drugs can disrupt endolysosomes, promote excess intracellular labile iron, and increase levels of ROS and oxidative stress. Such findings led the IARC to classify X-ray radiation and γ-radiation into Group 1 “carcinogenic to humans” for the brain [221].
Drug-induced de-acidification of endolysosomes release cations including Fe2+
Numerous drugs including chloroquine (CQ), fluoxetine, imipramine, dimebon, tamoxifen, rotenone, chlorpromazine, amitriptyline, and verapamil can de-acidify the lumen of endolysosomes (Figure 2) [222, 223]. Lysosomotropic weak bases, which de-acidify endolysosomes, release endolysosome calcium into the cytosol [224]. This is consistent with findings that collapse of endolysosome transmembrane pH gradients with monensin causes an increase in cytosolic calcium levels [224]. In addition, free labile iron in endolysosomes is released into the cytosol upon bafilomycin A1 treatment, a known v-ATPase proton pump inhibitor that de-acidifies endolysosomes (Figure 2) [201, 206] and bafilomycin A1-induced iron release from endolysosomes was decreased by DFO [206]. Thus, regulating the pH of endolysosomes may result in new therapeutic strategies [200].
Ethanol (EtOH) is a major constituent of alcohol beverages and induces brain damage by triggering inflammatory processes in glial cells through activation of Toll-like receptor 4 (TLR4) signaling [225]. The IARC has alcoholic beverages classified as Group 1 carcinogens for certain cancers, and TLR4 participates in ethanol-induced de-acidification of endolysosomes [226]. Additionally, smoking tobacco is associated with endolysosome stress, mitochondrial stress, and swelling of neural stem cells [227]. The increase in endolysosome pH by nicotine, likely due to it being a weak base and activating two-pore channels [228], can diffuse across the cell membrane and accumulate in endolysosomes by proton trapping [229, 230]. As noted earlier, the de-acidification of endolysosomes results in labile iron release into the cytosol and increased ROS production [206].
Endolysosome iron causes mitochondrial fragmentation and dysfunction
Mitochondria are dynamically regulated by fission (fragmentation) and fusion events [231]. Molecular mechanisms that control mitochondrial fission and fusion help preserve mitochondrial integrity through mitophagy and disposal of dysfunctional mitochondria. Mitochondrial fragmentation, also known as the thread-grain transition, is induced by oxidative stress, occurs during apoptosis, and results in the presence of numerous smaller mitochondria [232]; [233]. Mitochondrial fragmentation and mitophagy function to maintain healthy mitochondria; mitophagy removes mitochondria that have mitochondrial DNA (mtDNA) mutations and this helps reduce mtDNA heterogeneity and mitochondrial heteroplasmy [234].
Iron accumulation promotes increases in ROS levels, mitochondrial fragmentation, and cellular apoptosis, a chain of events that was blocked by DFO [235]. Similar results were demonstrated in hippocampal neuronal cells; FAC-induced iron accumulation triggered mitochondrial fragmentation and apoptosis that was prevented by DFO [236]. Many drugs can de-acidify endolysosomes, induce mitochondrial fragmentation, and cause loss of mitochondrial membrane potential including CQ, artesunate, diazepam, oligomycin, antimycin A, and rotenone. These effects that were blocked by DFO [223, 237–242]. Thus, endolysosome iron release appears to be an important event upstream of increased mitochondrial ROS, fragmentation, dysfunction, and apoptosis.
Effects of mitochondrial fragmentation on disease
Mitochondrial fragmentation causes mitochondria to divide into smaller, more numerous vesicles and this phenomenon has been implicated in oxidative stress-related apoptosis and various pathological phenotypes including initiation of brain tumors (Figure 1) [242–245]. Glioblastomas have an increased frequency of mitochondrial fission [246–248] and suggest that mitochondrial fragmentation may be more pronounced in stem cells [245]. DRP1, a regulator of mitochondrial fission, helps maintain the tumorigenic potential of brain tumor initiating cells (BTIC) and reductions in DRP1 causes loss of the BTIC phenotype and tumor formation [245]. In contrast, mitochondrial division inhibitor 1 (Mdivi-1) inhibits mitochondrial division and decreases BTIC tumor formation [245]; Mdivi-1 acidified endolysosomes and genetic inhibition of DRP1 with si-DRP1 de-acidified endolysosomes [249]. Although total levels of DRP1 are similar in normal and neoplastic brain tissues, phosphorylation of DRP1 (DRP1S616) is increased in glioblastomas and there is a strong inverse correlation between phosphorylation of DRP1 (S616) and poor glioblastoma patient survival [245].
Mitochondrial ROS promotes mtDNA mutations and carcinogenesis
mtDNA mutations in cancer occur in genes of complexes I, III, IV, and V and affect oxidative phosphorylation and the regulation of ROS [250]. Although still actively investigated, somatic and germline mtDNA mutations have been reported in numerous different cancer types including astrocytic tumors [251–254]. The importance of mtDNA mutations in neoplastic transformation [251, 255] is supported by findings that exchanging cancer cell mtDNA with other sources of mtDNA changes cancer cell phenotypes including production of ROS (Figure 2) [256–258]. Elevated levels of mitochondrial ROS are toxic, are strong mitogens, and damage mitochondria and mtDNA [250].
Endolysosome iron mediates oxidative-stress-induced DNA damage
DNA mutations known to induce malignancies can be a consequence of cellular oxidative stress and the generation of highly reactive and short-lived hydroxyl radicals in close proximity to DNA [259–263]. Similar to the labile iron pool found in the cytosol, a chelatable and labile iron pool exists in mammalian nuclei [204], and transition metals and hydrogen peroxide can induce hydroxylation and other DNA modifications [261, 262, 264–266]. The specific mechanisms by which iron enters the nucleus remain unclear. Labile iron bound to low molecular weight ligands may freely diffuse through nuclear pores and/or get actively (ATP-dependent) transported across nuclear membranes; nuclear uptake of iron can be blocked by DFO [267]. These findings suggest that endolysosomes are the source of nuclear free labile iron. DFO is able to protect against oxidative disruption induced by hydrogen peroxide and prevents strand breaks in nuclear DNA induced by hydrogen peroxide [268]. Thus, iron from endolysosomes can be translocated to the nucleus after endolysosome membrane rupture [268]; a form of lysosome membrane permeabilization (LMP) (Figure 2).
Oxidative DNA damage leads to DNA mutations and cells harboring DNA mutations that escape programmed cell death are capable of initiating tumors. Oxidative DNA damage of guanine, due to its low oxidation potential, leads to the formation of 8-oxo-7,8-dihydroguanine (8-oxo-dG) and incorporation into DNA [269]. In addition, oxidative stress and DNA damage via 8-oxo-dG directly inhibits telomerase transcriptional activity of TERT [269]. There is a direct correlation between increased stores of iron in the body with increased risks of cancer [270, 271], the mutagenicity of iron is mediated by ROS [272–274].
Human glioblastoma arises from cells in the subventricular zone (SVZ) that contain key driver mutations (p53, PTEN,, EGFR, and TERT) [275]; astrocyte-like neural stem cells (NSC) migrating from the SVZ led to the development of malignant gliomas in distant brain regions of the cortex. The loss of TERT, via genetic mutation or downstream of DNA damage via 8-oxo-dG, may be an early genetic event by which NSCs in the SVZ acquire more driver mutations that eventually develop into glioblastoma [275]. However, it remains uncertain whether the presence of 8-oxo-dG in DNA is sufficient to cause tumor formation in part because other pathological conditions associated with elevated oxidative DNA damage have no association with increased carcinogenesis.
Conclusions
Numerous risk factors including stress, tobacco, environmental pollutants, radiation, viral infection, diet, and infection are linked with chronic diseases such as cancer through the production of ROS [276], and increased levels of ferrous (Fe2+) iron can generate reactive oxygen species (ROS). ROS activates nuclear factor kappa-light-chain-enhancer of activated B cells [NF-κB], hypoxia-inducible factor-1α (HIF-1α), and signal transducer and activator of transcription 3 (STAT3) resulting in increased expression of proteins that increase inflammation, cellular transformation, cell survival, tumor cell proliferation and invasion, angiogenesis, and metastasis. ROS is also known to damage cellular proteins, lipids and DNA, and contribute to carcinogenesis [277, 278]. Chronic inflammation, which is one of the major mediators of tumorigenesis, is regulated by ROS and an association between ROS, DNA mutations, and malignant transformation has been clearly shown from studies of gain-of-function mutations in oncogenes and loss-of-function mutations in tumor suppressor genes [167, 279–281].
As discussed above, many factors contribute to elevated iron and other heavy metal levels in brain and endolysosomes contain a relatively large concentration of free labile iron. Once released from endolysosomes, iron can affect many different organelles through Fenton chemistry reactions and the production of ROS. Dispersion of the endolysosome labile iron can be caused by drug-induced de-acidification of endolysosomes, radiation or an overabundance of iron accumulation. Fe2+ released from endolysosomes can damage mitochondria and cause oxidative damage to nuclear DNA. However, despite links between dysregulated iron metabolism and carcinogenesis, relatively little has been done to directly test whether modulation of iron may be a valuable treatment strategy. Iron chelators initially used to treat acute iron overload have received some attention due to high levels of iron in cancer cells [282], and preliminary studies with these agents such as DFO have shown that they may have a role in inhibiting tumor growth [282, 283]. Although clinical trials of DFO in cancer therapy are limited, some positive results have been published. DFO was found to have a combined 88% full or partial response of tumor decreases in neuroblastoma in a clinical trial with 7% of cases progressing, However, significant side effects were reported that would need to be addressed before advancing these therapies further and argue for a more targeted approach. Another study found chelating iron with DFO enhanced fluorescence contrast to provide a more effective fluorescence-guided resection of brain tumors [284, 285]. Overall, while many key questions remain, we believe that there is sufficient evidence to suggest that regulation of iron homeostasis of endolysosomes plays a key role in carcinogenesis in the brain, and this may warrant additional consideration of therapeutic strategies which modulate iron for cancer treatment and prevention.
Acknowledgements
Grant support from the NIH (ES022030) awarded to J.E.O. is acknowledged. JDG acknowledges research support from the National Institute of General Medical Sciences under award numbers P30GM100329 and U54GM115458, the National Institute of Mental Health under award numbers R01MH100972, R01MH105329 and R01MH119000, the National Institute of Neurological Diseases and Stroke (NINDS) under award number 2R01NS065957, and the National Institute of Drug Abuse under award number 2R01DA032444.
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].Jordan TH, Structural geology of the Earth’s interior, Proceedings of the National Academy of Sciences 76(9) (1979) 4192–4200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Yehuda S, Brain iron deficiency. Biochemistry and behavior, Brain iron, neurochemical and behavioral aspects (1988). [Google Scholar]
- [3].Laskey J, Webb I, Schulman HM, Ponka P, Evidence that transferrin supports cell proliferation by supplying iron for DNA synthesis, Exp Cell Res 176(1) (1988) 87–95. [DOI] [PubMed] [Google Scholar]
- [4].Hill JM, Iron concentration reduced in ventral pallidum, globus pallidus, and substantia nigra by GABA-transaminase inhibitor, gamma-vinyl GABA, Brain Res 342(1) (1985) 18–25. [DOI] [PubMed] [Google Scholar]
- [5].Connor JR, Pavlick G, Karli D, Menzies SL, Palmer C, A histochemical study of iron-positive cells in the developing rat brain, J Comp Neurol 355(1) (1995) 111–23. [DOI] [PubMed] [Google Scholar]
- [6].Youdim MB, Ben-Shachar D, Yehuda S, Riederer P, The role of iron in the basal ganglion, Adv Neurol 53 (1990) 155–62. [PubMed] [Google Scholar]
- [7].Hulet SW, Hess EJ, Debinski W, Arosio P, Bruce K, Powers S, Connor JR, Characterization and distribution of ferritin binding sites in the adult mouse brain, J Neurochem 72(2) (1999) 868–74. [DOI] [PubMed] [Google Scholar]
- [8].Kerins MJ, Ooi A, The Roles of NRF2 in Modulating Cellular Iron Homeostasis, Antioxid Redox Signal 29(17) (2018) 1756–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Qian Z-M, Ke Y, Brain iron transport, Biological Reviews 94(5) (2019) 1672–1684. [DOI] [PubMed] [Google Scholar]
- [10].Rouault TA, Tong W-H, Iron–sulphur cluster biogenesis and mitochondrial iron homeostasis, Nature Reviews Molecular Cell Biology 6(4) (2005) 345–351. [DOI] [PubMed] [Google Scholar]
- [11].Andrews NC, Forging a field: the golden age of iron biology, Blood 112(2) (2008) 219–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Ali MK, Kim RY, Karim R, Mayall JR, Martin KL, Shahandeh A, Abbasian F, Starkey MR, Loustaud-Ratti V, Johnstone D, Milward EA, Hansbro PM, Horvat JC, Role of iron in the pathogenesis of respiratory disease, Int J Biochem Cell Biol 88 (2017) 181–195. [DOI] [PubMed] [Google Scholar]
- [13].Dielschneider RF, Henson ES, Gibson SB, Lysosomes as Oxidative Targets for Cancer Therapy, Oxid Med Cell Longev 2017 (2017) 3749157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Torti SV, Torti FM, Iron and cancer: more ore to be mined, Nat Rev Cancer 13(5) (2013) 342–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Fonseca-Nunes A, Jakszyn P, Agudo A, Iron and cancer risk--a systematic review and meta-analysis of the epidemiological evidence, Cancer Epidemiol Biomarkers Prev 23(1) (2014) 12–31. [DOI] [PubMed] [Google Scholar]
- [16].Manz DH, Blanchette NL, Paul BT, Torti FM, Torti SV, Iron and cancer: recent insights, Ann N Y Acad Sci 1368(1) (2016) 149–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Brawn K, Fridovich I, DNA strand scission by enzymically generated oxygen radicals, Arch Biochem Biophys 206(2) (1981) 414–9. [DOI] [PubMed] [Google Scholar]
- [18].Jackson JH, Schraufstatter IU, Hyslop PA, Vosbeck K, Sauerheber R, Weitzman SA, Cochrane CG, Role of oxidants in DNA damage. Hydroxyl radical mediates the synergistic DNA damaging effects of asbestos and cigarette smoke, J Clin Invest 80(4) (1987) 1090–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Schraufstätter I, Hyslop PA, Jackson JH, Cochrane CG, Oxidant-induced DNA damage of target cells, J Clin Invest 82(3) (1988) 1040–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Jackson JH, Potential molecular mechanisms of oxidant-induced carcinogenesis, Environ Health Perspect 102 Suppl 10(Suppl 10) (1994) 155–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Imlay JA, Linn S, DNA damage and oxygen radical toxicity, Science 240(4857) (1988) 1302–9. [DOI] [PubMed] [Google Scholar]
- [22].Fuss JO, Tsai CL, Ishida JP, Tainer JA, Emerging critical roles of Fe-S clusters in DNA replication and repair, Biochim Biophys Acta 1853(6) (2015) 1253–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Leftin A, Ben-Chetrit N, Klemm F, Joyce JA, Koutcher JA, Iron imaging reveals tumor and metastasis macrophage hemosiderin deposits in breast cancer, PLoS One 12(9) (2017) e0184765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Behin A, Hoang-Xuan K, Carpentier AF, Delattre JY, Primary brain tumours in adults, Lancet 361(9354) (2003) 323–31. [DOI] [PubMed] [Google Scholar]
- [25].de Robles P, Fiest KM, Frolkis AD, Pringsheim T, Atta C, St Germaine-Smith C, Day L, Lam D, Jette N, The worldwide incidence and prevalence of primary brain tumors: a systematic review and meta-analysis, Neuro Oncol 17(6) (2015) 776–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Poon MTC, Sudlow CLM, Figueroa JD, Brennan PM, Longer-term (>/= 2 years) survival in patients with glioblastoma in population-based studies pre- and post-2005: a systematic review and meta-analysis, Scientific reports 10(1) (2020) 11622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Qian ZM, Tang PL, Mechanisms of iron uptake by mammalian cells, Biochim Biophys Acta 1269(3) (1995) 205–14. [DOI] [PubMed] [Google Scholar]
- [28].Liuzzi JP, Aydemir F, Nam H, Knutson MD, Cousins RJ, Zip14 (Slc39a14) mediates non-transferrin-bound iron uptake into cells, Proc Natl Acad Sci U S A 103(37) (2006) 13612–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Chen Y, Fan Z, Yang Y, Gu C, Iron metabolism and its contribution to cancer (Review), Int J Oncol 54(4) (2019) 1143–1154. [DOI] [PubMed] [Google Scholar]
- [30].González A, Gálvez N, Martín J, Reyes F, Pérez-Victoria I, Dominguez-Vera JM, Identification of the key excreted molecule by Lactobacillus fermentum related to host iron absorption, Food Chem 228 (2017) 374–380. [DOI] [PubMed] [Google Scholar]
- [31].Li Y, Jiang H, Huang G, Protein Hydrolysates as Promoters of Non-Haem Iron Absorption, Nutrients 9(6) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Staroń R, Lipiński P, Lenartowicz M, Bednarz A, Gajowiak A, Smuda E, Krzeptowski W, Pieszka M, Korolonek T, Hamza I, Swinkels DW, Van Swelm RPL, Starzyński RR, Dietary hemoglobin rescues young piglets from severe iron deficiency anemia: Duodenal expression profile of genes involved in heme iron absorption, PLOS ONE 12(7) (2017) e0181117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Coates TD, Physiology and pathophysiology of iron in hemoglobin-associated diseases, Free Radic Biol Med 72 (2014) 23–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Donovan A, Lima CA, Pinkus JL, Pinkus GS, Zon LI, Robine S, Andrews NC, The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis, Cell Metab 1(3) (2005) 191–200. [DOI] [PubMed] [Google Scholar]
- [35].Sokolov AV, Voynova IV, Kostevich VA, Vlasenko AY, Zakharova ET, Vasilyev VB, Comparison of interaction between ceruloplasmin and lactoferrin/transferrin: to bind or not to bind, Biochemistry (Moscow) 82(9) (2017) 1073–1078. [DOI] [PubMed] [Google Scholar]
- [36].Gerlach M, Ben-Shachar D, Riederer P, Youdim MB, Altered brain metabolism of iron as a cause of neurodegenerative diseases?, J Neurochem 63(3) (1994) 793–807. [DOI] [PubMed] [Google Scholar]
- [37].Mulligan M, Althaus B, Linder MC, Non-ferritin, non-heme iron pools in rat tissues, Int J Biochem 18(9) (1986) 791–8. [DOI] [PubMed] [Google Scholar]
- [38].Weaver J, Pollack S, Zhan H, Low molecular weight iron from guinea pig reticulocytes isolated by Sephadex G-25 chromatography, Eur J Haematol 43(4) (1989) 321–7. [DOI] [PubMed] [Google Scholar]
- [39].Triggs WJ, Willmore LJ, In vivo lipid peroxidation in rat brain following intracortical Fe2+ injection, J Neurochem 42(4) (1984) 976–80. [DOI] [PubMed] [Google Scholar]
- [40].Bishop GM, Robinson SR, Quantitative analysis of cell death and ferritin expression in response to cortical iron: implications for hypoxia-ischemia and stroke, Brain Res 907(1–2) (2001) 175–87. [DOI] [PubMed] [Google Scholar]
- [41].Rolfs A, Hediger MA, Metal ion transporters in mammals: structure, function and pathological implications, J Physiol 518(Pt 1) (1999) 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Crichton RR, Wilmet S, Legssyer R, Ward RJ, Molecular and cellular mechanisms of iron homeostasis and toxicity in mammalian cells, J Inorg Biochem 91(1) (2002) 9–18. [DOI] [PubMed] [Google Scholar]
- [43].Morgan EH, Chelator-mediated iron efflux from reticulocytes, Biochim Biophys Acta 733(1) (1983) 39–50. [DOI] [PubMed] [Google Scholar]
- [44].Martines AM, Masereeuw R, Tjalsma H, Hoenderop JG, Wetzels JF, Swinkels DW, Iron metabolism in the pathogenesis of iron-induced kidney injury, Nat Rev Nephrol 9(7) (2013) 385–98. [DOI] [PubMed] [Google Scholar]
- [45].Ohgami RS, Campagna DR, Greer EL, Antiochos B, McDonald A, Chen J, Sharp JJ, Fujiwara Y, Barker JE, Fleming MD, Identification of a ferrireductase required for efficient transferrin-dependent iron uptake in erythroid cells, Nat Genet 37(11) (2005) 1264–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Ohgami RS, Campagna DR, McDonald A, Fleming MD, The Steap proteins are metalloreductases, Blood 108(4) (2006) 1388–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Gordan R, Wongjaikam S, Gwathmey JK, Chattipakorn N, Chattipakorn SC, Xie L-H, Involvement of cytosolic and mitochondrial iron in iron overload cardiomyopathy: an update, Heart failure reviews 23(5) (2018) 801–816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Zecca L, Youdim MB, Riederer P, Connor JR, Crichton RR, Iron, brain ageing and neurodegenerative disorders, Nat Rev Neurosci 5(11) (2004) 863–73. [DOI] [PubMed] [Google Scholar]
- [49].Chiou B, Neal EH, Bowman AB, Lippmann ES, Simpson IA, Connor JR, Endothelial cells are critical regulators of iron transport in a model of the human blood–brain barrier, Journal of Cerebral Blood Flow & Metabolism 39(11) (2018) 2117–2131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Duck KA, Neely EB, Simpson IA, Connor JR, A role for sex and a common HFE gene variant in brain iron uptake, Journal of Cerebral Blood Flow & Metabolism 38(3) (2017) 540–548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Simpson IA, Ponnuru P, Klinger ME, Myers RL, Devraj K, Coe CL, Lubach GR, Carruthers A, Connor JR, A novel model for brain iron uptake: introducing the concept of regulation, J Cereb Blood Flow Metab 35(1) (2015) 48–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Khan AI, Liu J, Dutta P, Iron transport kinetics through blood-brain barrier endothelial cells, Biochimica et Biophysica Acta (BBA) - General Subjects 1862(5) (2018) 1168–1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].McCarthy RC, Kosman DJ, Iron transport across the blood-brain barrier: development, neurovascular regulation and cerebral amyloid angiopathy, Cell Mol Life Sci 72(4) (2015) 709–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Jefferies WA, Brandon MR, Hunt SV, Williams AF, Gatter KC, Mason DY, Transferrin receptor on endothelium of brain capillaries, Nature 312(5990) (1984) 162–3. [DOI] [PubMed] [Google Scholar]
- [55].Bradbury MW, Transport of iron in the blood-brain-cerebrospinal fluid system, J Neurochem 69(2) (1997) 443–54. [DOI] [PubMed] [Google Scholar]
- [56].Malecki EA, Devenyi AG, Beard JL, Connor JR, Existing and emerging mechanisms for transport of iron and manganese to the brain, J Neurosci Res 56(2) (1999) 113–22. [DOI] [PubMed] [Google Scholar]
- [57].Aisen P, Entry of iron into cells: a new role for the transferrin receptor in modulating iron release from transferrin, Ann Neurol 32 Suppl (1992) S62–8. [DOI] [PubMed] [Google Scholar]
- [58].Qian ZM, Shen X, Brain iron transport and neurodegeneration, Trends Mol Med 7(3) (2001) 103–8. [DOI] [PubMed] [Google Scholar]
- [59].McCarthy RC, Kosman DJ, Ferroportin and exocytoplasmic ferroxidase activity are required for brain microvascular endothelial cell iron efflux, J Biol Chem 288(24) (2013) 17932–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].McCarthy RC, Kosman DJ, Glial Cell Ceruloplasmin and Hepcidin Differentially Regulate Iron Efflux from Brain Microvascular Endothelial Cells, PLOS ONE 9(2) (2014) e89003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Burkhart A, Skjørringe T, Johnsen KB, Siupka P, Thomsen LB, Nielsen MS, Thomsen LL, Moos T, Expression of Iron-Related Proteins at the Neurovascular Unit Supports Reduction and Reoxidation of Iron for Transport Through the Blood-Brain Barrier, Mol Neurobiol 53(10) (2016) 7237–7253. [DOI] [PubMed] [Google Scholar]
- [62].Moos T, Morgan EH, A morphological study of the developmentally regulated transport of iron into the brain, Dev Neurosci 24(2–3) (2002) 99–105. [DOI] [PubMed] [Google Scholar]
- [63].Banks WA, Kastin AJ, Fasold MB, Barrera CM, Augereau G, Studies of the slow bidirectional transport of iron and transferrin across the blood-brain barrier, Brain Res Bull 21(6) (1988) 881–5. [DOI] [PubMed] [Google Scholar]
- [64].Moos T, Increased accumulation of transferrin by motor neurons of the mouse mutant progressive motor neuronopathy (pmn/pmn), J Neurocytol 24(5) (1995) 389–98. [DOI] [PubMed] [Google Scholar]
- [65].Graeber MB, Raivich G, Kreutzberg GW, Increase of transferrin receptors and iron uptake in regenerating motor neurons, J Neurosci Res 23(3) (1989) 342–5. [DOI] [PubMed] [Google Scholar]
- [66].Giometto B, Bozza F, Argentiero V, Gallo P, Pagni S, Piccinno MG, Tavolato B, Transferrin receptors in rat central nervous system. An immunocytochemical study, J Neurol Sci 98(1) (1990) 81–90. [DOI] [PubMed] [Google Scholar]
- [67].Dickinson TK, Connor JR, Immunohistochemical analysis of transferrin receptor: regional and cellular distribution in the hypotransferrinemic (hpx) mouse brain, Brain Res 801(1–2) (1998) 171–81. [DOI] [PubMed] [Google Scholar]
- [68].Qian ZM, Liao QK, To Y, Ke Y, Tsoi YK, Wang GF, Ho KP, Transferrin-bound and transferrin free iron uptake by cultured rat astrocytes, Cell Mol Biol (Noisy-le-grand) 46(3) (2000) 541–8. [PubMed] [Google Scholar]
- [69].Du F, Qian C, Qian ZM, Wu XM, Xie H, Yung WH, Ke Y, Hepcidin directly inhibits transferrin receptor 1 expression in astrocytes via a cyclic AMP-protein kinase A pathway, Glia 59(6) (2011) 936–45. [DOI] [PubMed] [Google Scholar]
- [70].Rathore KI, Redensek A, David S, Iron homeostasis in astrocytes and microglia is differentially regulated by TNF-α and TGF-β1, Glia 60(5) (2012) 738–50. [DOI] [PubMed] [Google Scholar]
- [71].Rathnasamy G, Ling EA, Kaur C, Hypoxia inducible factor-1α mediates iron uptake which induces inflammatory response in amoeboid microglial cells in developing periventricular white matter through MAP kinase pathway, Neuropharmacology 77 (2014) 428–40. [DOI] [PubMed] [Google Scholar]
- [72].Todorich B, Zhang X, Slagle-Webb B, Seaman WE, Connor JR, Tim-2 is the receptor for H-ferritin on oligodendrocytes, J Neurochem 107(6) (2008) 1495–505. [DOI] [PubMed] [Google Scholar]
- [73].Todorich B, Zhang X, Connor JR, H-ferritin is the major source of iron for oligodendrocytes, Glia 59(6) (2011) 927–35. [DOI] [PubMed] [Google Scholar]
- [74].Hulet SW, Heyliger SO, Powers S, Connor JR, Oligodendrocyte progenitor cells internalize ferritin via clathrin-dependent receptor mediated endocytosis, J Neurosci Res 61(1) (2000) 52–60. [DOI] [PubMed] [Google Scholar]
- [75].Han J, Day JR, Connor JR, Beard JL, Gene expression of transferrin and transferrin receptor in brains of control vs. iron-deficient rats, Nutr Neurosci 6(1) (2003) 1–10. [PubMed] [Google Scholar]
- [76].Ke Y, Ming Qian Z, Iron misregulation in the brain: a primary cause of neurodegenerative disorders, Lancet Neurol 2(4) (2003) 246–53. [DOI] [PubMed] [Google Scholar]
- [77].Schipper HM, Bernier L, Mehindate K, Frankel D, Mitochondrial iron sequestration in dopamine-challenged astroglia: role of heme oxygenase-1 and the permeability transition pore, J Neurochem 72(5) (1999) 1802–11. [DOI] [PubMed] [Google Scholar]
- [78].Huang E, Ong WY, Connor JR, Distribution of divalent metal transporter-1 in the monkey basal ganglia, Neuroscience 128(3) (2004) 487–96. [DOI] [PubMed] [Google Scholar]
- [79].Takeda A, Devenyi A, Connor JR, Evidence for non-transferrin-mediated uptake and release of iron and manganese in glial cell cultures from hypotransferrinemic mice, J Neurosci Res 51(4) (1998) 454–62. [DOI] [PubMed] [Google Scholar]
- [80].Leitner DF, Connor JR, Functional roles of transferrin in the brain, Biochim Biophys Acta 1820(3) (2012) 393–402. [DOI] [PubMed] [Google Scholar]
- [81].Kato Y, Ozawa S, Miyamoto C, Maehata Y, Suzuki A, Maeda T, Baba Y, Acidic extracellular microenvironment and cancer, Cancer cell international 13(1) (2013) 89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Stubbs M, McSheehy PM, Griffiths JR, Bashford CL, Causes and consequences of tumour acidity and implications for treatment, Mol Med Today 6(1) (2000) 15–9. [DOI] [PubMed] [Google Scholar]
- [83].Moos T, Morgan EH, The significance of the mutated divalent metal transporter (DMT1) on iron transport into the Belgrade rat brain, J Neurochem 88(1) (2004) 233–45. [DOI] [PubMed] [Google Scholar]
- [84].Selim MH, Ratan RR, The role of iron neurotoxicity in ischemic stroke, Ageing Res Rev 3(3) (2004) 345–53. [DOI] [PubMed] [Google Scholar]
- [85].Moos T, Morgan EH, Evidence for low molecular weight, non-transferrin-bound iron in rat brain and cerebrospinal fluid, J Neurosci Res 54(4) (1998) 486–94. [DOI] [PubMed] [Google Scholar]
- [86].Burdo JR, Menzies SL, Simpson IA, Garrick LM, Garrick MD, Dolan KG, Haile DJ, Beard JL, Connor JR, Distribution of divalent metal transporter 1 and metal transport protein 1 in the normal and Belgrade rat, J Neurosci Res 66(6) (2001) 1198–207. [DOI] [PubMed] [Google Scholar]
- [87].Wu LJ, Leenders AG, Cooperman S, Meyron-Holtz E, Smith S, Land W, Tsai RY, Berger UV, Sheng ZH, Rouault TA, Expression of the iron transporter ferroportin in synaptic vesicles and the blood-brain barrier, Brain Res 1001(1–2) (2004) 108–17. [DOI] [PubMed] [Google Scholar]
- [88].Zarruk JG, Berard JL, Passos dos Santos R, Kroner A, Lee J, Arosio P, David S, Expression of iron homeostasis proteins in the spinal cord in experimental autoimmune encephalomyelitis and their implications for iron accumulation, Neurobiol Dis 81 (2015) 93–107. [DOI] [PubMed] [Google Scholar]
- [89].Pelizzoni I, Zacchetti D, Smith CP, Grohovaz F, Codazzi F, Expression of divalent metal transporter 1 in primary hippocampal neurons: reconsidering its role in non-transferrin-bound iron influx, J Neurochem 120(2) (2012) 269–78. [DOI] [PubMed] [Google Scholar]
- [90].Loeffler DA, LeWitt PA, Juneau PL, Sima AA, Nguyen HU, DeMaggio AJ, Brickman CM, Brewer GJ, Dick RD, Troyer MD, Kanaley L, Increased regional brain concentrations of ceruloplasmin in neurodegenerative disorders, Brain Res 738(2) (1996) 265–74. [DOI] [PubMed] [Google Scholar]
- [91].Loeffler DA, Sima AA, LeWitt PA, Ceruloplasmin immunoreactivity in neurodegenerative disorders, Free Radic Res 35(2) (2001) 111–8. [DOI] [PubMed] [Google Scholar]
- [92].Klomp LW, Farhangrazi ZS, Dugan LL, Gitlin JD, Ceruloplasmin gene expression in the murine central nervous system, J Clin Invest 98(1) (1996) 207–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Klomp LW, Gitlin JD, Expression of the ceruloplasmin gene in the human retina and brain: implications for a pathogenic model in aceruloplasminemia, Hum Mol Genet 5(12) (1996) 1989–96. [DOI] [PubMed] [Google Scholar]
- [94].Hudson DM, Curtis SB, Smith VC, Griffiths TAM, Wong AYK, Scudamore CH, Buchan AMJ, MacGillivray RTA, Human hephaestin expression is not limited to enterocytes of the gastrointestinal tract but is also found in the antrum, the enteric nervous system, and pancreatic β-cells, American Journal of Physiology-Gastrointestinal and Liver Physiology 298(3) (2009) G425–G432. [DOI] [PubMed] [Google Scholar]
- [95].Ji C, Steimle BL, Bailey DK, Kosman DJ, The Ferroxidase Hephaestin But Not Amyloid Precursor Protein is Required for Ferroportin-Supported Iron Efflux in Primary Hippocampal Neurons, Cell Mol Neurobiol 38(4) (2018) 941–954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Schulz K, Vulpe CD, Harris LZ, David S, Iron efflux from oligodendrocytes is differentially regulated in gray and white matter, J Neurosci 31(37) (2011) 13301–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Hallgren B, Sourander P, The effect of age on the non-haemin iron in the human brain, J Neurochem 3(1) (1958) 41–51. [DOI] [PubMed] [Google Scholar]
- [98].Schenck JF, Zimmerman EA, High-field magnetic resonance imaging of brain iron: birth of a biomarker?, NMR Biomed 17(7) (2004) 433–45. [DOI] [PubMed] [Google Scholar]
- [99].Morris CM, Candy JM, Keith AB, Oakley AE, Taylor GA, Pullen RG, Bloxham CA, Gocht A, Edwardson JA, Brain iron homeostasis, J Inorg Biochem 47(3–4) (1992) 257–65. [DOI] [PubMed] [Google Scholar]
- [100].Koeppen AH, The history of iron in the brain, J Neurol Sci 134 Suppl (1995) 1–9. [DOI] [PubMed] [Google Scholar]
- [101].Aisen P, Wessling-Resnick M, Leibold EA, Iron metabolism, Curr Opin Chem Biol 3(2) (1999) 200–6. [DOI] [PubMed] [Google Scholar]
- [102].Theil EC, Ferritin: structure, gene regulation, and cellular function in animals, plants, and microorganisms, Annu Rev Biochem 56 (1987) 289–315. [DOI] [PubMed] [Google Scholar]
- [103].Wixom RL, Prutkin L, Munro HN, Hemosiderin: nature, formation, and significance, Int Rev Exp Pathol 22 (1980) 193–225. [PubMed] [Google Scholar]
- [104].Iancu TC, Ferritin and hemosiderin in pathological tissues, Electron Microsc Rev 5(2) (1992) 209–29. [DOI] [PubMed] [Google Scholar]
- [105].Núñez-Millacura C, Tapia V, Muñoz P, Maccioni RB, Núñez MT, An oxidative stress-mediated positive-feedback iron uptake loop in neuronal cells, J Neurochem 82(2) (2002) 240–8. [DOI] [PubMed] [Google Scholar]
- [106].Breuer W, Epsztejn S, Cabantchik ZI, Dynamics of the cytosolic chelatable iron pool of K562 cells, FEBS Lett 382(3) (1996) 304–8. [DOI] [PubMed] [Google Scholar]
- [107].Wagner KR, Sharp FR, Ardizzone TD, Lu A, Clark JF, Heme and Iron Metabolism: Role in Cerebral Hemorrhage, Journal of Cerebral Blood Flow & Metabolism 23(6) (2003) 629–652. [DOI] [PubMed] [Google Scholar]
- [108].Crowe A, Morgan EH, Iron and transferrin uptake by brain and cerebrospinal fluid in the rat, Brain Res 592(1–2) (1992) 8–16. [DOI] [PubMed] [Google Scholar]
- [109].Götz ME, Double K, Gerlach M, Youdim MB, Riederer P, The relevance of iron in the pathogenesis of Parkinson’s disease, Ann N Y Acad Sci 1012 (2004) 193–208. [DOI] [PubMed] [Google Scholar]
- [110].Beal MF, Mitochondrial dysfunction in neurodegenerative diseases, Biochim Biophys Acta 1366(1–2) (1998) 211–23. [DOI] [PubMed] [Google Scholar]
- [111].Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L, The role of iron in brain ageing and neurodegenerative disorders, Lancet Neurol 13(10) (2014) 1045–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Burdo JR, Martin J, Menzies SL, Dolan KG, Romano MA, Fletcher RJ, Garrick MD, Garrick LM, Connor JR, Cellular distribution of iron in the brain of the Belgrade rat, Neuroscience 93(3) (1999) 1189–1196. [DOI] [PubMed] [Google Scholar]
- [113].Zecca L, Gallorini M, Schünemann V, Trautwein AX, Gerlach M, Riederer P, Vezzoni P, Tampellini D, Iron, neuromelanin and ferritin content in the substantia nigra of normal subjects at different ages: consequences for iron storage and neurodegenerative processes, J Neurochem 76(6) (2001) 1766–73. [DOI] [PubMed] [Google Scholar]
- [114].Angelova DM, Brown DR, Iron, Aging, and Neurodegeneration, Metals 5(4) (2015). [Google Scholar]
- [115].Yan F, He N, Lin H, Li R, Iron deposition quantification: Applications in the brain and liver, J Magn Reson Imaging 48(2) (2018) 301–317. [DOI] [PubMed] [Google Scholar]
- [116].Ying W, Han SK, Miller JW, Swanson RA, Acidosis potentiates oxidative neuronal death by multiple mechanisms, J Neurochem 73(4) (1999) 1549–56. [DOI] [PubMed] [Google Scholar]
- [117].Levy YS, Streifler JY, Panet H, Melamed E, Offen D, Hemin-induced apoptosis in PC12 and neuroblastoma cells: implications for local neuronal death associated with intracerebral hemorrhage, Neurotoxicity research 4(7) (2002) 609–616. [DOI] [PubMed] [Google Scholar]
- [118].Kajikawa H, Ohta T, Yoshikawa Y, Funatsu N, Yamamoto M, Someda K, Cerebral vasospasm and hemoglobins—clinical and experimental studies, Neurologia medico-chirurgica 19(1) (1979) 61–71. [DOI] [PubMed] [Google Scholar]
- [119].Regan RF, Panter SS, Neurotoxicity of hemoglobin in cortical cell culture, Neuroscience letters 153(2) (1993) 219–222. [DOI] [PubMed] [Google Scholar]
- [120].Turner CP, Bergeron M, Matz P, Zegna A, Noble LJ, Panter SS, Sharp FR, Heme oxygenase-1 is induced in glia throughout brain by subarachnoid hemoglobin, Journal of Cerebral Blood Flow & Metabolism 18(3) (1998) 257–273. [DOI] [PubMed] [Google Scholar]
- [121].Koeppen AH, Dickson AC, McEvoy JA, The cellular reactions to experimental intracerebral hemorrhage, Journal of the neurological sciences 134 (1995) 102–112. [DOI] [PubMed] [Google Scholar]
- [122].Hua YA, Nakamura T, Keep RF, Wu J, Schallert T, Hoff JT, Xi G, Long-term effects of experimental intracerebral hemorrhage: the role of iron, Journal of neurosurgery 104(2) (2006) 305–312. [DOI] [PubMed] [Google Scholar]
- [123].Briat JF, Curie C, Gaymard F, Iron utilization and metabolism in plants, Curr Opin Plant Biol 10(3) (2007) 276–82. [DOI] [PubMed] [Google Scholar]
- [124].Beutler E, Gelbart T, West C, Lee P, Adams M, Blackstone R, Pockros P, Kosty M, Venditti CP, Phatak PD, Mutation analysis in hereditary hemochromatosis, Blood Cells, Molecules, and Diseases 22(2) (1996) 187–194. [DOI] [PubMed] [Google Scholar]
- [125].Cross AJ, Leitzmann MF, Gail MH, Hollenbeck AR, Schatzkin A, Sinha R, A prospective study of red and processed meat intake in relation to cancer risk, PLoS Med 4(12) (2007) e325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Huncharek M, Kupelnick B, A meta-analysis of maternal cured meat consumption during pregnancy and the risk of childhood brain tumors, Neuroepidemiology 23(1–2) (2004) 78–84. [DOI] [PubMed] [Google Scholar]
- [127].Bánfalvi G, Cellular effects of heavy metals, Springer; 2011. [Google Scholar]
- [128].Bowen HJM, Trace elements in biochemistry, Academic Press, London; New York, 1966. [Google Scholar]
- [129].McCall AS, Cummings CF, Bhave G, Vanacore R, Page-McCaw A, Hudson BG, Bromine is an essential trace element for assembly of collagen IV scaffolds in tissue development and architecture, Cell 157(6) (2014) 1380–1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Li L, Xu G, Shao H, Zhang Z-H, Pan X-F, Li J-Y, Analysis of Blood Concentrations of Zinc, Germanium, and Lead and Relevant Environmental Factors in a Population Sample from Shandong Province, China, International journal of environmental research and public health, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Salt DE, Blaylock M, Kumar NPBA, Dushenkov V, Ensley BD, Chet I, Raskin I, Phytoremediation: A Novel Strategy for the Removal of Toxic Metals from the Environment Using Plants, Bio/Technology 13(5) (1995) 468–474. [DOI] [PubMed] [Google Scholar]
- [132].Karri V, Schuhmacher M, Kumar V, Heavy metals (Pb, Cd, As and MeHg) as risk factors for cognitive dysfunction: A general review of metal mixture mechanism in brain, Environ Toxicol Pharmacol 48 (2016) 203–213. [DOI] [PubMed] [Google Scholar]
- [133].Zhu Y, Costa M, Metals and molecular carcinogenesis, Carcinogenesis 41(9) (2020) 1161–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].costa M, The Toxicity of Carcinogenic Metals to the Brain., J Carcinog Mutagen 11(354) (2020). [Google Scholar]
- [135].Mulware SJ, Comparative Trace Elemental Analysis in Cancerous and Noncancerous Human Tissues Using PIXE, Journal of Biophysics 2013 (2013) 192026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Chandra S, Parker DJ, Barth RF, Pannullo SC, Quantitative imaging of magnesium distribution at single-cell resolution in brain tumors and infiltrating tumor cells with secondary ion mass spectrometry (SIMS), J Neurooncol 127(1) (2016) 33–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Zhuang G, Zhou Y, Lu H, Lu W, Zhou M, Wang Y, Tan M, Concentration of rare earth elements, As, and Th in human brain and brain tumors, determined by neutron activation analysis, Biol Trace Elem Res 53(1–3) (1996) 45–9. [DOI] [PubMed] [Google Scholar]
- [138].Brun A, Brunk U, Histochemical indications for lysosomal localization of heavy metals in normal rat brain and liver, J Histochem Cytochem 18(11) (1970) 820–7. [DOI] [PubMed] [Google Scholar]
- [139].Squibb KS, Fowler BA, Relationship between metal toxicity to subcellular systems and the carcinogenic response, Environmental health perspectives 40 (1981) 181–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Genchi G, Sinicropi MS, Lauria G, Carocci A, Catalano A, The Effects of Cadmium Toxicity, International journal of environmental research and public health 17(11) (2020) 3782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Crane RK, Lipmann F, The effect of arsenate on aerobic phosphorylation, J Biol Chem 201(1) (1953) 235–43. [PubMed] [Google Scholar]
- [142].Sunderman FW Jr., Esfahani M, Nickel carbonyl inhibition of RNA polymerase activity in hepatic nuclei, Cancer Res 28(12) (1968) 2565–7. [PubMed] [Google Scholar]
- [143].Stoll RE, White JF, Miya TS, Bousquet WF, Effects of cadmium on nucleic acid and protein synthesis in rat liver, Toxicology and Applied Pharmacology 37(1) (1976) 61–74. [DOI] [PubMed] [Google Scholar]
- [144].Cilliers K, Muller CJF, Page BJ, Trace Element Concentration Changes in Brain Tumors: A Review, The Anatomical Record 303(5) (2020) 1293–1299. [DOI] [PubMed] [Google Scholar]
- [145].Andrási E, Suhajda M, Sáray I, Bezúr L, Ernyei L, Réffy A, Concentration of elements in human brain: glioblastoma multiforme, Sci Total Environ 139–140 (1993) 399–402. [DOI] [PubMed] [Google Scholar]
- [146].Hadfield MG, Adera T, Smith B, Fortner-Burton CA, Gibb RD, Mumaw V, Human brain tumors and exposure to metal and non-metal elements: a case-control study, J Environ Pathol Toxicol Oncol 17(1) (1998) 1–9. [PubMed] [Google Scholar]
- [147].Tapper UAS, Malmqvist KG, Brun A, Salford LG, Elemental regional distribution in human brain tumours — PIXE analysis of biopsy and autopsy samples, Nuclear Instruments and Methods in Physics Research Section B: Beam Interactions with Materials and Atoms 22(1) (1987) 176–178. [Google Scholar]
- [148].Rajadhyaksha MM, Turel ZR, Multielemental determination in normal, benign, and cancerous tissues of the human brain, Journal of Radioanalytical and Nuclear Chemistry 156(2) (1992) 341–347. [Google Scholar]
- [149].Wandzilak A, Czyzycki M, Radwanska E, Adamek D, Geraki K, Lankosz M, X-ray fluorescence study of the concentration of selected trace and minor elements in human brain tumours, Spectrochimica Acta Part B: Atomic Spectroscopy 114 (2015) 52–57. [Google Scholar]
- [150].Al-Saleh I, Shinwari N, Levels of cadmium, lead, and mercury in human brain tumors, Biological trace element research 79(3) (2001) 197–203. [DOI] [PubMed] [Google Scholar]
- [151].Dehnhardt M, Zoriy MV, Khan Z, Reifenberger G, Ekström TJ, Sabine Becker J, Zilles K, Bauer A, Element distribution is altered in a zone surrounding human glioblastoma multiforme, J Trace Elem Med Biol 22(1) (2008) 17–23. [DOI] [PubMed] [Google Scholar]
- [152].Szczerbowska-Boruchowska M, Lankosz M, Adamek D, First step toward the “fingerprinting” of brain tumors based on synchrotron radiation X-ray fluorescence and multiple discriminant analysis, J Biol Inorg Chem 16(8) (2011) 1217–26. [DOI] [PubMed] [Google Scholar]
- [153].Clark RE, IARC Monographs on the Evaluation of Carcinogenic Risks to Humans: Chromium, Nickel and Welding. Volume 49. Based on a Meeting Held in Lyon, 5–13 June 1989, The Quarterly Review of Biology 68(3) (1993) 472–473. [Google Scholar]
- [154].Tchounwou PB, Yedjou CG, Patlolla AK, Sutton DJ, Heavy metal toxicity and the environment, Exp Suppl 101 (2012) 133–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [155].Belyaeva EA, Dymkowska D, Wieckowski MR, Wojtczak L, Mitochondria as an important target in heavy metal toxicity in rat hepatoma AS-30D cells, Toxicol Appl Pharmacol 231(1) (2008) 34–42. [DOI] [PubMed] [Google Scholar]
- [156].Lee WK, Thévenod F, Cell organelles as targets of mammalian cadmium toxicity, Arch Toxicol 94(4) (2020) 1017–1049. [DOI] [PubMed] [Google Scholar]
- [157].Chatterjee S, Kundu S, Bhattacharyya A, Mechanism of cadmium induced apoptosis in the immunocyte, Toxicol Lett 177(2) (2008) 83–9. [DOI] [PubMed] [Google Scholar]
- [158].Chandravanshi LP, Gupta R, Shukla RK, Developmental Neurotoxicity of Arsenic: Involvement of Oxidative Stress and Mitochondrial Functions, Biol Trace Elem Res 186(1) (2018) 185–198. [DOI] [PubMed] [Google Scholar]
- [159].Chandravanshi LP, Shukla RK, Sultana S, Pant AB, Khanna VK, Early life arsenic exposure and brain dopaminergic alterations in rats, Int J Dev Neurosci 38 (2014) 91–104. [DOI] [PubMed] [Google Scholar]
- [160].Namgung U, Xia Z, Arsenic induces apoptosis in rat cerebellar neurons via activation of JNK3 and p38 MAP kinases, Toxicol Appl Pharmacol 174(2) (2001) 130–8. [DOI] [PubMed] [Google Scholar]
- [161].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]
- [162].Mochizuki H, Arsenic Neurotoxicity in Humans, International journal of molecular sciences 20(14) (2019) 3418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Solomon E, Borrow J, Goddard AD, Chromosome aberrations and cancer, Science 254(5035) (1991) 1153–60. [DOI] [PubMed] [Google Scholar]
- [164].Bos JL, ras oncogenes in human cancer: a review, Cancer Res 49(17) (1989) 4682–9. [PubMed] [Google Scholar]
- [165].Hollstein M, Sidransky D, Vogelstein B, Harris CC, p53 mutations in human cancers, Science 253(5015) (1991) 49–53. [DOI] [PubMed] [Google Scholar]
- [166].Schwab M, Oncogene amplification in neoplastic development and progression of human cancers, Crit Rev Oncog 2(1) (1990) 35–51. [PubMed] [Google Scholar]
- [167].Valko M, Morris H, Cronin MT, Metals, toxicity and oxidative stress, Curr Med Chem 12(10) (2005) 1161–208. [DOI] [PubMed] [Google Scholar]
- [168].Waisberg M, Joseph P, Hale B, Beyersmann D, Molecular and cellular mechanisms of cadmium carcinogenesis, Toxicology 192(2–3) (2003) 95–117. [DOI] [PubMed] [Google Scholar]
- [169].Torreilles J, Guerin MC, Slaoui-Hasnaoui A, Nickel (II) complexes of histidyl-peptides as Fenton-reaction catalysts, Free Radic Res Commun 11(1–3) (1990) 159–66. [DOI] [PubMed] [Google Scholar]
- [170].Hug SJ, Leupin O, Iron-catalyzed oxidation of arsenic(III) by oxygen and by hydrogen peroxide: pH-dependent formation of oxidants in the Fenton reaction, Environ Sci Technol 37(12) (2003) 2734–42. [DOI] [PubMed] [Google Scholar]
- [171].Robb SJ, Robb-Gaspers LD, Scaduto RC Jr, Thomas AP, Connor JR, Influence of calcium and iron on cell death and mitochondrial function in oxidatively stressed astrocytes, Journal of neuroscience research 55(6) (1999) 674–686. [DOI] [PubMed] [Google Scholar]
- [172].Gul N, Shah MT, Khan S, Khattak NU, Muhammad S, Arsenic and heavy metals contamination, risk assessment and their source in drinking water of the Mardan District, Khyber Pakhtunkhwa, Pakistan, J Water Health 13(4) (2015) 1073–84. [DOI] [PubMed] [Google Scholar]
- [173].Grimsrud TK, Berge SR, Martinsen JI, Andersen A, Lung cancer incidence among Norwegian nickel-refinery workers 1953–2000, J Environ Monit 5(2) (2003) 190–7. [DOI] [PubMed] [Google Scholar]
- [174].Wesseling C, Pukkala E, Neuvonen K, Kauppinen T, Boffetta P, Partanen T, Cancer of the brain and nervous system and occupational exposures in Finnish women, J Occup Environ Med 44(7) (2002) 663–8. [DOI] [PubMed] [Google Scholar]
- [175].Siesjö BK, Agardh CD, Bengtsson F, Free radicals and brain damage, Cerebrovasc Brain Metab Rev 1(3) (1989) 165–211. [PubMed] [Google Scholar]
- [176].Baader SL, Bruchelt G, Carmine TC, Lode HN, Rieth AG, Niethammer D, Ascorbic-acid-mediated iron release from cellular ferritin and its relation to the formation of DNA strand breaks in neuroblastoma cells, J Cancer Res Clin Oncol 120(7) (1994) 415–21. [DOI] [PubMed] [Google Scholar]
- [177].Breuer W, Greenberg E, Cabantchik ZI, Newly delivered transferrin iron and oxidative cell injury, FEBS Lett 403(2) (1997) 213–9. [DOI] [PubMed] [Google Scholar]
- [178].Rauhala P, Chiueh CC, Effects of atypical antioxidative agents, S-nitrosoglutathione and manganese, on brain lipid peroxidation induced by iron leaking from tissue disruption, Ann N Y Acad Sci 899 (2000) 238–54. [DOI] [PubMed] [Google Scholar]
- [179].Ward RJ, Kühn LC, Kaldy P, Florence A, Peters TJ, Crichton RR, Control of cellular iron homeostasis by iron-responsive elements in vivo, Eur J Biochem 220(3) (1994) 927–31. [DOI] [PubMed] [Google Scholar]
- [180].Halliwell B, Gutteridge JMC, Free radicals in biology and medicine, Oxford University Press, USA: 2015. [Google Scholar]
- [181].Jung M, Mertens C, Tomat E, Brüne B, Iron as a Central Player and Promising Target in Cancer Progression, Int J Mol Sci 20(2) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Zhou L, Zhao B, Zhang L, Wang S, Dong D, Lv H, Shang P, Alterations in Cellular Iron Metabolism Provide More Therapeutic Opportunities for Cancer, Int J Mol Sci 19(5) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [183].Basuli D, Tesfay L, Deng Z, Paul B, Yamamoto Y, Ning G, Xian W, McKeon F, Lynch M, Crum CP, Hegde P, Brewer M, Wang X, Miller LD, Dyment N, Torti FM, Torti SV, Iron addiction: a novel therapeutic target in ovarian cancer, Oncogene 36(29) (2017) 4089–4099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Forciniti S, Greco L, Grizzi F, Malesci A, Laghi L, Iron Metabolism in Cancer Progression, Int J Mol Sci 21(6) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [185].Ludwig H, Evstatiev R, Kornek G, Aapro M, Bauernhofer T, Buxhofer-Ausch V, Fridrik M, Geissler D, Geissler K, Gisslinger H, Koller E, Kopetzky G, Lang A, Rumpold H, Steurer M, Kamali H, Link H, Iron metabolism and iron supplementation in cancer patients, Wien Klin Wochenschr 127(23–24) (2015) 907–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Shen Y, Li X, Dong D, Zhang B, Xue Y, Shang P, Transferrin receptor 1 in cancer: a new sight for cancer therapy, Am J Cancer Res 8(6) (2018) 916–931. [PMC free article] [PubMed] [Google Scholar]
- [187].Kim K, Mitra S, Wu G, Berka V, Song J, Yu Y, Poget S, Wang DN, Tsai AL, Zhou M, Six-Transmembrane Epithelial Antigen of Prostate 1 (STEAP1) Has a Single b Heme and Is Capable of Reducing Metal Ion Complexes and Oxygen, Biochemistry 55(48) (2016) 6673–6684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Han M, Xu R, Wang S, Yang N, Ni S, Zhang Q, Xu Y, Zhang X, Zhang C, Wei Y, Ji J, Huang B, Zhang D, Chen A, Li W, Bjerkvig R, Li X, Wang J, Six-Transmembrane Epithelial Antigen of Prostate 3 Predicts Poor Prognosis and Promotes Glioblastoma Growth and Invasion, Neoplasia 20(6) (2018) 543–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Marnett LJ, Oxy radicals, lipid peroxidation and DNA damage, Toxicology 181–182 (2002) 219–22. [DOI] [PubMed] [Google Scholar]
- [190].Bedard K, Krause KH, The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology, Physiol Rev 87(1) (2007) 245–313. [DOI] [PubMed] [Google Scholar]
- [191].Veith A, Moorthy B, ROLE OF CYTOCHROME P450S IN THE GENERATION AND METABOLISM OF REACTIVE OXYGEN SPECIES, Curr Opin Toxicol 7 (2018) 44–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [192].Rhee SG, Yang KS, Kang SW, Woo HA, Chang TS, Controlled elimination of intracellular H(2)O(2): regulation of peroxiredoxin, catalase, and glutathione peroxidase via post-translational modification, Antioxid Redox Signal 7(5–6) (2005) 619–26. [DOI] [PubMed] [Google Scholar]
- [193].Kress GJ, Dineley KE, Reynolds IJ, The relationship between intracellular free iron and cell injury in cultured neurons, astrocytes, and oligodendrocytes, Journal of Neuroscience 22(14) (2002) 5848–5855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [194].Snyder A, The Role of Glia in Neurotoxicity, (1996).
- [195].Landis DMD, The early reactions of non-neuronal cells to brain injury, Annual review of neuroscience 17(1) (1994) 133–151. [DOI] [PubMed] [Google Scholar]
- [196].Makar TK, Nedergaard M, Preuss A, Gelbard AS, Perumal AS, Cooper AJL, Vitamin E, ascorbate, glutathione, glutathicne disulfide, and enzymes of glutathione metabolism in cultures of chick astrocytes and neurons: evidence that astrocytes play an important role in antioxidative processes in the brain, Journal of neurochemistry 62(1) (1994) 45–53. [DOI] [PubMed] [Google Scholar]
- [197].Aschner M, The functional significance of brain metallothioneins, The FASEB journal 10(10) (1996) 1129–1136. [DOI] [PubMed] [Google Scholar]
- [198].Raps SP, Lai JCK, Hertz L, Cooper AJL, Glutathione is present in high concentrations in cultured astrocytes but not in cultured neurons, Brain research 493(2) (1989) 398–401. [DOI] [PubMed] [Google Scholar]
- [199].Oide T, Yoshida K, Kaneko K, Ohta M, Arima K, Iron overload and antioxidative role of perivascular astrocytes in aceruloplasminemia, Neuropathology and applied neurobiology 32(2) (2006) 170–176. [DOI] [PubMed] [Google Scholar]
- [200].Halcrow P, Datta G, Ohm JE, Soliman ML, Chen X, Geiger JD, Role of endolysosomes and pH in the pathogenesis and treatment of glioblastoma, Cancer Reports 2(6) (2019) e1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [201].Halcrow PW, Khan N, Datta G, Ohm JE, Chen X, Geiger JD, Importance of measuring endolysosome, cytosolic, and extracellular pH in understanding the pathogenesis of and possible treatments for glioblastoma multiforme, Cancer Reports 2(6) (2019) e1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [202].Yu Z, Persson HL, Eaton JW, Brunk UT, Intralysosomal iron: a major determinant of oxidant-induced cell death, Free Radical Biology and Medicine 34(10) (2003) 1243–1252. [DOI] [PubMed] [Google Scholar]
- [203].Persson HL, Yu Z, Tirosh O, Eaton JW, Brunk UT, Prevention of oxidant-induced cell death by lysosomotropic iron chelators, Free Radical Biology and Medicine 34(10) (2003) 1295–1305. [DOI] [PubMed] [Google Scholar]
- [204].Petrat F, de Groot H, Rauen U, Subcellular distribution of chelatable iron: a laser scanning microscopic study in isolated hepatocytes and liver endothelial cells, Biochem J 356(Pt 1) (2001) 61–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [205].Richmond HG, Induction of sarcoma in the rat by iron-dextran complex, Br Med J 1(5127) (1959) 947–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [206].Uchiyama A, Kim JS, Kon K, Jaeschke H, Ikejima K, Watanabe S, Lemasters JJ, Translocation of iron from lysosomes into mitochondria is a key event during oxidative stress-induced hepatocellular injury, Hepatology 48(5) (2008) 1644–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [207].Kurz T, Eaton JW, Brunk UT, The role of lysosomes in iron metabolism and recycling, Int J Biochem Cell Biol 43(12) (2011) 1686–97. [DOI] [PubMed] [Google Scholar]
- [208].Cable H, Lloyd JB, Cellular uptake and release of two contrasting iron chelators, J Pharm Pharmacol 51(2) (1999) 131–4. [DOI] [PubMed] [Google Scholar]
- [209].Lloyd JB, Cable H, Rice-Evans C, Evidence that desferrioxamine cannot enter cells by passive diffusion, Biochemical Pharmacology 41(9) (1991) 1361–1363. [DOI] [PubMed] [Google Scholar]
- [210].Doulias PT, Christoforidis S, Brunk UT, Galaris D, Endosomal and lysosomal effects of desferrioxamine: protection of HeLa cells from hydrogen peroxide-induced DNA damage and induction of cell-cycle arrest, Free Radic Biol Med 35(7) (2003) 719–28. [DOI] [PubMed] [Google Scholar]
- [211].Kurz T, Gustafsson B, Brunk UT, Intralysosomal iron chelation protects against oxidative stress-induced cellular damage, The FEBS Journal 273(13) (2006) 3106–3117. [DOI] [PubMed] [Google Scholar]
- [212].Glickstein H, Ben El R, Shvartsman M, Cabantchik ZI, Intracellular labile iron pools as direct targets of iron chelators: a fluorescence study of chelator action in living cells, Blood 106(9) (2005) 3242–3250. [DOI] [PubMed] [Google Scholar]
- [213].Alhajala HS, Joshi D, Al-Gizawiy MM, Hedlund B, Elford HL, Schmainda KM, Chitambar CR, EXTH-40. TUMOR IRON STATUS SENSITIZES GLIOBLASTOMA CELLS TO THE CYTOTOXICITY OF AN INHIBITOR OF THE IRON-DEPENDENT M2 SUBUNIT OF RIBONUCLEOTIDE REDUCTASE, Neuro-Oncology 19(suppl_6) (2017) vi81–vi81. [Google Scholar]
- [214].Hanson LR, Roeytenberg A, Martinez PM, Coppes VG, Sweet DC, Rao RJ, Marti DL, Hoekman JD, Matthews RB, Frey WH 2nd, Panter SS, Intranasal deferoxamine provides increased brain exposure and significant protection in rat ischemic stroke, The Journal of pharmacology and experimental therapeutics 330(3) (2009) 679–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [215].Smith SW, The role of chelation in the treatment of other metal poisonings, Journal of medical toxicology : official journal of the American College of Medical Toxicology 9(4) (2013) 355–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [216].Molina-Jijón E, Zarco-Márquez G, Medina-Campos ON, Zataraín-Barrón ZL, Hernández-Pando R, Pinzón E, Zavaleta RM, Tapia E, Pedraza-Chaverri J, Deferoxamine pretreatment prevents Cr(VI)-induced nephrotoxicity and oxidant stress: role of Cr(VI) chelation, Toxicology 291(1–3) (2012) 93–101. [DOI] [PubMed] [Google Scholar]
- [217].Warnick RE, Yook E, Relationship between pin type and depth of skull penetration during frame placement for Gamma Knife radiosurgery, Journal of radiosurgery and SBRT 6(3) (2019) 241–246. [PMC free article] [PubMed] [Google Scholar]
- [218].Chen H-E, Lin J-F, Tsai T-F, Lin Y-C, Chou K-Y, Hwang T, Chloroquine induces lysosomal membrane permeability mediated apoptotic cell death in bladder cancer cells, Urological Science 27 (2016) S5. [Google Scholar]
- [219].Halcrow PW, Geiger JD, Chen X, Overcoming Chemoresistance: Altering pH of Cellular Compartments by Chloroquine and Hydroxychloroquine, Frontiers in Cell and Developmental Biology 9 (2021) 170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [220].Persson HL, Kurz T, Eaton JW, Brunk UT, Radiation-induced cell death: importance of lysosomal destabilization, The Biochemical journal 389(Pt 3) (2005) 877–884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [221].C. International Agency for Research on, IARC monographs on the evaluation of the carcinogenic risk of chemicals to humans. Vol. 22. Some non-nutritive sweetening agents, Distributed for IARC by WHO, Geneva, Switzerland: 1980. [PubMed] [Google Scholar]
- [222].Lu S, Sung T, Lin N, Abraham RT, Jessen BA, Lysosomal adaptation: How cells respond to lysosomotropic compounds, PLOS ONE 12(3) (2017) e0173771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [223].Mader BJ, Pivtoraiko VN, Flippo HM, Klocke BJ, Roth KA, Mangieri LR, Shacka JJ, Rotenone inhibits autophagic flux prior to inducing cell death, ACS chemical neuroscience 3(12) (2012) 1063–1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [224].Styrt B, Klempner MS, Lysosomotropic amines modulate neutrophil calcium homeostasis, J Cell Physiol 135(2) (1988) 309–16. [DOI] [PubMed] [Google Scholar]
- [225].Alfonso-Loeches S, Pascual-Lucas M, Blanco AM, Sanchez-Vera I, Guerri C, Pivotal role of TLR4 receptors in alcohol-induced neuroinflammation and brain damage, J Neurosci 30(24) (2010) 8285–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [226].Pla A, Pascual M, Guerri C, Autophagy Constitutes a Protective Mechanism against Ethanol Toxicity in Mouse Astrocytes and Neurons, PLOS ONE 11(4) (2016) e0153097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [227].Zahedi A, Phandthong R, Chaili A, Leung S, Omaiye E, Talbot P, Mitochondrial Stress Response in Neural Stem Cells Exposed to Electronic Cigarettes, iScience 16 (2019) 250–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [228].Halcrow PW, Lakpa KL, Khan N, Afghah Z, Miller N, Datta G, Chen X, Geiger JD, HIV-1 gp120-Induced Endolysosome de-Acidification Leads to Efflux of Endolysosome Iron, and Increases in Mitochondrial Iron and Reactive Oxygen Species, J Neuroimmune Pharmacol (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [229].Thyberg J, Hedin U, Stenseth K, Nilsson J, Effects of nicotine on the fine structure of cultivated mouse peritoneal macrophages, Acta Pathologica Microbiologica Scandinavica Series A: Pathology 91(1–6) (1983) 23–30. [DOI] [PubMed] [Google Scholar]
- [230].Govind AP, Vezina P, Green WN, Nicotine-induced upregulation of nicotinic receptors: underlying mechanisms and relevance to nicotine addiction, Biochemical pharmacology 78(7) (2009) 756–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [231].Suen DF, Norris KL, Youle RJ, Mitochondrial dynamics and apoptosis, Genes Dev 22(12) (2008) 1577–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [232].Cassidy-Stone A, Chipuk JE, Ingerman E, Song C, Yoo C, Kuwana T, Kurth MJ, Shaw JT, Hinshaw JE, Green DR, Nunnari J, Chemical inhibition of the mitochondrial division dynamin reveals its role in Bax/Bak-dependent mitochondrial outer membrane permeabilization, Dev Cell 14(2) (2008) 193–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [233].Pletjushkina OY, Lyamzaev KG, Popova EN, Nepryakhina OK, Ivanova OY, Domnina LV, Chernyak BV, Skulachev VP, Effect of oxidative stress on dynamics of mitochondrial reticulum, Biochim Biophys Acta 1757(5–6) (2006) 518–24. [DOI] [PubMed] [Google Scholar]
- [234].Hahn A, Zuryn S, The Cellular Mitochondrial Genome Landscape in Disease, Trends Cell Biol 29(3) (2019) 227–240. [DOI] [PubMed] [Google Scholar]
- [235].Zheng Q, Zhao Y, Guo J, Zhao S, Fei C, Xiao C, Wu D, Wu L, Li X, Chang C, Iron overload promotes mitochondrial fragmentation in mesenchymal stromal cells from myelodysplastic syndrome patients through activation of the AMPK/MFF/Drp1 pathway, Cell Death & Disease 9(5) (2018) 515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [236].Park J, Lee DG, Kim B, Park SJ, Kim JH, Lee SR, Chang KT, Lee HS, Lee DS, Iron overload triggers mitochondrial fragmentation via calcineurin-sensitive signals in HT-22 hippocampal neuron cells, Toxicology 337 (2015) 39–46. [DOI] [PubMed] [Google Scholar]
- [237].Li Z, Zhu Y.-t., Xiang M, Qiu J.-l., Luo S.-q., Lin F, Enhanced lysosomal function is critical for paclitaxel resistance in cancer cells: reversed by artesunate, Acta Pharmacologica Sinica 42(4) (2021) 624–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [238].Hamacher-Brady A, Stein HA, Turschner S, Toegel I, Mora R, Jennewein N, Efferth T, Eils R, Brady NR, Artesunate activates mitochondrial apoptosis in breast cancer cells via iron-catalyzed lysosomal reactive oxygen species production, J Biol Chem 286(8) (2011) 6587–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [239].Gasmi A, Peana M, Noor S, Lysiuk R, Menzel A, Gasmi Benahmed A, Bjørklund G, Chloroquine and hydroxychloroquine in the treatment of COVID-19: the never-ending story, Applied Microbiology and Biotechnology 105(4) (2021) 1333–1343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [240].Vorobjev IA, Zorov DB, Diazepam inhibits cell respiration and induces fragmentation of mitochondrial reticulum, FEBS Lett 163(2) (1983) 311–4. [DOI] [PubMed] [Google Scholar]
- [241].Poliakova IA, Zorov DB, Leĭkina MI, [Structure-activity changes of mitochondria from cultured cells upon disruption of energy metabolism], Dokl Akad Nauk 342(4) (1995) 553–5. [PubMed] [Google Scholar]
- [242].Zorov DB, Vorobjev IA, Popkov VA, Babenko VA, Zorova LD, Pevzner IB, Silachev DN, Zorov SD, Andrianova NV, Plotnikov EY, Lessons from the Discovery of Mitochondrial Fragmentation (Fission): A Review and Update, Cells 8(2) (2019) 175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [243].Cho B, Choi SY, Cho HM, Kim HJ, Sun W, Physiological and pathological significance of dynamin-related protein 1 (drp1)-dependent mitochondrial fission in the nervous system, Exp Neurobiol 22(3) (2013) 149–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [244].Tanwar DK, Parker DJ, Gupta P, Spurlock B, Alvarez RD, Basu MK, Mitra K, Crosstalk between the mitochondrial fission protein, Drp1, and the cell cycle is identified across various cancer types and can impact survival of epithelial ovarian cancer patients, Oncotarget 7(37) (2016) 60021–60037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [245].Xie Q, Wu Q, Horbinski CM, Flavahan WA, Yang K, Zhou W, Dombrowski SM, Huang Z, Fang X, Shi Y, Ferguson AN, Kashatus DF, Bao S, Rich JN, Mitochondrial control by DRP1 in brain tumor initiating cells, Nature neuroscience 18(4) (2015) 501–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [246].Guido C, Whitaker-Menezes D, Lin Z, Pestell RG, Howell A, Zimmers TA, Casimiro MC, Aquila S, Ando S, Martinez-Outschoorn UE, Sotgia F, Lisanti MP, Mitochondrial fission induces glycolytic reprogramming in cancer-associated myofibroblasts, driving stromal lactate production, and early tumor growth, Oncotarget 3(8) (2012) 798–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [247].Wan Y-Y, Zhang J-F, Yang Z-J, Jiang L-P, Wei YF, Lai Q-N, Wang J-B, Xin H-B, Han X-J, Involvement of Drp1 in hypoxia-induced migration of human glioblastoma U251 cells, Oncol Rep 32(2) (2014) 619–626. [DOI] [PubMed] [Google Scholar]
- [248].Wan YY, Zhang JF, Yang ZJ, Jiang LP, Wei YF, Lai QN, Wang JB, Xin HB, Han XJ, Involvement of Drp1 in hypoxia-induced migration of human glioblastoma U251 cells, Oncol Rep 32(2) (2014) 619–26. [DOI] [PubMed] [Google Scholar]
- [249].Liu W, Ye C, Cheng Q, Zhang X, Yao L, Li Q, Huang J, Liu Y, Zou Z, Wang H, Yan J, Zhu Y, Wang C, Ai D, Macrophage Raptor Deficiency-Induced Lysosome Dysfunction Exacerbates Nonalcoholic Steatohepatitis, Cellular and molecular gastroenterology and hepatology 7(1) (2019) 211–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [250].Wallace DC, Mitochondria and cancer, Nature reviews. Cancer 12(10) (2012) 685–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [251].Horton TM, Petros JA, Heddi A, Shoffner J, Kaufman AE, Graham SD Jr., Gramlich T, Wallace DC, Novel mitochondrial DNA deletion found in a renal cell carcinoma, Genes Chromosomes Cancer 15(2) (1996) 95–101. [DOI] [PubMed] [Google Scholar]
- [252].Pereira L, Soares P, Máximo V, Samuels DC, Somatic mitochondrial DNA mutations in cancer escape purifying selection and high pathogenicity mutations lead to the oncocytic phenotype: pathogenicity analysis of reported somatic mtDNA mutations in tumors, BMC Cancer 12 (2012) 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [253].Chinnery PF, Samuels DC, Elson J, Turnbull DM, Accumulation of mitochondrial DNA mutations in ageing, cancer, and mitochondrial disease: is there a common mechanism?, Lancet 360(9342) (2002) 1323–5. [DOI] [PubMed] [Google Scholar]
- [254].Brandon M, Baldi P, Wallace DC, Mitochondrial mutations in cancer, Oncogene 25(34) (2006) 4647–62. [DOI] [PubMed] [Google Scholar]
- [255].Meierhofer D, Mayr JA, Fink K, Schmeller N, Kofler B, Sperl W, Mitochondrial DNA mutations in renal cell carcinomas revealed no general impact on energy metabolism, Br J Cancer 94(2) (2006) 268–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [256].Howell AN, Sager R, Tumorigenicity and its suppression in cybrids of mouse and Chinese hamster cell lines, Proc Natl Acad Sci U S A 75(5) (1978) 2358–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [257].Shidara Y, Yamagata K, Kanamori T, Nakano K, Kwong JQ, Manfredi G, Oda H, Ohta S, Positive contribution of pathogenic mutations in the mitochondrial genome to the promotion of cancer by prevention from apoptosis, Cancer Res 65(5) (2005) 1655–63. [DOI] [PubMed] [Google Scholar]
- [258].Ishikawa K, Takenaga K, Akimoto M, Koshikawa N, Yamaguchi A, Imanishi H, Nakada K, Honma Y, Hayashi J, ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis, Science 320(5876) (2008) 661–4. [DOI] [PubMed] [Google Scholar]
- [259].Cadet J, Delatour T, Douki T, Gasparutto D, Pouget J-P, Ravanat J-L, Sauvaigo S, Hydroxyl radicals and DNA base damage, Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis 424(1) (1999) 9–21. [DOI] [PubMed] [Google Scholar]
- [260].Chatgilialoglu C, O’Neill P, Free radicals associated with DNA damage, Exp Gerontol 36(9) (2001) 1459–71. [DOI] [PubMed] [Google Scholar]
- [261].Halliwell B, Gutteridge JM, Role of free radicals and catalytic metal ions in human disease: an overview, Methods Enzymol 186 (1990) 1–85. [DOI] [PubMed] [Google Scholar]
- [262].Halliwell B, Aruoma OI, DNA damage by oxygen-derived species. Its mechanism and measurement in mammalian systems, FEBS Lett 281(1–2) (1991) 9–19. [DOI] [PubMed] [Google Scholar]
- [263].Ames BN, Shigenaga MK, Hagen TM, Oxidants, antioxidants, and the degenerative diseases of aging, Proceedings of the National Academy of Sciences of the United States of America 90(17) (1993) 7915–7922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [264].Meneghini R, Iron homeostasis, oxidative stress, and DNA damage, Free Radic Biol Med 23(5) (1997) 783–92. [DOI] [PubMed] [Google Scholar]
- [265].Galey JB, Potential use of iron chelators against oxidative damage, Adv Pharmacol 38 (1997) 167–203. [DOI] [PubMed] [Google Scholar]
- [266].Chevion M, A site-specific mechanism for free radical induced biological damage: the essential role of redox-active transition metals, Free Radic Biol Med 5(1) (1988) 27–37. [DOI] [PubMed] [Google Scholar]
- [267].Gurgueira SA, Meneghini R, An ATP-dependent iron transport system in isolated rat liver nuclei, J Biol Chem 271(23) (1996) 13616–20. [DOI] [PubMed] [Google Scholar]
- [268].Kurz T, Leake A, Von Zglinicki T, Brunk UT, Relocalized redox-active lysosomal iron is an important mediator of oxidative-stress-induced DNA damage, Biochemical Journal 378(3) (2004) 1039–1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [269].Poetsch AR, The genomics of oxidative DNA damage, repair, and resulting mutagenesis, Computational and Structural Biotechnology Journal 18 (2020) 207–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [270].Stevens RG, Beasley RP, Blumberg BS, Iron-binding proteins and risk of cancer in Taiwan, J Natl Cancer Inst 76(4) (1986) 605–10. [DOI] [PubMed] [Google Scholar]
- [271].Selby JV, Friedman GD, Epidemiologic evidence of an association between body iron stores and risk of cancer, Int J Cancer 41(5) (1988) 677–82. [DOI] [PubMed] [Google Scholar]
- [272].Park SH, Aust AE, Participation of iron and nitric oxide in the mutagenicity of asbestos in hgprt-, gpt+ Chinese hamster V79 cells, Cancer Res 58(6) (1998) 1144–8. [PubMed] [Google Scholar]
- [273].Chao CC, Park SH, Aust AE, Participation of nitric oxide and iron in the oxidation of DNA in asbestos-treated human lung epithelial cells, Arch Biochem Biophys 326(1) (1996) 152–7. [DOI] [PubMed] [Google Scholar]
- [274].Aust AE, Eveleigh JF, Mechanisms of DNA oxidation, Proc Soc Exp Biol Med 222(3) (1999) 246–52. [DOI] [PubMed] [Google Scholar]
- [275].Lee JH, Lee JE, Kahng JY, Kim SH, Park JS, Yoon SJ, Um J-Y, Kim WK, Lee J-K, Park J, Kim EH, Lee J-H, Lee J-H, Chung W-S, Ju YS, Park S-H, Chang JH, Kang S-G, Lee JH, Human glioblastoma arises from subventricular zone cells with low-level driver mutations, Nature 560(7717) (2018) 243–247. [DOI] [PubMed] [Google Scholar]
- [276].Gupta SC, Hevia D, Patchva S, Park B, Koh W, Aggarwal BB, Upsides and downsides of reactive oxygen species for cancer: the roles of reactive oxygen species in tumorigenesis, prevention, and therapy, Antioxidants & redox signaling 16(11) (2012) 1295–1322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [277].Benhar M, Engelberg D, Levitzki A, ROS, stress-activated kinases and stress signaling in cancer, EMBO Rep 3(5) (2002) 420–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [278].D’Autréaux B, Toledano MB, ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis, Nat Rev Mol Cell Biol 8(10) (2007) 813–24. [DOI] [PubMed] [Google Scholar]
- [279].Jackson AL, Loeb LA, The contribution of endogenous sources of DNA damage to the multiple mutations in cancer, Mutat Res 477(1–2) (2001) 7–21. [DOI] [PubMed] [Google Scholar]
- [280].Marnett LJ, Oxyradicals and DNA damage, carcinogenesis 21(3) (2000) 361–370. [DOI] [PubMed] [Google Scholar]
- [281].Cooke MS, Evans MD, Dizdaroglu M, Lunec J, Oxidative DNA damage: mechanisms, mutation, and disease, The FASEB Journal 17(10) (2003) 1195–1214. [DOI] [PubMed] [Google Scholar]
- [282].Zhao R, Planalp RP, Ma R, Greene BT, Jones BT, Brechbiel MW, Torti FM, Torti SV, Role of zinc and iron chelation in apoptosis mediated by tachpyridine, an anti-cancer iron chelator, Biochem Pharmacol 67(9) (2004) 1677–88. [DOI] [PubMed] [Google Scholar]
- [283].Richardson DR, Iron chelators as therapeutic agents for the treatment of cancer, Crit Rev Oncol Hematol 42(3) (2002) 267–81. [DOI] [PubMed] [Google Scholar]
- [284].Donfrancesco A, De Bernardi B, Carli M, Mancini A, Nigro M, De Sio L, Casale F, Bagnulo S, Helson L, Deb G, Deferoxamine followed by cyclophosphamide, etoposide, carboplatin, thiotepa, induction regimen in advanced neuroblastoma: Preliminary results, European Journal of Cancer 31(4) (1995) 612–615. [DOI] [PubMed] [Google Scholar]
- [285].Valdés PA, Samkoe K, O’Hara JA, Roberts DW, Paulsen KD, Pogue BW, Deferoxamine iron chelation increases delta-aminolevulinic acid induced protoporphyrin IX in xenograft glioma model, Photochem Photobiol 86(2) (2010) 471–5. [DOI] [PMC free article] [PubMed] [Google Scholar]