Abstract
Iron is a key element for mitochondrial function and homeostasis, which is also crucial for maintaining the neuronal system, but too much iron promotes oxidative stress. A large body of evidence has indicated that abnormal iron accumulation in the brain is associated with various neurogenerative diseases such as Huntington’s disease, Alzheimer’s disease, Parkinson’s disease, and Friedreich’s ataxia. However, it is still unclear how irregular iron status contributes to the development of neuronal disorders. Hence, the current review provides an update on the causal effects of iron overload in the development and progression of neurodegenerative diseases and discusses important roles of mitochondrial iron homeostasis in these disease conditions. Furthermore, this review discusses potential therapeutic targets for the treatments of iron overload-linked neurodegenerative diseases.
Keywords: Brain iron, Iron overload, Iron transport, Mitochondria, Neuron
1. INTRODUCTION
Iron, an essential metal nutrient, is a fundamental element for many metabolic processes, including electron transfer, oxygen transport and storage, mitochondrial function, and cellular growth and differentiation. Such biochemical reactions in the body are processed by iron-containing proteins such as hemoglobin, iron-sulfur enzymes, and heme-containing enzymes. Thus, iron homeostasis is controlled via multiple iron regulatory processes: iron uptake, storage, transfer, and export to other cells or organs.
In the brain, iron is involved in many physiological activities such as mitochondrial respiration and energy production, myelin synthesis, neurotransmitter synthesis and metabolism, and DNA replication (1). Given that neuronal mitochondrial respiration is in charge of about 20% of the total body oxygen consumption (2), the maintenance of physiological iron levels is critical for proper brain function. Indeed, dysregulated iron levels have been reported to be associated with several neurodegenerative diseases, including Friedreich’s ataxia (FA), Huntington’s disease (HD), Alzheimer’s disease (AD), Parkinson’s disease (PD), and neurodegeneration with brain iron accumulation (NBIA) (3–7). Studies have also pointed out that many neurodegenerative disorders are characterized by abnormal iron status in the mitochondria, the major iron recipient of the cell (8–10). In this review, we aim to summarize important roles of mitochondrial iron in neuronal function and to provide current understanding about the pathophysiology related to mitochondrial dysfunction in neurodegenerative diseases.
2. MITOCHONDRIA AND IRON
2.1. Biochemistry of Iron
Iron is a d-block transition metal whose primary oxidation states in biological conditions are ferrous (II), ferric (III), and ferryl (IV) states. The unoccupied d orbitals also allow iron to bind to ligands, including oxygen, nitrogen, and sulfur atoms (11, 12). Thus, iron has a tendency to easily donate its electrons, which contributes to various biochemical processes (13). In normal cellular conditions, ferric iron (Fe3+) constitutes the common stable form of bioavailable iron that is associated with various biological complexes. Meanwhile, ferrous iron (Fe2+) can be found in ferritin, heme synthesis, and transmembrane transport. Despite its importance in biochemical processes, free ferrous iron is highly reactive and can cause cellular damage (13, 14), and is therefore stored and chaperoned throughout the cell with the help of several iron binding and transport proteins (e.g., ferritin or hemosiderin) (13). For example, ferritin is a highly conserved protein among both vertebrate and invertebrate species, and two subunits of ferritin, L-ferritin (FTL) and H-ferritin (FTH), are essential for iron storage in vertebrate cells.
2.2. Cellular Iron Homeostasis
Iron, after its intestinal absorption or export out of iron-storage organs (e.g., liver), binds to transferrin (Tf) as a ferric form in the plasma, and Tf-bound iron (TBI) is taken up by the cell through endocytosis mediated by transferrin receptor 1 (TfR1), which is the major iron transport mechanism under normal physiological conditions (15–17). Intracellular Fe3+ is then reduced by ferrireductase Steap3 to Fe2+ in the endosome and is transported to the cytoplasm by the divalent metal transporter 1 (DMT1). This reduced iron (Fe2+) is either stored in ferritin protein, labile iron pool or delivered to mitochondria (18).
In the cytosol, iron concentration is regulated by iron regulatory proteins (IRP1 and IRP2) that bind to the iron-responsive elements (IREs) within the 5’ or 3’ untranslated regions (UTR) of target mRNAs and regulate the expression of various iron processing proteins, including TfR1 and iron storage protein (i.e., ferritin). Thus, the levels of these iron-associated proteins are precisely regulated by IRPs, and any decrease in extracellular iron levels can promote the IRP-IRE interaction, resulting in increased stability of TfR1 mRNA and decreased translation of ferritin (19). Conversely, when the cell is iron sufficient, a 4Fe-4S cluster is bound to IRP1, while IRP2 undergoes iron-mediated proteasomal degradation. The depletion of IRP2 not only causes the degradation of TfR1 mRNA, but also promotes ferritin translation. Those outcomes lead to a decrease in iron transport into the cell, as well as an increase in iron storage by ferritin, thereby preventing iron-induced oxidative damage. The IRP2-mediated iron response is thought to be dependent on oxygen level as well as FBXL5 protein binding, which further activates ubiquitination-dependent IRP2 degradation (20). However, a recent study suggests that IRP2 may not require FBXL5 protein binding to modulate its iron response and it can regulate cellular iron levels independent of IRP1 (21). The underlying mechanism has been proposed to be dependent on iron-sulfur (Fe-S) cluster binding to the IRP2 protein itself, since an inhibition of Fe-S biogenesis in IRP1-knockout cells had similar iron responses as in IRP2-knockout cells (21). However, more studies are warranted to fully understand the Fe-S dependent iron response linked with IRP2, as well as its potential applications in iron-associated disorders (Fig. 1A).
Fig. 1: Cellular and mitochondrial iron homeostasis.
A: Iron dependent cellular iron homeostasis. From circulation, iron is imported into the cell 1) mostly as transferrin-bound iron (TBI) through TfR1-mediated endocytosis, which releases iron from the transferrin due to the difference in pH and is transferred to the cytosol as Fe2+ by DMT 1 or 2) to lesser degree as non-transferrin-bound iron by DMT1 directly into the cytosol. Iron is then transported to various organelles or labile iron pool, and excess iron is stored in ferritin protein. Cellular iron regulatory system is dependent on the availability of iron itself in the cell. During increased iron levels, there is an increased availability of Fe-S clusters (dependent on iron level) bound to the IRPs, therefore decreasing IRP-IRE interactions. These interactions result in decreased iron uptake by transferrin receptor (TfR), increased iron storage by ferritin (FTN), and increased iron export by ferroportin (FPN).
B: Mitochondrial iron regulation. Iron is imported into the intermembrane space via several proteins (e.g., porins, DMT1). Mitoferrin (MFRN) then imports the iron into the mitochondrial matrix, where it is utilized for Fe-S cluster biogenesis by the iron sulfur cluster assembly complex, as well as for energy production by the ETC. Excess iron is not only stored in mitochondria specific ferritin, but also exported by iron exporters such as ABCB7/8 in either elemental form or Fe-S clusters. Upon iron overload in the mitochondria, free iron content level is elevated, by which the Fenton reaction can produce highly reactive oxygen species, leading to increased oxidative damage.
2.3. Mitochondrial Iron Homeostasis
Mitochondria are vital for the survival of nearly all eukaryotic cells and organisms since they are essential for energy metabolism along with the generation of various intermediate metabolites (e.g., acetyl CoA, citrate, succinate, and fumarate) (22, 23) and the regulation of trace elements (e.g., copper, manganese, zinc, and iron) (24–26). Therefore, mitochondrial regulations of these elements and metabolites are critical for proper mitochondrial functions.
2.3.1. Iron and mitochondrial iron-sulfur cluster.
Iron plays an important role in the mitochondria where it is utilized in three major pathways: heme synthesis, Fe-S cluster biogenesis, and iron storage in the mitochondria-specific ferritin (FtMt) (15). Heme synthesis and Fe-S cluster biogenesis are highly interdependent because enzymes (e.g., ferrochelatase) involved in the heme synthesis pathway contain an Fe-S cluster (27, 28). Fe-S clusters are versatile prosthetic groups for the proteins involved in several essential cellular processes such as electron transfer, energy production, gene expression, and iron metabolism. These clusters, composed of ferrous/ferric iron and inorganic sulfide, are allowed for delocalization of electron cloud over both Fe and S, which is essential for the versatility of Fe-S clusters as co-factors (29). The synthesis of Fe-S clusters is facilitated by over 20 different protein components in the cytosol, mitochondria, and nucleus, where sulfur is sourced from cystine and is combined with iron to form a Fe-S cluster on a scaffold protein (30, 31). This Fe-S cluster is then incorporated into an apoprotein, which is utilized for required biochemical activities. It has been suggested that the machinery of Fe-S cluster synthesis was inherited from bacterial ancestors of mitochondria, and this system has been conserved in eukaryotes from yeast to human (31). Because of the versatility of the Fe-S cluster biosynthesis and its importance for essential cellular functions, any disruption in the Fe-S cluster formation can impair the overall iron homeostasis of the mitochondria and can be linked with severe diseases such as FA and Fe-S assembly enzyme myopathy (32, 33).
2.3.2. Mitochondrial Iron Regulation.
Mitochondria are a bilayer organelle. The outer mitochondrial membrane (OMM) is highly permeable to ions and small uncharged molecules via pore-forming membrane proteins (porins), while an inner mitochondrial membrane (IMM) is impermeable to most ions and small molecules. Porins in the OMM, such as voltage-dependent anion channel (VDAC), allow the transport of most of the metabolites involved in energy production, including pyruvate, ATP, cations, and others (34). Although the impermeability of IMM creates a high electrochemical gradient between the intermembrane space and the matrix, ions and molecules can be transported only through IMM-specific proteins. As a result, mitochondrial iron uptake is regulated by IMM protein mitoferrins (MFRNs), members of the mitochondrial carrier family (MCF) (15). MFRN1, also referred to as SLC25A37, is involved in erythroid cell development. Meanwhile, MFRN2, or SLC25A28, is ubiquitously expressed in different tissues (35–38), although the exact roles of MFRN2 have not been completely understood (35, 39).
In addition to the iron import and utilization mechanisms, it is also important for mitochondria to regularly export iron in the form of heme, Fe-S clusters, as well as elemental iron. Failure of proper iron export from the mitochondria can cause severe mitochondrial iron overload as seen in FA (27, 40) and various cardiomyopathies (41, 42). Although the mechanism of mitochondrial iron export is not clearly determined, it has been suggested that the members of the mitochondrial ABC family (e.g., ABCB7 (43) and ABCB8 (42)) are involved in mitochondrial transport of iron. In particular, ABCB7 is found to export mitochondrial iron via the formation of the Fe-S cluster (44, 45). Notably, it has been reported that ABCB7 could contribute to the regulation of cytosolic iron status since the deletion of ABCB7 gene hampers the formation of cytosolic proteins containing the Fe-S cluster (43, 46). The exact roles of ABCB8 have yet to be defined, although it appears to support mitochondrial export of iron (42) (Fig. 1B).
3. ROLE OF IRON IN NEURONAL FUNCTION
3.1. Iron and neurochemical processes
Iron is involved in the regulation of various neuronal functions, in which several neurotransmitters such as dopamine, norepinephrine (NE), epinephrine, and 5-hydroxytryptamine (5-HT; serotonin) are involved, and these neurotransmitters are synthesized by several iron-containing enzymes, including tyrosine hydroxylase, phenylalanine hydroxylase, and tryptophan hydroxylase (12). For example, studies have shown that iron-deficient children are more vulnerable to anxiety and/or depression (47). These conditions appear to have a significant relationship with altered brain homeostasis, including myelination, neurotransmission (e.g., monoamine metabolism), and energy metabolism (48).
The dopaminergic system is tightly regulated by changes in iron status. It has been suggested that decreased whole-brain iron levels may result in altered dopaminergic system in different brain regions. The dopamine receptor 1 (D1R) was reported to be lowered in the caudate putamen and prefrontal cortex in postweaning iron-deficient rats (11, 49). Nevertheless, contradictory findings have been also reported, as increased or decreased extracellular dopamine levels are observed in the striatum or prefrontal cortex, respectively, of rats exposed to postweaning iron deficiency (50, 51).
5-HT and NE are also reported to be altered by cellular iron status, as radioligand binding studies have demonstrated reduced 5-HT transporter densities in the striatum of iron-deficient mice (52). Moreover, extracellular NE levels are also found to be increased in iron-deficient rats, while NE transporter levels in the brain are decreased in the iron-deficiency condition (53, 54).
In addition, iron is required for the production and maintenance of myelin, which is the fatty white matter that insulates axons and helps preserve signaling (12). Brain iron deficiency leads to abnormal expression of myelin-related proteins, lipids, and cholesterol, since 80% of the brain cholesterol content is expressed as myelin (55–58). Lack of myelination causes delayed neuronal conduction, which was shown to be associated with retardation of reflexes in iron-deficient infants (59, 60).
3.2. Brain iron trafficking and regulation
Dietary iron (Fe3+) is reduced in the small intestine by the duodenal cytochrome B to Fe2+, which is then taken up by DMT1 into the duodenal epithelium and exits to the blood by a basolateral membrane transporter ferroportin (FPN). In the blood, Fe2+ is oxidized back to Fe3+ by ceruloplasmin or hephaestin and then binds to circulating transferrin (Tf). As described above, the iron-bound Tf interacts with TfR1 on the microvasculature of the blood-brain barrier (BBB) of the brain, which allows iron to enter the brain by endocytosis and transcytosis within various cell types in the central nervous system (CNS) (61–63). Once in the brain, iron is utilized for cellular metabolism or stored in ferritin when in excess. Alternatively, other cells including astrocytes and oligodendrocytes can take up excess iron in the low molecular weight form, such as iron-citrate, ATP, or ascorbate, which collectively represent the non-transferrin-bound iron (NTBI)-mediated transport pathway (64, 65).
In the brain, iron is more abundantly distributed in the white matter compared with the gray matter since significant iron staining is observed in oligodendrocytes of the matured brain (62, 66). Iron is highly required in oligodendrocytes which exhibit high metabolic rate and are responsible for axon myelination, and the Tim-2 receptor-mediated endocytosis of interstitial FTH has been identified as the major source of iron for oligodendrocytes due to their lack of TfRs (65, 67, 68). However, little is known about how and where iron is released from FTH and transported across the membrane into the cytosol. Although oligodendrocytes can synthesize Tf, its secretion from the cells is very limited (1). Instead, Tf is mainly synthesized and secreted by the choroid plexus, so that iron (Fe3+) is further transported to glia and neurons (11, 64, 65). Neurons can take up iron from Tf primarily via TfR1 as well as DMT1, and can export excess iron via FPN, the only known iron exporter, out of neuronal cells (8). Dysregulation of proteins involved in iron export may lead to iron accumulation in the brain and is further associated with neurodegeneration disorders such as aceruloplasminemia (ACP) (69) and PD (70) (Fig. 2).
Fig. 2: Brain Iron Homeostasis.
After entering the brain capillary endothelial cells (BCEC), transferrin-bound iron (TBI) is either 1) directly released into the brain via transcytosis or 2) converted to Fe2+ in the endosome after internalization of transferrin receptor 1 (TfR1) that is released to cytosol via divalent metal transporter 1 (DMT1) and then exported to the brain through ferroportin (FPN), forming the labile iron pool (LIP). Hepcidin could downregulate the expression of FPN and consequently trap iron in the cells. The labile iron in the brain is either stored in ferritin (FTN) or oxidized to Fe3+ by ceruloplasmin (CP), and Fe3+ is further bound to extracellular Tf secreted from the choroid plexus. Fe3+ that is released from astrocytes can bind to low molecular weight compounds such as ATP, ascorbate, and citrate (i.e., NTBI, non-transferrin-bound iron). Astrocytes also take up labile irons via DMT1 or other pathways such as TfR1, wherein those irons are then either stored in FTN or exported via FPN. CP can promote Fe2+ release by FPN. The extracellular TBI is taken up by neurons by TfR1-mediated endocytosis and undergoes internalization and acidification in endosomes where iron is released. The released Fe3+ is further reduced to Fe2+ and exported into the cytosol by DMT1, contributing to the LIP of neurons. Iron is exported from neurons by FPN, which is stabilized by amyloid precursor protein (APP). Microglia takes up iron and export ferritin through unknown pathways (possibly DMT1 and FPN), and the exported ferritin is further imported into oligodendrocytes by T-cell immunoglobulin mucin domain 2 protein (Tim-2).
3.3. Dysregulation of brain iron
Although iron status is tightly regulated in the brain, abnormally high or low iron levels are directly or indirectly involved in the pathophysiology of several neurodegenerative diseases, including AD, HD, PD, FA, and amyotrophic lateral sclerosis (ALS). Iron deficiency may impair the synthesis of essential iron-containing proteins and consequently hamper normal physiological functions. Untreated systemic iron deficiency can lead to iron-deficiency anemia, which is the most common type of anemia since the body lacks iron required to produce hemoglobin (71). Nevertheless, it has been shown that anemia is not only caused by iron deficiency, but also linked with chronic inflammation (72).
Recently, hepcidin, the master regulator of systemic iron homeostasis, is often elevated during inflammation (73) and has also been identified as a key player in brain iron homeostasis by controlling FPN expression post-translationally (74, 75). One of the possible sources of hepcidin in the brain is systemic hepcidin that can cross the BBB via transcytosis, although the exact mechanism remains unknown (76–78). In the brain, it has been also suggested that local cellular source of hepcidin is available from astrocytes or microglia (74, 76). Like systemic hepcidin, brain hepcidin appears to regulate iron homeostasis by downregulating the expression of FPN and iron importers such as DMT1. Both cell (79) and animal (80) studies have shown that increased hepcidin expression in the brain leads to both decreased cellular iron uptake (by decreasing TfR1 and DMT1) and decreased release (by decreasing FPN), although mixed results have been reported by different groups. A recent study has shown that overexpression of brain hepcidin in astrocytes leads to decreased brain iron level in AD mice (81). On the other hand, in studies using 6-hydroxydopamine (6-OHDA)-induced PD cell culture models, elevated levels of both hepcidin and DMT1 along with decreased expression of FPN were noted, resulting in iron accumulation (82). This suggests an independent regulation of DMT1 by 6-OHDA-induced inflammation apart from the hepcidin pathway, as reported by Jiang et al. (83). It was also reported that hepcidin knockdown in neuronal cells attenuated cellular iron burden by upregulation of FPN1 expression (82, 84), which is inconsistent with findings in the AD model. More studies are needed to identify and verify the molecular mechanisms of hepcidin and FPN in brain iron homeostasis.
Iron overload is also associated with several neuronal diseases. It has been well-recognized that age-related brain iron accumulation is linked with neurodegenerative diseases (e.g., PD and AD), and other iron overload diseases are caused by genetic mutations, which include hemochromatosis (i.e., Hfe), aceruloplasminemia (i.e., CP), and FA (i.e., FXN) (64, 85, 86). It is generally believed that excess free iron in the brain interacts with oxygen molecules to form reactive oxygen species (ROS) through the Fenton and Haber-Weiss reactions, which leads to oxidative stress in the neuronal cells (61, 87). Therefore, increased non-heme iron promotes the generation of ROS and consequently cellular damage and tissue fibrosis in the systemic organs (88).
Abnormal iron status can cause neurodegenerative diseases via mitochondrial dysfunction since iron is required for the formation of Fe-S clusters that are critical factors for oxidative phosphorylation (89). It has been reported that disruption of iron homeostasis results in altered mitochondrial morphology and function, as well as impaired mitochondrial DNA (90, 91). As a result, mitochondrial dysfunction has been recognized as an important hallmark for neurodegenerative diseases with iron dyshomeostasis. For instance, Huang et al. reported that iron overload results in reduced mitochondrial energy production along with induction of apoptosis, which is accompanied by mitophagic signaling (e.g., PINK1) (89).
3.4. Brain Iron and Mitochondrial Dynamics
Mitochondria are highly dynamic organelles so that they alter their functional and structural characteristics in response to various environmental changes. The process of fission or fusion allows mitochondria to divide into multiple smaller mitochondria or to combine into larger mitochondria, respectively, which not only contributes to attaining functional structures of mitochondria within a cell, but also facilitates the maintenance of mitochondrial homeostasis (92–94). Although the detailed mechanisms of mitochondrial fission and fusion are out of the scope of this review, it is important to understand how these mitochondrial dynamics are regulated by cellular iron status.
Mitochondrial fission and fusion are dependent on the common primary regulator, dynamin-related family proteins. The DRP1 (Dynamin-Related Protein 1) is a primary fission protein that is present in the cytosol and interacts with fission protein 1 (FIS 1) (95). During fission, DRP1 forms a ring structure around the mitochondria to constrict the OMM at the site of fission, creating smaller spherical mitochondria. On the other hand, the process of mitochondrial fusion is mediated by two primary proteins, MFN (Mitofusin) 1 and 2, and the dynamin-like GTPase protein Optic Atrophy 1 (OPA1) (94). The MFN proteins mediate the fusion of OMM, whereas the IMM fusion is mediated by the OPA1 protein (96). In normal cells, mitochondrial fission-fusion is well balanced, however this balance is disturbed in many disease conditions including neurodegenerative diseases. Following the impaired mitochondrial fission-fusion balance, mitochondrial morphology and activity are abnormally regulated due to diminished respiratory and bioenergetic capacities (97).
Along with the important roles of iron in mitochondrial oxidative function (98), dysregulated iron homeostasis has been also shown to be linked with an imbalance in mitochondrial fission and fusion in the central area. For example, several neurodevelopmental and neurodegenerative diseases, such as autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), and PD, have been found to be associated with prenatal and postnatal iron deficiency that results not only in depleted mitochondrial ATP production, but also in dysregulated mitochondrial dynamics and density in the neuronal cells (99, 100). In particular, iron-deficient neurons were shown to accelerate the fission process, as evidenced by decreased mitochondrial size, as well as reduced expression of fusion (OPA1) and increased expression of fission (DRP1) proteins (97).
In addition to iron deficiency, iron overload is also commonly observed in many neurodegenerative diseases (e.g., AD, PD, and HD) (101–103), and this excess iron has been shown not only to be linked with oxidative stress, but also to alter mitochondrial dynamics to prefer fission, rendering more fractioned mitochondria (104, 105). Although exact mechanisms are poorly understood, the iron overload-induced mitochondrial fission-fusion imbalance is likely to be secondary to the primary cause of neurodegenerative diseases, except for the Charcot-Marie-Tooth disease in which MFN2 defect plays a primary role (106). Although elevated DRP1 activity (i.e., fission) has been reported in iron overload-related neurodegenerative diseases (104, 107–109), there is no direct evidence that increased iron levels alter the fission-fusion balance towards fission. Nevertheless, it has been suggested that two mechanisms may be involved in iron overload-induced mitochondrial fission. First, iron overload-associated ROS production can stimulate mitochondrial fission through activating DRP1, as the amelioration of excess ROS was shown to prevent mitochondrial fragmentation as well as neurodegeneration process (109, 110). Furthermore, in neuronal cells with a deficiency of peroxiredoxin 5 that is important for eliminating H2O2, mitochondrial fragmentation and neurodegenerative characteristics were observed (108). Second, in neuron, iron overload-induced mitochondrial fission is probably linked with calcium signaling, as DRP1 activity has been shown to be regulated by cellular calcium levels. For example, elevated iron levels appear to accelerate calcineurin activity along with increased intracellular calcium levels, which could upregulate mitochondrial fission by DRP1 dephosphorylation at serine 637 and affect neurodegeneration (107, 111).
Collectively, altered mitochondrial dynamics are hallmarks of neurodegenerative diseases that are affected by either iron deficiency or overload, although more studies are warranted to fully understand a causal relationship between iron status and mitochondrial fission-fusion balance in neuronal cells.
4. NEURODEGENERATIVE DISEASES AND IRON OVERLOAD
Multiple factors contribute to the development of neurodegeneration, including aging, oxidative stress, neuroinflammation, and neuronal or protein dysregulation (e.g., dopaminergic neurons in PD and protein aggregation in AD). In addition, it has been revealed that both iron dysregulation and mitochondrial dysfunction are critical causal factors for neurodegenerative diseases. Ferroptosis, as an iron-dependent programmed cell death, has also been implicated in neurodegenerative disorders, including AD and PD (112). Ferroptosis is characterized by depletion of glutathione (GSH) and lipid peroxidation, with distinct morphological changes such as shrunken mitochondria (113) and ruptured mitochondrial outer membrane (114). Glutathione peroxidase 4 (GPX4) plays an important role in the regulation of ferroptosis, by catalyzing the reduction of lipid peroxides along with GSH (112). Furthermore, GSH binds to Fe2+ in the labile iron pool (LIP) and prevents it from being oxidized, acting as a ligand for ferrous iron and providing a substrate in the synthesis of the Fe-S cluster proteins (115).
While brain iron levels increase with age, upregulated iron-related proteins are observed in multiple brain regions of the elderly (64). For example, higher levels of ferritin and heme oxygenase-1 (HO-1) are detected in the substantia nigra (116) and hippocampus (117), respectively, which is linked with increased oxidative stress in the brain of older people. Nevertheless, the causality between iron accumulation and aging-related neurodegeneration has not been completely understood. Iron accumulation in the brain is likely to alter iron-related gene expression, which contributes to neurodegenerative diseases. Many of these disease conditions are also significantly correlated with mitochondrial dysfunction in the CNS, which is affected by iron transport and metabolism. In the following sections, several notable neurodegenerative diseases associated with iron overload are discussed with a focus on mitochondrial iron regulation.
4.1. Friedreich’s Ataxia
Friedreich’s ataxia (FA) is an autosomal recessive disorder that is caused by the mutation of FXN gene located on the ninth chromosome that codes for mitochondrial frataxin protein (118, 119). The mutation for FXN is most commonly identified by the intronic expansion of GAA trinucleotide repeat of the FXN gene (119). The trinucleotide repeats in FA patients can reach over 1000 GAA copies, and it has been shown that the number of these repeats is inversely proportional to the age of onset for the disease (118). However, trinucleotide expansion is not the only cause for abnormal FXN expression since several epigenetic factors (120), FXN point mutation (121, 122), and compound heterozygosity for GAA expansion (123) have also been observed in a smaller fraction of FA patients. This disease is commonly characterized by the loss of positional awareness, muscle dystrophy, hypertrophic cardiomyopathy, and diabetes.
Mature human FXN is a 210-amino acid protein (~23 kDa), which is essential for iron homeostasis and metabolism (124), as well as many enzymatic actions as co-factor. Frataxin is a nuclear encoded protein that is later transported to mitochondria (32) wherein it contributes to the production of Fe-S clusters since FXN-ablated cells show reduced abundances of Fe-S cluster proteins (125). FXN has also been shown to interact with ISD11, a component of the Fe-S cluster assembly complex, supporting the idea that FXN is required for the formation of Fe-S clusters (126, 127). These clusters are also involved in several key proteins including mitochondrial respiratory chain components (e.g., complex I and II), aerobic metabolic enzymes (e.g., aconitase, citrate, fumarase), and other proteins in the nucleus (for DNA synthesis and repair mechanism) and cytoplasm (for ribosome biogenesis) (128, 129).
Under normal iron status, healthy cells promote the production of Fe-S clusters, while inhibiting the import of extra iron by the action of iron regulatory proteins (IRP1 and IRP2) (130, 131). However, in FA, even in normal iron status, the Fe-S cluster formation is diminished, and other iron regulatory system (IRP1, TfR, and ferritin) is also dysregulated. Interestingly, a recent study showed that a long-term running exercise has an effect to restore IRP1 expression without changing FXN levels using KIKO mice, a Knock in-Knock out FA mouse model (81), suggesting that a molecular mechanism of mitochondrial iron homeostasis may be independent of FXN in FA.
Reactive oxygen species (ROS) are mainly produced from the mitochondrial electron transport chain (ETC) as byproducts of energy production. In FA, it has been reported that ROS are produced in an uncontrolled manner, which can react with proteins, nucleic acids, and lipids, causing their damages (132). In FXN-deficient cells, Fe-S cluster formation is ablated in the ETC, which not only increases electron leakage but also results in ROS production in the mitochondria (133). For instance, the ratio of reduced to oxidized glutathione (GSH/GSSH) was significantly decreased in FA cells (134), suggesting FA-induced increase in oxidative stress (e.g., H2O2 production). The increased H2O2 and free iron in the mitochondria may facilitate the production of hydroxyl radical (•HO) via the Fenton reaction (132). However, hydroxyl radicals are not the only ROS that are produced in FA cells, and the peroxides also react with membrane lipids to produce carbon centered radicals (R•) that will further react with oxygen to produce reactive hydroperoxyl radicals (ROO•) (133). They are highly reactive species that can transfuse through membranes, which causes oxidative stress-induced damages. Furthermore, it has been shown that a loss of FXN in FA dorsal root ganglia blunts the expression of Nrf2 (Nuclear Factor Erythroid 2-related Factor 2), which in turn inhibits the expression antioxidants such as glutathione S-transferase, peroxiredoxin, and glutaredoxin (135). Together these changes decrease the antioxidative capacity and sensitize the cells to oxidative stress.
Neurodegeneration in FA is known to lead speech impairment, progressive ataxia, and an overall decrease in cognitive capabilities (135). It has been shown that FA-linked neurodegeneration is associated with defects in Fe-S cluster assembly unit where FXN plays a key role for the cluster formation with other proteins (136). Nevertheless, myopathy patients with Fe-S cluster assembly mutations are found to have no neurodegenerative symptoms, although they have FA-like characteristics such as iron accumulation as well as decreased aconitase activity (137). Thus, it may be plausible that other factors are responsible for FA-linked neurodegeneration other than FXN-based Fe-S cluster formation unit. More studies are required to elucidate the mechanisms of disease.
Furthermore, ferroptosis has been identified as a possible mechanism for neurodegeneration in FA patients (138, 139). In neuronal cells, Nrf2 (a key regulator of ferroptosis) has been shown to regulate expression of several oxidative stress resistance genes such as HO-1, thioredoxin (Trx), and glutamate cystein ligase (GCL) (135, 140, 141). Although Nrf2 is usually located in the cytoplasm, it is translocated to the nucleus under oxidative stress where it binds to the antioxidant-responsive element (ARE) in the DNA and induces transcription of antioxidant/cytoprotective genes (140). Both in vivo and in vitro studies have shown that Nrf2 expression is dependent on FXN levels in neuronal cells and tissues (138, 142). In FA, lower FXN levels appear to downregulate Nrf2 expression in neurons, which renders these cells more susceptible to oxidative damage and results in ferroptosis (138, 143). These studies suggest Nrf2-linked ferroptosis as a chief pathway for FA neurodegeneration (138, 142). Other than its role in ferroptosis, Nrf2 has also been shown to be important for neurogenesis (143). A recent study shows that reduced Nrf2 expression in FA neural stem cells inhibits impairment in neurogenesis (138, 143). However, upon recovery of Nrf2 function, the proliferation and regeneration pattern of these stem cells were significantly improved (138, 143). Recovering of Nrf2 also triggers axonal re-growth, which can possibly help in the formation of better neural networks (143). The increasing evidence for the role of Nrf2 in FA neurodegeneration suggests its possibility as a therapeutic target. On the other hand, oxidative stress has been shown to alter the mitochondrial fission/fusion balance in FA (144), so it is plausible that restoration of Nrf2 expression could correct the impaired mitochondrial dynamics possibly by alleviating oxidative stress, although more studies are needed to verify this idea.
Currently, pharmacological treatments for FA include agents that are designed to reduce oxidative stress as well as to enhance mitochondrial function (e.g., idebenone, coenzyme Q10, A001, and omaveloxolone), anti-inflammatory agents (e.g., methylprednisolone), iron chelator (e.g., deferiprone), FXN modifiers or mimetics (e.g., carbamylated erythropoietin, ubiquitin competitors, and CTI-1601), and agents that induce FXN gene expression (e.g., RG2833, nicotinamide, interferon gamma, and resveratrol) (145). Recently, mitochondrial fragmentation (i.e., DRP1) has been identified as a therapeutic target for FA patients, as reversal of mitochondrial fragmentation appeared to restore impaired bioenergetics and ATP production in FA patient-derived cells (146). Nevertheless, further studies are required to determine the efficacy and safety of these therapeutic options in FA patients.
4.2. Huntington’s Disease
Huntington’s Disease (HD) is a progressive neurodegenerative disease caused by the expansion of a CAG trinucleotide repeat in exon 1 of the huntingtin (HTT) gene (147). Western European ancestry are more vulnerable to HD, with the prevalence of ~12 individuals per 100,000, due to their genetic differences in the HTT gene (148). Typical clinical symptoms of HD include chorea (involuntary movements), neuropsychiatric disturbances, dementia, and weight loss (149), which may be related to impaired mitochondrial ATP synthesis (150). Mutant HTT not only induces neuronal loss in the basal ganglia, but also leads to movement and cognitive deficits (151). In early stage of HD, destruction of neurons and astrogliosis in the striatum is reported to accompany iron overload (152, 153). As the disease develops to the advanced stage, the neuronal damage starts to affect the cortex and hippocampus, as well as respiratory chain (154, 155).
Studies have reported that healthy HTT is responsible for mediating endocytosed Fe2+ that is required for oxidative energy production (149, 156). Therefore, its dysregulation is responsible for the functional impairment of mitochondria. For example, multiple studies have reported that HTT mutation induced-neuronal death is linked with disrupted mitochondria energy metabolism, including decreased mitochondrial respiratory complexes II-IV and aconitase activity (157, 158), abnormal mitochondrial trafficking (159), and impaired mitochondrial ATP production (160). Since HTT is involved in nuclear gene expression, its mutation is believed to be correlated with disrupted transcription, as well as import of mitochondrial proteins (161–164). Mutant HTT is likely to increase dynamin-related protein-1 (Drp1), a mitochondrial fission GTPase protein, in the postmortem brain of HD patients (165), which results in favor of fission in the fission-fusion dynamic balance of mitochondria (166).
Iron overload, in the case of HD, appears to be an early event instead of a founding event (151) (Fig. 3B). Magnetic resonance imaging (MRI) studies showed changes in brain iron metabolism in early stage of HD (167, 168). Iron accumulation is observed in the basal ganglia (153, 169), microglia (170), and myelinating oligodendrocytes of HD patients, which further leads to increased oxidative stress and damage to adjacent neurons (171). Consistent with HD patients, HD animal models also show increased iron and ferritin levels in neurons and glia (170, 172). One hypothesis of the relationship between increased brain iron and HD is the dysregulated iron homeostatic pathways that are affected by mutant HTT. Niu et al. demonstrated that mutation of HTT induces upregulation of IRP1, Tf, and TfR in both in vivo and in vitro studies, along with increased iron levels in the striatum and cortex in 12-week-old HD mice, although no HD symptoms were observed at this age (173). Multiple studies using different chelators have been conducted to investigate whether the progression of HD can be ameliorated by removal of excess brain iron. Notably, chelation of excess brain iron by deferoxamine (DFO) improves behavioral and pathological symptoms in HD mice (172) and HD cells (174), respectively. Another metal-binding compound clioquinol was shown to ameliorate neurodegenerative symptoms in HD mice, as well as to downregulate mutant HTT expression in PC12 cells (175). However, most studies are limited to early-stage HD, and more studies are needed to determine the role of iron in late-stage HD.
Fig. 3: Neurodegenerative diseases with brain iron accumulation.
A: Friedreich’s ataxia. In Friedreich’s ataxia (FA), mutations of the mitochondrial protein frataxin (FXN) lead to impaired Fe-S cluster biogenesis which further leads to disruption of iron metabolism along with increased mitochondrial iron contents. Excessive accumulation of iron leads to the production of reactive oxygen species (ROS), causing lipid peroxidation and cell death. Mutation of frataxin has also been shown to impair the translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), a key player for antioxidant gene expressions, into the nucleus, which not only impairs antioxidant response, but also contributes to the ROS production.
B: Iron dysregulation in Huntington’s disease. In Huntington’s disease (HD), the presence of mutant Huntington (mHTT) leads to increased sensitivity of NMDA receptors, which not only increases influx of extracellular calcium (Ca2+), but also results in the activation of neurotoxic Rhes, rendering more toxic soluble mHTT. The mHTT interacts with mitochondria and it results in the impaired mitochondrial membrane permeability as well as the efflux of Ca2+ from mitochondria. Increased cytosolic Ca2+ leads to the activation of neuronal nitric oxide synthase (nNOS), which binds to NMDA receptors via Dexras1, releasing NO that eventually forms peroxynitrite radicals in the presence of superoxides. The nNOS also binds to divalent metal transporter 1 (DMT1), which results in enhanced Fe2+ influx, as well as increased oxidative stress via the Fenton reaction, while interacting with H2O2 released from superoxide dismutase (SOD). These reactions ultimately lead to increased lipid peroxidation and neuronal cell death.
C: Iron dysregulation in Alzheimer’s disease. Alzheimer’s disease (AD) is characterized by the aggregation of Aβ plaque and phosphorylated Tau protein (neurofibrillary tangle, NFT). In normal condition, APP stabilizes ferroportin (FPN) to facilitate iron export, and Tau protein supports the trafficking of APP transport to the cell surface. In AD, Tau is phosphorylated and aggregated to NFT, leading to less Tau available to facilitate APP transport, and APP is cleaved to generate Aβ, so the iron efflux via FPN is impaired, leading to iron accumulation in cells. Iron can bind to Aβ and promotes its aggregation, leading to disrupted mitochondrial membrane permeability, producing oxidative stress within the mitochondria. Iron dysregulation also alters Aβ metabolism at post-transcriptional level and causes DNA damage and mutation. Taken together, these disruptions lead to increased release of Ca2+, apoptosis-inducing factor (AIF) and cytochrome c, resulting in apoptosis and neuronal cell death.
D: Iron dysregulation in Parkinson’s disease. In Parkinson’s disease (PD), Fe2+ levels are elevated by both increased DMT1-mediated import and neuroinflammation, which leads to oxidative damage by the Fenton reaction. The reduction from Fe3+ to Fe2+ is accelerated in the presence of α-synuclein, while the oxidation from Fe2+ to Fe3+ is inhibited due to a lack of ceruloplasmin (CP). The Fe2+ overload induced-oxidative stress exacerbates the aggregation of misfolded α-synuclein, by which Lewy bodies are accumulated and it could be the cause of dementia. Disrupted Tf/TfR2 transport system can lead to an iron accumulation in mitochondria, which may induce mitochondrial dysfunction along with mitochondrial oxidative stress. Another characteristic of PD is decreased release of dopamine, which also promotes the misfold of α-synuclein, either directly or indirectly leading to neuronal cell death.
Apart from iron accumulation, impaired iron regulatory system is likely to be linked with mitochondrial dysfunction in the brain of HD patients where it is featured by increased expression on mitoferrin 2 (MFRN2) (176), a mitochondrial iron importer from the cytosol (36). To test whether mitochondria are a key site of iron dysregulation in HD, Agrawal et al. found increased labile iron in the mitochondria of both human and mouse HD brain at an advanced stage, which was shown to be rescued by a chelating agent, deferiprone (DFP), suggesting that mitochondrial labile iron overload might be a mediator of HD mitochondrial dysfunction, as well as disease progression (149). In addition, the expression patterns of heme-based mitochondrial respiratory chain complexes and Fe-S cluster enzymes are altered in the striatum of HD patients (177, 178). Also, FXN was found to be downregulated in the brains of mouse and human HD subjects, which is a common feature of FA (described above) (149). It appears plausible that both deficient FXN and oxidative stress could play an important role in impaired mitochondrial Fe-S cluster biosynthesis in HD (159).
4.3. Alzheimer’s Disease
Alzheimer’s Disease (AD) is a chronic multifactorial neurodegenerative condition associated with progressive dysfunction and degeneration of neurons, which leads to progressive cognitive decline and dementia (179). In the brain of AD patients, the accumulation of extracellular β-amyloid (Aβ) plaques and the deposit of neurofibrillary tangles (NFTs) formed by hyperphosphorylation of tau (pTau) proteins are considered to be the main pathophysiological features. The etiology of AD is diverse with a combination of genetic and environmental factors so that the specific mechanisms of its onset and corresponding treatment are still unclear. Growing evidence has shown that iron plays an important role in the early progression of AD and is involved in the development of Aβ plaques and NFTs (180).
Abnormal iron levels result in diminished iron regulatory enzyme activity, which not only increases Aβ production through its binding with amyloid precursor proteins (APP) and tau proteins (181), but also promotes oxidative stress and cytotoxicity along with the plaques (182, 183). The intracellular iron has been reported to accelerate Aβ deposition and production at the post-transcriptional level. For instance, the increased iron level upregulates the expression of APP proteins via the regulation of the atypical IRE in 5’UTR of APP transcripts, leading to increased Aβ level as well as aggregation (184–186). However, it has been argued that the Aβ conjugated with iron could be a protective response of the neuron from oxidative stress through sequestering iron (187, 188). Nevertheless, the cellular mechanisms of iron in the progression of AD are still unclear, which warrants further studies.
Similar to Aβ, the aggregation of pTau leads to the deposition of NFTs via tauopathy, which is induced by Fe3+ (189). In physiological conditions, tau promotes the stabilization and regulation of microtubule and axonal transport, which is critical for neurotransmission as well as iron homeostasis (190). However, upon CDK5/P25 complex and GSK-3b, the Fe3+-induced pTau hinders the microtubule stabilization in NFTs (191). Meanwhile, the aggregation of tau in NFTs induces the expression of HO-1 and also increases the release of Fe2+ (192), which elevates oxidative stress and further promotes the aggregation of pTau (193). In addition to cellular damage, the iron-associated tau protein deposition hampers iron export, which in turn results in iron trapping in the cells and leads to a vicious cycle (194). Therefore, iron chelation therapies have been applied and shown to have promising efficacies on ameliorating AD symptoms. For example, Guo et al. revealed that DFO has a significant effect to inhibit the iron-induced pTau aggregation in an AD mouse model (191). Other chelators such as clioquinol and 5,7-dichloro-2-((dimethylamino)methyl)quinolin-8-ol (PBT2) were also shown to decrease Aβ1–42 levels and improved cognitive function in AD patients (195, 196).
In addition, oxidative stress has been suggested to contribute to the production of Aβ (197, 198), which appears to be linked with abnormal mitochondrial function and morphology. These dysregulated mitochondria have been identified in the early stage of AD, which is featured by reduced ATP production in the brain (8). In the mitochondria, for example, an active γ-secretase complex cleaves APP into Aβ, which inhibits mitochondria complex IV through Aβ-bound alcohol dehydrogenase (ABAD) that produces ROS (192). Thus, mitochondria are recognized as major organelle for ROS and oxidative stress (151), but further studies are required to better understand how iron-related oxidative stress is linked with mitochondrial dysfunction in AD (Fig. 3C).
4.4. Parkinson’s Disease
Parkinson’s Disease (PD) is the second common neurodegenerative disease of the elderly, in which dopaminergic neuronal loss and iron accumulation in the substantia nigra (SN) pars compacta have been recognized as pathological features (199). Iron accumulation within the SN has been well reported using different analytical methods, including inductively-coupled plasma mass spectrometry (ICP-MS) (190), MRI (200), and atomic absorption spectroscopy (201, 202). The exact molecular mechanism of PD has not yet been fully understood due to the complex pathology of the disease, although other factors such as misfolded proteins, impaired ubiquitin-proteasome, dysfunction of mitochondria, and oxidative stress are considered for the pathogenesis of PD (203).
In PD, iron binds to α-synuclein, an abundant protein in dopamine neurons, and accelerates its aggregation, causing the production of toxic hydroxyl radical in the SN (12). In the affected dopaminergic neurons, iron is also thought to contribute to the formation of Lewy bodies, which are intraneuronal proteinaceous inclusions that result in an aggregation of α-synuclein and are considered to be a pathological hallmark of PD (199). This clustered α-synuclein disturbs cytosolic and mitochondrial environments and dopamine transporters, which further results in activation of cell death (183) (Fig. 3D). Moreover, PD patients also show increased levels of ferritin-loaded microglia in the SN, where degenerating and neuromelanin-loaded dopaminergic cells are shown to have more reactive microglia (204). The upregulation of both glial cells by the debris and toxins released by dying neurons appear to contribute to increased neuromelanin, which, upon binding to iron, leads to the release of neurotoxic factors including tumor necrosis factor-α, inflammation marker interleukin-6 (IL-6), and nitric oxide (183). The immunoreactivity of HO-1 is also reported to be increased in the neuropil of the substantia nigra (205).
As a redox-active iron pool, mitochondrial dysfunction in PD has been studied as early as in the 1980s, when four college students developed Parkinson-like symptoms after the injection of drugs that contain MPTP, a compound whose metabolite MPP+ inhibits the mitochondrial complex I capacity (206, 207). More studies have also confirmed dysregulated mitochondrial complex I activity and content levels in the brain of PD patients (208, 209). This lessened mitochondrial complex I capacity seems to elevate mitochondrial iron uptake and ROS production (210). It has been identified that iron chelation- and gene-based therapies are effective to restore ferritin and to further rescue the parkinsonism symptoms in animal models of PD (12). In a pilot clinical trial with early-stage PD patients, the DFP treatment reduced iron overload in the substantia nigra and significantly improved the motor ability (211), indicating that chelation therapies have the potential to slow down the disease progression and provide treatment options for further long-term therapy.
PD has been shown to compromise iron regulatory system along with mutations in iron-related proteins such as Tf (212), IRP2 (213), ferritin (214), and DMT1 (215). For instance, ferritin levels are decreased in the brain of PD patients, indicating reduced iron storage capacity (216). DMT1 appears to be increased in nigral dopamine neurons and microglia, which may not only contribute to the increased capacity of Fe2+ transport from the extracellular space into the cytosol, but also increase the production of ROS (217). Therefore, iron accumulation is likely to exacerbate mitochondrial dysfunction in PD patients (199).
4.5. Neurodegeneration with Brain Iron Accumulation (NBIA)
Neurodegeneration with brain iron accumulation (NBIA) is a group of rare inherited monogenic diseases characterized by iron accumulation in different parts of the brain, especially within the basal ganglia, with the symptoms of painful dystonia, parkinsonism, neuropsychiatric abnormalities, and early death. There is no cure for NBIA disorders. NBIA displays a marked genetic heterogeneity, wherein the mutations of each specific gene lead to the failure of each protein for essential cellular functions. Ten different subtypes of NBIA are summarized in Table 1 (218, 219). The majority of these genes are found in the mitochondria, including PANK2, CoPAN, PLA2G6, C19orf12, and ATP13A2, and it is thus important to understand iron regulatory mechanisms and its connections between mitochondria function and neurodegeneration in the brain (220).
Table 1.
NBIA disorder | Gene | Symptoms |
---|---|---|
PKAN (Pantothenate Kinase-Associated Neurodegeneration) | PANK2 |
Classic PKAN: gait problems, progressive dystonia, retinal degeneration, dysarthria, rigidity, spasticity, hyperreflexia, and extensor toe signs Later onset PKAN: speech difficulty, psychiatric symptoms |
PLAN (PLA2G6-Associated Neurodegeneration) | PLA2G6 |
INAD (infantile neuroaxonal dystrophy): hypotonia, spasticity, optic atrophy aNAD (atypical neuroaxonal dystrophy): speech delay, dystonia, behavior changes PLA2G6-related dystonia-parkinsonism: dystonia, neuropsychiatric changes, slowness, poor balance and rigidity |
MPAN (Mitochondrial membrane Protein-Associated Neurodegeneration) | C19orf12 | Dystonia, spasticity, weakness, optic atrophy, and neuropsychiatric changes |
BPAN (Beta-propeller Protein-Associated Neurodegeneration) | WDR45 | Childhood development delay and seizures, slow motor, and cognitive gains (mutant on the X chromosome, lethal in most males) |
FAHN (Fatty Acid Hydroxylase-associated Neurodegeneration) | FA2H | Leg dystonia, weakness and falling, optic atrophy, dysphagia (difficulty swallowing), progressive intellectual impairment |
CoPAN (COASY Protein-Associated Neurodegeneration) | COASY |
Early in the disease: spastic-dystonic paraparesis in lower limbs Later in the disease: dystonia of the mouth and jaw, speech problems caused by dysarthria |
Woodhouse-Sakati Syndrome | DCAF17 | In Saudi Arabian population: dystonia, hair loss, diabetes, hearing loss, gonadal dysfunction, and mental retardation |
Neuroferritinopathy | FTL | Dystonia, chorea (jerky movements), subtle cognitive decline |
Aceruloplasminemia | CP | Iron accumulation in brain as well as other organs, retinal degeneration, diabetes, ataxia, involuntary movement, anemia |
Kufor-Rakeb syndrome | ATP13A2 | Juvenile parkinsonism, dementia, abnormal eye movements and involuntary jerking of facial and finger muscles |
Although being called NBIA, only two subtypes are directly related to iron metabolism: aceruloplasminemia (ACP), which is associated with recessive mutations in ceruloplasmin (CP), and neuroferritinopathy, which is associated with mutations in ferritin light chain 1 (FTL1). These two proteins and their subtypes are involved in different mechanisms, such as lipid metabolism, mitochondrial function, coenzyme A (CoA) metabolism, and autophagy (6).
Ceruloplasmin, encoded by CP gene, is the only existing ferroxidase that is expressed as a glycosylphosphatidylinositol (GPI)-linked form in astrocytes and catalyzes the peroxidation of Fe2+-Tf to Fe3+-Tf (220, 221). Aceruloplasminemia (ACP) that is caused by the mutation of CP gene induces the absence of the corresponding protein in the plasma and is characterized by extrapyramidal symptoms, ataxia, progressive CNS, and retinal neurodegeneration (183, 222). The mutations of the CP gene cause neuronal structural modifications, so that the type I copper binding site fails to incorporate copper into the apo-CP, an unstable apo form of CP that will be rapidly degraded in the plasma, leading to retention of CP proteins in the endoplasmic reticulum (ER), as well as impaired ferroxidase activity (223, 224). Consequently, iron enters the CNS as Fe2+ where it is not oxidized, leading to excess NTBI. In addition, mutations of CP also contribute to the destabilization of FPN on the cell surface, so that iron export is impaired (223). Collectively, in ACP patients, cellular iron is accumulated in astrocytes and is not delivered to the neurons, which causes neuronal cell death due to iron deficiency (225).
Neuroferritinopathy is an autosomal inherited, late-onset basal ganglia disease linked to mutations in FTL1 that is one of the two ferritin subunits. Individuals with neuroferritinopathy show nuclear and cytoplasmic ferritin accumulation in both neurons and glia of the striatum, as well as cerebellar cortex, which contributes to increased cellular labile iron pool along with elevated ROS production and oxidative stress (183, 226, 227). Consistently, it has been suggested that the iron accumulation-linked oxidative stress causes mitochondrial dysfunction and cell damage, as in the neurodegenerative rodent model, markers of oxidative stress (e.g., lipid peroxidation) are increased along with the dysregulated iron regulatory system (e.g., TfR1 and IRP1) (228).
5. CONCLUSIONS
There has been increased attention on the roles of iron in the CNS since mitochondrial function and structure are largely dependent on the optimal range of iron level in the central tissues. In particular, neurodegenerative diseases have shown to be notably linked with irregular iron status, which also causes impaired mitochondrial function along with oxidative stress. Here, we discussed how dysregulated iron homeostasis alters the mitochondrial system in multiple neurodegenerative diseases: Alzheimer’s disease, Huntington’s disease, Parkinson’s disease, neurodegeneration with brain iron accumulation, and Friedreich’s ataxia. Remarkably, we underscore that iron overload may be an important causal and/or susceptibility factor for these neurodegenerative diseases through impinging on mitochondrial functions and structures. The present review also summarized current understanding on therapeutic targets of these neurodegenerative diseases, while focusing on the mitochondrial iron system. In addition to approaches with various iron chelators, we have also discussed mitochondria-specific targets that include mitochondrial dynamics (i.e., fission and fusion), as well as mitochondrial iron regulatory system. More mechanistic and systematic studies are required to better understand the underlying mechanisms of neurodegenerative diseases associated with iron overload and associated mitochondrial dysfunction, and these efforts will help to identify novel therapeutic strategies that can prevent the development of neurodegenerative diseases and ameliorate disease symptoms or slow down the disease progression.
ACKNOWLEDGMENTS
This work was supported in part by the NIH HL143020 and AG074472 (J.K.).
Abbreviations:
- ACP
aceruloplasminemia
- AD
Alzheimer’s disease
- APP
amyloid precursor protein
- BBB
blood-brain barrier
- BCEC
brain capillary endothelial cells
- CNS
central nervous system
- CP
ceruloplasmin
- DMT1
divalent metal transporter 1
- ER
endoplasmic reticulum
- ETC
electron transport chain
- FPN
ferroportin
- FA
Friedreich’s Aataxia
- FTH
ferritin heavy chain
- FTL
ferritin light chain
- FtMt
mitochondria specific ferritin
- FXN
frataxin
- GPX
glutathione peroxidase
- GSH
glutathione
- HD
Huntington’s disease
- HO-1
heme oxygenase 1
- HTT
huntingtin
- IMM
inner mitochondrial membrane
- IRE
iron responsive element
- IRP
iron regulatory protein
- MFRN
mitoferrin
- NBIA
neurodegeneration with brain iron accumulation
- Nrf2
nuclear factor erythroid 2-related factor 2
- NTBI
non-transferrin-bound iron
- OMM
outer mitochondrial membrane
- PD
Parkinson’s disease
- ROS
reactive oxygen species
- SN
substantia nigra
- TBI
transferrin-bound iron
- Tf
transferrin
- TfR1
transferrin receptor 1
- Tim-2
T-cell immunoglobulin mucin domain 2 protein
- UTR
untranslated region
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.
Declaration of interests
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
REFERENCES
- 1.Qian ZM, Ke Y. Brain iron transport. Biological Reviews. 2019;94(5):1672–84. [DOI] [PubMed] [Google Scholar]
- 2.Moos T, Morgan EH. The metabolism of neuronal iron and its pathogenic role in neurological disease: review. Ann N Y Acad Sci. 2004;1012:14–26. [DOI] [PubMed] [Google Scholar]
- 3.Ayton S, Lei P, Bush AI. Metallostasis in Alzheimer’s disease. Free Radic Biol Med. 2013;62:76–89. [DOI] [PubMed] [Google Scholar]
- 4.Hare DJ, Double KL. Iron and dopamine: a toxic couple. Brain. 2016;139(Pt 4):1026–35. [DOI] [PubMed] [Google Scholar]
- 5.Liu C, Liang MC, Soong TW. Nitric Oxide, Iron and Neurodegeneration. Front Neurosci. 2019;13:114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Meyer E, Kurian MA, Hayflick SJ. Neurodegeneration with Brain Iron Accumulation: Genetic Diversity and Pathophysiological Mechanisms. Annu Rev Genomics Hum Genet. 2015;16:257–79. [DOI] [PubMed] [Google Scholar]
- 7.Li K Iron Pathophysiology in Friedreich’s Ataxia. Adv Exp Med Biol. 2019;1173:125–43. [DOI] [PubMed] [Google Scholar]
- 8.Ndayisaba A, Kaindlstorfer C, Wenning GK. Iron in Neurodegeneration - Cause or Consequence? Front Neurosci. 2019;13:180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Huang J, Chen S, Hu L, Niu H, Sun Q, Li W, et al. Mitoferrin-1 is Involved in the Progression of Alzheimer’s Disease Through Targeting Mitochondrial Iron Metabolism in a Caenorhabditis elegans Model of Alzheimer’s Disease. Neuroscience. 2018;385:90–101. [DOI] [PubMed] [Google Scholar]
- 10.Shokolenko I, Venediktova N, Bochkareva A, Wilson GL, Alexeyev MF. Oxidative stress induces degradation of mitochondrial DNA. Nucleic Acids Res. 2009;37(8):2539–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Beard JL. Iron biology in immune function, muscle metabolism and neuronal functioning. J Nutr. 2001;131(2S-2):568S–79S; discussion 80S. [DOI] [PubMed] [Google Scholar]
- 12.Hare D, Ayton S, Bush A, Lei P. A delicate balance: Iron metabolism and diseases of the brain. Front Aging Neurosci. 2013;5:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Aisen P, Enns C, Wessling-Resnick M. Chemistry and biology of eukaryotic iron metabolism. Int J Biochem Cell Biol. 2001;33(10):940–59. [DOI] [PubMed] [Google Scholar]
- 14.Wang J, Pantopoulos K. Regulation of cellular iron metabolism. Biochem J. 2011. ;434(3):365–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lane DJ, Merlot AM, Huang ML, Bae DH, Jansson PJ, Sahni S, et al. Cellular iron uptake, trafficking and metabolism: Key molecules and mechanisms and their roles in disease. Biochim Biophys Acta. 2015;1853(5):1130–44. [DOI] [PubMed] [Google Scholar]
- 16.Gammella E, Buratti P, Cairo G, Recalcati S. The transferrin receptor: the cellular iron gate. Metallomics. 2017;9(10):1367–75. [DOI] [PubMed] [Google Scholar]
- 17.Kawabata H. Transferrin and transferrin receptors update. Free Radic Biol Med. 2019;133:46–54. [DOI] [PubMed] [Google Scholar]
- 18.Ohgami RS, Campagna DR, Greer EL, Antiochos B, McDonald A, Chen J, et al. Identification of a ferrireductase required for efficient transferrin-dependent iron uptake in erythroid cells. Nat Genet. 2005;37(11):1264–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Pantopoulos K Iron metabolism and the IRE/IRP regulatory system: an update. Ann N Y Acad Sci. 2004;1012:1–13. [DOI] [PubMed] [Google Scholar]
- 20.Wang H, Shi H, Rajan M, Canarie ER, Hong S, Simoneschi D, et al. FBXL5 Regulates IRP2 Stability in Iron Homeostasis via an Oxygen-Responsive [2Fe2S] Cluster. Mol Cell. 2020;78(1):31–41 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Terzi EM, Sviderskiy VO, Alvarez SW, Whiten GC, Possemato R. Iron-sulfur cluster deficiency can be sensed by IRP2 and regulates iron homeostasis and sensitivity to ferroptosis independent of IRP1 and FBXL5. Science Advances. 2021;7(22):eabg4302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Frezza C Mitochondrial metabolites: undercover signalling molecules. Interface Focus. 2017;7(2):20160100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Martinez-Reyes I, Chandel NS. Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun. 2020;11(1):102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Gille G, Reichmann H. Iron-dependent functions of mitochondria--relation to neurodegeneration. J Neural Transm (Vienna). 2011;118(3):349–59. [DOI] [PubMed] [Google Scholar]
- 25.Walter PB, Knutson MD, Paler-Martinez A, Lee S, Xu Y, Viteri FE, et al. Iron deficiency and iron excess damage mitochondria and mitochondrial DNA in rats. Proc Natl Acad Sci U S A. 2002;99(4):2264–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kashiv Y, Austin JR 2nd, Lai B, Rose V, Vogt S, El-Muayed M. Imaging trace element distributions in single organelles and subcellular features. Sci Rep. 2016;6:21437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Huang ML, Becker EM, Whitnall M, Suryo Rahmanto Y, Ponka P, Richardson DR. Elucidation of the mechanism of mitochondrial iron loading in Friedreich’s ataxia by analysis of a mouse mutant. Proc Natl Acad Sci U S A. 2009;106(38):16381–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Liu G, Sil D, Maio N, Tong WH, Bollinger JM Jr., Krebs C, et al. Heme biosynthesis depends on previously unrecognized acquisition of iron-sulfur cofactors in human amino-levulinic acid dehydratase. Nat Commun. 2020;11(1):6310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Johnson DC, Dean DR, Smith AD, Johnson MK. Structure, function, and formation of biological iron-sulfur clusters. Annu Rev Biochem. 2005;74:247–81. [DOI] [PubMed] [Google Scholar]
- 30.Lill R, Muhlenhoff U. Maturation of iron-sulfur proteins in eukaryotes: mechanisms, connected processes, and diseases. Annu Rev Biochem. 2008;77:669–700. [DOI] [PubMed] [Google Scholar]
- 31.Lill R, Dutkiewicz R, Elsasser HP, Hausmann A, Netz DJ, Pierik AJ, et al. Mechanisms of iron-sulfur protein maturation in mitochondria, cytosol and nucleus of eukaryotes. Biochim Biophys Acta. 2006;1763(7):652–67. [DOI] [PubMed] [Google Scholar]
- 32.Clark E, Johnson J, Dong Yi N, Mercado-Ayon E, Warren N, Zhai M, et al. Role of frataxin protein deficiency and metabolic dysfunction in Friedreich ataxia, an autosomal recessive mitochondrial disease. Neuronal Signaling. 2018;2(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Saha PP, Kumar SKP, Srivastava S, Sinha D, Pareek G, D’Silva P. The presence of multiple cellular defects associated with a novel G50E iron-sulfur cluster scaffold protein (ISCU) mutation leads to development of mitochondrial myopathy. J Biol Chem. 2014;289(15):10359–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kuhlbrandt W. Structure and function of mitochondrial membrane protein complexes. BMC Biol. 2015;13:89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Seguin A, Jia X, Earl AM, Li L, Wallace J, Qiu A, et al. The mitochondrial metal transporters mitoferrin1 and mitoferrin2 are required for liver regeneration and cell proliferation in mice. J Biol Chem. 2020;295(32):11002–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Shaw GC, Cope JJ, Li L, Corson K, Hersey C, Ackermann GE, et al. Mitoferrin is essential for erythroid iron assimilation. Nature. 2006;440(7080):96–100. [DOI] [PubMed] [Google Scholar]
- 37.Jain A, Dashner ZS, Connolly EL. Mitochondrial Iron Transporters (MIT1 and MIT2) Are Essential for Iron Homeostasis and Embryogenesis in Arabidopsis thaliana. Front Plant Sci. 2019;10:1449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Paradkar PN, Zumbrennen KB, Paw BH, Ward DM, Kaplan J. Regulation of mitochondrial iron import through differential turnover of mitoferrin 1 and mitoferrin 2. Mol Cell Biol. 2009;29(4):1007–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Li H, Zhao H, Hao S, Shang L, Wu J, Song C, et al. Iron regulatory protein deficiency compromises mitochondrial function in murine embryonic fibroblasts. Sci Rep. 2018;8(1):5118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Santos MM, Miranda CJ, Levy JE, Montross LK, Cossée M, Sequeiros J, et al. Iron metabolism in mice with partial frataxin deficiency. The Cerebellum. 2003;2(2):146–53. [DOI] [PubMed] [Google Scholar]
- 41.Cardenas-Rodriguez M, Chatzi A, Tokatlidis K. Iron-sulfur clusters: from metals through mitochondria biogenesis to disease. J Biol Inorg Chem. 2018;23(4):509–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ichikawa Y, Bayeva M, Ghanefar M, Potini V, Sun L, Mutharasan RK, et al. Disruption of ATP-binding cassette B8 in mice leads to cardiomyopathy through a decrease in mitochondrial iron export. Proc Natl Acad Sci U S A. 2012;109(11):4152–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Pondarre C, Antiochos BB, Campagna DR, Clarke SL, Greer EL, Deck KM, et al. The mitochondrial ATP-binding cassette transporter Abcb7 is essential in mice and participates in cytosolic iron-sulfur cluster biogenesis. Hum Mol Genet. 2006;15(6):953–64. [DOI] [PubMed] [Google Scholar]
- 44.Qi W, Li J, Cowan JA. A structural model for glutathione-complexed iron-sulfur cluster as a substrate for ABCB7-type transporters. Chem Commun (Camb). 2014;50(29):3795–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pearson SA, Wachnowsky C, Cowan JA. Defining the mechanism of the mitochondrial Atm1p [2Fe–2S] cluster exporter†. Metallomics. 2020;12(6):902–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zutz A, Gompf S, Schagger H, Tampe R. Mitochondrial ABC proteins in health and disease. Biochim Biophys Acta. 2009; 1787(6):681–90. [DOI] [PubMed] [Google Scholar]
- 47.Lozoff B, Jimenez E, Hagen J, Mollen E, Wolf AW. Poorer behavioral and developmental outcome more than 10 years after treatment for iron deficiency in infancy. Pediatrics. 2000; 105(4):E51. [DOI] [PubMed] [Google Scholar]
- 48.Kim J, Wessling-Resnick M. Iron and mechanisms of emotional behavior. J Nutr Biochem. 2014;25(11): 1101–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Erikson KM, Jones BC, Hess EJ, Zhang Q, Beard JL. Iron deficiency decreases dopamine D1 and D2 receptors in rat brain. Pharmacol Biochem Behav. 2001;69(3-4):409–18. [DOI] [PubMed] [Google Scholar]
- 50.Chen Q, Beard JL, Jones BC. Abnormal rat brain monoamine metabolism in iron deficiency anemia. The Journal of Nutritional Biochemistry. 1995;6(9):486–93. [Google Scholar]
- 51.Li Y, Kim J, Buckett PD, Bohlke M, Maher TJ, Wessling-Resnick M. Severe postnatal iron deficiency alters emotional behavior and dopamine levels in the prefrontal cortex of young male rats. J Nutr. 2011; 141 (12):2133–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Morse AC, Beard JL, Azar MR, Jones BC. Sex and Genetics are Important Cofactors in Assessing the Impact of Iron Deficiency on the Developing Mouse Brain. Nutr Neurosci. 1999;2(5):323–35. [DOI] [PubMed] [Google Scholar]
- 53.Burhans MS, Dailey C, Beard Z, Wiesinger J, Murray-Kolb L, Jones BC, et al. Iron deficiency: differential effects on monoamine transporters. Nutr Neurosci. 2005;8(1):31–8. [DOI] [PubMed] [Google Scholar]
- 54.Bianco LE, Wiesinger J, Earley CJ, Jones BC, Beard JL. Iron deficiency alters dopamine uptake and response to L-DOPA injection in Sprague-Dawley rats. J Neurochem. 2008;106(1):205–15. [DOI] [PubMed] [Google Scholar]
- 55.Ortiz E, Pasquini JM, Thompson K, Felt B, Butkus G, Beard J, et al. Effect of manipulation of iron storage, transport, or availability on myelin composition and brain iron content in three different animal models. J Neurosci Res. 2004;77(5):681–9. [DOI] [PubMed] [Google Scholar]
- 56.Oloyede OB, Folayan AT, Odutuga AA. Effects of low-iron status and deficiency of essential fatty acids on some biochemical constituents of rat brain. Biochem Int. 1992;27(5):913–22. [PubMed] [Google Scholar]
- 57.Moller HE, Bossoni L, Connor JR, Crichton RR, Does MD, Ward RJ, et al. Iron, Myelin, and the Brain: Neuroimaging Meets Neurobiology. Trends Neurosci. 2019;42(6):384–401. [DOI] [PubMed] [Google Scholar]
- 58.Zywicke HA, van Gelderen P, Connor JR, Burdo JR, Garrick MD, Dolan KG, et al. Microscopic R2* mapping of reduced brain iron in the Belgrade rat. Ann Neurol. 2002;52(1):102–5. [DOI] [PubMed] [Google Scholar]
- 59.Armony-Sivan R, Eidelman AI, Lanir A, Sredni D, Yehuda S. Iron Status and Neurobehavioral Development of Premature Infants. Journal of Perinatology. 2004;24(12):757–62. [DOI] [PubMed] [Google Scholar]
- 60.Algarin C, Peirano P, Garrido M, Pizarro F, Lozoff B. Iron deficiency anemia in infancy: long-lasting effects on auditory and visual system functioning. Pediatr Res. 2003;53(2):217–23. [DOI] [PubMed] [Google Scholar]
- 61.Salvador GA. Iron in neuronal function and dysfunction. Biofactors. 2010;36(2):103–10. [DOI] [PubMed] [Google Scholar]
- 62.Connor JR, Fine RE. Development of transferrin-positive oligodendrocytes in the rat central nervous system. J Neurosci Res. 1987;17(1):51–9. [DOI] [PubMed] [Google Scholar]
- 63.Connor JR, Menzies SL. Cellular management of iron in the brain. J Neurol Sci. 1995;134 Suppl:33–44. [DOI] [PubMed] [Google Scholar]
- 64.Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L. The role of iron in brain ageing and neurodegenerative disorders. Lancet Neurol. 2014;13(10):1045–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Leitner DF, Connor JR. Functional roles of transferrin in the brain. Biochim Biophys Acta. 2012;1820(3):393–402. [DOI] [PubMed] [Google Scholar]
- 66.LeVine SM, Macklin WB. Iron-enriched oligodendrocytes: a reexamination of their spatial distribution. J Neurosci Res. 1990;26(4):508–12. [DOI] [PubMed] [Google Scholar]
- 67.Todorich B, Zhang X, Connor JR. H-ferritin is the major source of iron for oligodendrocytes. Glia. 2011;59(6):927–35. [DOI] [PubMed] [Google Scholar]
- 68.Todorich B, Zhang X, Slagle-Webb B, Seaman WE, Connor JR. Tim-2 is the receptor for H-ferritin on oligodendrocytes. J Neurochem. 2008;107(6):1495–505. [DOI] [PubMed] [Google Scholar]
- 69.Calder GL, Lee MH, Sachithanandan N, Bell S, Zeimer H, MacIsaac RJ. Aceruloplasminaemia: a disorder of diabetes and neurodegeneration. Intern Med J. 2017;47(1):115–8. [DOI] [PubMed] [Google Scholar]
- 70.Xu H, Liu X, Xia J, Yu T, Qu Y, Jiang H, et al. Activation of NMDA receptors mediated iron accumulation via modulating iron transporters in Parkinson’s disease. FASEB J. 2018:fj201800060RR. [DOI] [PubMed] [Google Scholar]
- 71.Naigamwalla DZ, Webb JA, Giger U. Iron deficiency anemia. Can Vet J. 2012;53(3):250–6. [PMC free article] [PubMed] [Google Scholar]
- 72.Peng YY, Uprichard J. Ferritin and iron studies in anaemia and chronic disease. Ann Clin Biochem. 2017;54(1):43–8. [DOI] [PubMed] [Google Scholar]
- 73.D’Angelo G Role of hepcidin in the pathophysiology and diagnosis of anemia. Blood Res. 2013;48(1):10–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Vela D. Hepcidin, an emerging and important player in brain iron homeostasis. J Transl Med. 2018;16(1):25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Krause A, Neitz S, Magert HJ, Schulz A, Forssmann WG, Schulz-Knappe P, et al. LEAP-1, a novel highly disulfide-bonded human peptide, exhibits antimicrobial activity. FEBS Lett. 2000;480(2–3):147–50. [DOI] [PubMed] [Google Scholar]
- 76.Xiong XY, Liu L, Wang FX, Yang YR, Hao JW, Wang PF, et al. Toll-Like Receptor 4/MyD88-Mediated Signaling of Hepcidin Expression Causing Brain Iron Accumulation, Oxidative Injury, and Cognitive Impairment After Intracerebral Hemorrhage. Circulation. 2016;134(14):1025–38. [DOI] [PubMed] [Google Scholar]
- 77.Raha-Chowdhury R, Raha AA, Forostyak S, Zhao JW, Stott SR, Bomford A. Expression and cellular localization of hepcidin mRNA and protein in normal rat brain. BMC Neurosci. 2015;16:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hofer T, Perry G. Nucleic acid oxidative damage in Alzheimer’s disease-explained by the hepcidin-ferroportin neuronal iron overload hypothesis? J Trace Elem Med Biol. 2016;38:1–9. [DOI] [PubMed] [Google Scholar]
- 79.Du F, Qian C, Qian ZM, Wu XM, Xie H, Yung WH, et al. Hepcidin directly inhibits transferrin receptor 1 expression in astrocytes via a cyclic AMP-protein kinase A pathway. Glia. 2011;59(6):936–45. [DOI] [PubMed] [Google Scholar]
- 80.Du F, Qian ZM, Luo Q, Yung WH, Ke Y. Hepcidin Suppresses Brain Iron Accumulation by Downregulating Iron Transport Proteins in Iron-Overloaded Rats. Mol Neurobiol. 2015;52(1):101–14. [DOI] [PubMed] [Google Scholar]
- 81.Zhao H, Lewellen BM, Wilson RJ, Cui D, Drake JC, Zhang M, et al. Long-term voluntary running prevents the onset of symptomatic Friedreich’s ataxia in mice. Sci Rep. 2020;10(1):6095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Chen D, Kanthasamy AG, Reddy MB. EGCG Protects against 6-OHDA-Induced Neurotoxicity in a Cell Culture Model. Parkinsons Dis. 2015;2015:843906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Jiang H, Song N, Xu H, Zhang S, Wang J, Xie J. Up-regulation of divalent metal transporter 1 in 6-hydroxydopamine intoxication is IRE/IRP dependent. Cell Res. 2010;20(3):345–56. [DOI] [PubMed] [Google Scholar]
- 84.Xu Q, Kanthasamy AG, Jin H, Reddy MB. Hepcidin Plays a Key Role in 6-OHDA Induced Iron Overload and Apoptotic Cell Death in a Cell Culture Model of Parkinson’s Disease. Parkinsons Dis. 2016;2016:8684130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Fleming RE, Ponka P. Iron overload in human disease. N Engl J Med. 2012;366(4):348–59. [DOI] [PubMed] [Google Scholar]
- 86.Gao G, Li J, Zhang Y, Chang YZ. Cellular Iron Metabolism and Regulation. Adv Exp Med Biol. 2019;1173:21–32. [DOI] [PubMed] [Google Scholar]
- 87.Halliwell B Reactive oxygen species and the central nervous system. J Neurochem. 1992;59(5):1609–23. [DOI] [PubMed] [Google Scholar]
- 88.Anderson GJ. Mechanisms of iron loading and toxicity. Am J Hematol. 2007;82(12 Suppl):1128–31. [DOI] [PubMed] [Google Scholar]
- 89.Huang XT, Liu X, Ye CY, Tao LX, Zhou H, Zhang HY. Iron-induced energy supply deficiency and mitochondrial fragmentation in neurons. J Neurochem. 2018;147(6):816–30. [DOI] [PubMed] [Google Scholar]
- 90.Jarvis JH, Jacobs A. Morphological abnormalities in lymphocyte mitochondria associated with iron-deficiency anaemia. J Clin Pathol. 1974;27(12):973–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Walter PB, Knutson MD, Paler-Martinez A, Lee S, Xu Y, Viteri FE, et al. Iron deficiency and iron excess damage mitochondria and mitochondrial DNA in rats. Proceedings of the National Academy of Sciences. 2002;99(4):2264–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Knott AB, Perkins G, Schwarzenbacher R, Bossy-Wetzel E. Mitochondrial fragmentation in neurodegeneration. Nat Rev Neurosci. 2008;9(7):505–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Bertholet AM, Delerue T, Millet AM, Moulis MF, David C, Daloyau M, et al. Mitochondrial fusion/fission dynamics in neurodegeneration and neuronal plasticity. Neurobiol Dis. 2016;90:3–19. [DOI] [PubMed] [Google Scholar]
- 94.Frank S Dysregulation of mitochondrial fusion and fission: an emerging concept in neurodegeneration. Acta Neuropathologica. 2006;111(2):93–100. [DOI] [PubMed] [Google Scholar]
- 95.Smirnova E, Griparic L, Shurland D-L, Bliek AMvd. Dynamin-related Protein Drp1 Is Required for Mitochondrial Division in Mammalian Cells. Molecular Biology of the Cell. 2001;12(8):2245–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Hoppins S, Lackner L, Nunnari J. The Machines that Divide and Fuse Mitochondria. Annual Review of Biochemistry. 2007;76(1):751–80. [DOI] [PubMed] [Google Scholar]
- 97.Bastian TW, von Hohenberg WC, Georgieff MK, Lanier LM. Chronic Energy Depletion due to Iron Deficiency Impairs Dendritic Mitochondrial Motility during Hippocampal Neuron Development. J Neurosci. 2019;39(5):802–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Couch VA, Medvedev ES, Stuchebrukhov AA. Electrostatics of the FeS clusters in respiratory complex I. Biochim Biophys Acta. 2009;1787(10):1266–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wiegersma AM, Dalman C, Lee BK, Karlsson H, Gardner RM. Association of Prenatal Maternal Anemia With Neurodevelopmental Disorders. JAMA Psychiatry. 2019;76(12):1294–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Yang W, Liu B, Gao R, Snetselaar LG, Strathearn L, Bao W. Association of Anemia with Neurodevelopmental Disorders in a Nationally Representative Sample of US Children. The Journal of Pediatrics. 2021;228:183–9.e2. [DOI] [PubMed] [Google Scholar]
- 101.Sipe JC, Lee P, Beutler E. Brain iron metabolism and neurodegenerative disorders. Dev Neurosci. 2002;24(2–3):188–96. [DOI] [PubMed] [Google Scholar]
- 102.Urrutia P, Mena N, Nunez M. The interplay between iron accumulation, mitochondrial dysfunction, and inflammation during the execution step of neurodegenerative disorders. Frontiers in Pharmacology. 2014;5(38). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ward RJ, Zucca FA, Duyn JH, Crichton RR, Zecca L. The role of iron in brain ageing and neurodegenerative disorders. The Lancet Neurology. 2014;13(10):1045–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Joshi AU, Saw NL, Shamloo M, Mochly-Rosen D. Drp1/Fis1 interaction mediates mitochondrial dysfunction, bioenergetic failure and cognitive decline in Alzheimer’s disease. Oncotarget. 2018;9(5):6128–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Park J, Seo J, Won J, Yeo HG, Ahn YJ, Kim K, et al. Abnormal Mitochondria in a Non-human Primate Model of MPTP-induced Parkinson’s Disease: Drp1 and CDK5/p25 Signaling. Exp Neurobiol. 2019;28(3):414–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Rocha AG, Franco A, Krezel AM, Rumsey JM, Alberti JM, Knight WC, et al. MFN2 agonists reverse mitochondrial defects in preclinical models of Charcot-Marie-Tooth disease type 2A. Science. 2018;360(6386):336–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Park J, Lee DG, Kim B, Park S-J, Kim J-H, Lee S-R, et al. Iron overload triggers mitochondrial fragmentation via calcineurin-sensitive signals in HT-22 hippocampal neuron cells. Toxicology. 2015;337:39–46. [DOI] [PubMed] [Google Scholar]
- 108.Lee DG, Kam MK, Lee SR, Lee HJ, Lee DS. Peroxiredoxin 5 deficiency exacerbates iron overload-induced neuronal death via ER-mediated mitochondrial fission in mouse hippocampus. Cell Death Dis. 2020;11(3):204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Lee DG, Kam MK, Kim KM, Kim HS, Kwon O-S, Lee H-S, et al. Peroxiredoxin 5 prevents iron overload-induced neuronal death by inhibiting mitochondrial fragmentation and endoplasmic reticulum stress in mouse hippocampal HT-22 cells. The International Journal of Biochemistry & Cell Biology. 2018;102:10–9. [DOI] [PubMed] [Google Scholar]
- 110.Kim MH, Lee HJ, Lee S-R, Lee H-S, Huh J-W, Bae YC, et al. Peroxiredoxin 5 Inhibits Glutamate-Induced Neuronal Cell Death through the Regulation of Calcineurin-Dependent Mitochondrial Dynamics in HT22 Cells. Molecular and Cellular Biology. 2019;39(20):e00148–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lee DG, Park J, Lee H-S, Lee S-R, Lee D-S. Iron overload-induced calcium signals modulate mitochondrial fragmentation in HT-22 hippocampal neuron cells. Toxicology. 2016;365:17–24. [DOI] [PubMed] [Google Scholar]
- 112.Masaldan S, Bush AI, Devos D, Rolland AS, Moreau C. Striking while the iron is hot: Iron metabolism and ferroptosis in neurodegeneration. Free Radic Biol Med. 2019;133:221–33. [DOI] [PubMed] [Google Scholar]
- 113.Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol. 2014;16(12):1180–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Hider RC, Kong XL. Glutathione: a key component of the cytoplasmic labile iron pool. Biometals. 2011;24(6):1179–87. [DOI] [PubMed] [Google Scholar]
- 116.Connor JR, Snyder BS, Arosio P, Loeffler DA, LeWitt P. A quantitative analysis of isoferritins in select regions of aged, parkinsonian, and Alzheimer’s diseased brains. J Neurochem. 1995;65(2):717–24. [DOI] [PubMed] [Google Scholar]
- 117.Hirose W, Ikematsu K, Tsuda R. Age-associated increases in heme oxygenase-1 and ferritin immunoreactivity in the autopsied brain. Leg Med (Tokyo). 2003;5 Suppl 1 :S360–6. [DOI] [PubMed] [Google Scholar]
- 118.Koeppen AH. Friedreich’s ataxia: pathology, pathogenesis, and molecular genetics. J Neurol Sci. 2011. ;303(1-2): 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Campuzano V, Montermini L, Molto MD, Pianese L, Cossee M, Cavalcanti F, et al. Friedreich’s ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion. Science. 1996;271 (5254): 1423–7. [DOI] [PubMed] [Google Scholar]
- 120.Evans-Galea MV, Carrodus N, Rowley SM, Corben LA, Tai G, Saffery R, et al. FXN methylation predicts expression and clinical outcome in Friedreich ataxia. Ann Neurol. 2012;71(4):487–97. [DOI] [PubMed] [Google Scholar]
- 121.Gellera C, Castellotti B, Mariotti C, Mineri R, Seveso V, Didonato S, et al. Frataxin gene point mutations in Italian Friedreich ataxia patients. Neurogenetics. 2007;8(4):289–99. [DOI] [PubMed] [Google Scholar]
- 122.Heidari MM, Khatami M, Pourakrami J. Novel Point Mutations in Frataxin Gene in Iranian Patients with Friedreich’s Ataxia. Iran J Child Neurol. 2014;8(1):32–6. [PMC free article] [PubMed] [Google Scholar]
- 123.Galea CA, Huq A, Lockhart PJ, Tai G, Corben LA, Yiu EM, et al. Compound heterozygous FXN mutations and clinical outcome in friedreich ataxia. Ann Neurol. 2016;79(3):485–95. [DOI] [PubMed] [Google Scholar]
- 124.Guo L, Wang Q, Weng L, Hauser LA, Strawser CJ, Mesaros C, et al. Characterization of a new N-terminally acetylated extra-mitochondrial isoform of frataxin in human erythrocytes. Sci Rep. 2018;8(1): 17043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Martelli A, Wattenhofer-Donzé M, Schmucker S, Bouvet S, Reutenauer L, Puccio H. Frataxin is essential for extramitochondrial Fe-S cluster proteins in mammalian tissues. Human Molecular Genetics. 2007;16(22):2651–8. [DOI] [PubMed] [Google Scholar]
- 126.Fox NG, Yu X, Feng X, Bailey HJ, Martelli A, Nabhan JF, et al. Structure of the human frataxin-bound iron-sulfur cluster assembly complex provides insight into its activation mechanism. Nat Commun. 2019;10(1):2210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Das D, Patra S, Bridwell-Rabb J, Barondeau DP. Mechanism of frataxin “bypass’ in human iron-sulfur cluster biosynthesis with implications for Friedreich’s ataxia. J Biol Chem. 2019;294(23):9276–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Khodour Y, Kaguni LS, Stiban J. Chapter Seven - Iron–sulfur clusters in nucleic acid metabolism: Varying roles of ancient cofactors. In: Zhao L, Kaguni LS, editors. The Enzymes. 45: Academic Press; 2019. p. 225–56. [DOI] [PubMed] [Google Scholar]
- 129.Melber A, Winge DR. Steps Toward Understanding Mitochondrial Fe/S Cluster Biogenesis. Methods Enzymol. 2018;599:265–92. [DOI] [PubMed] [Google Scholar]
- 130.Moroishi T, Nishiyama M, Takeda Y, Iwai K, Nakayama KI. The FBXL5-IRP2 axis is integral to control of iron metabolism in vivo. Cell Metab. 2011. ;14(3):339–51. [DOI] [PubMed] [Google Scholar]
- 131.Vashisht AA, Zumbrennen KB, Huang X, Powers DN, Durazo A, Sun D, et al. Control of iron homeostasis by an iron-regulated ubiquitin ligase. Science. 2009;326(5953):718–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Schulz JB, Dehmer T, Schöls L, Mende H, Hardt C, Vorgerd M, et al. Oxidative stress in patients with Friedreich ataxia. Neurology. 2000;55(11):1719–21. [DOI] [PubMed] [Google Scholar]
- 133.Armstrong JS, Khdour O, Hecht SM. Does oxidative stress contribute to the pathology of Friedreich’s ataxia? A radical question. FASEB J. 2010;24(7):2152–63. [DOI] [PubMed] [Google Scholar]
- 134.Tan G, Napoli E, Taroni F, Cortopassi G. Decreased expression of genes involved in sulfur amino acid metabolism in frataxin-deficient cells. Hum Mol Genet. 2003; 12(14): 1699–711. [DOI] [PubMed] [Google Scholar]
- 135.Shan Y, Schoenfeld RA, Hayashi G, Napoli E, Akiyama T, Iodi Carstens M, et al. Frataxin deficiency leads to defects in expression of antioxidants and Nrf2 expression in dorsal root ganglia of the Friedreich’s ataxia YG8R mouse model. Antioxid Redox Signal. 2013;19(13):1481–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Schmucker S, Martelli A, Colin F, Page A, Wattenhofer-Donzé M, Reutenauer L, et al. Mammalian Frataxin: An Essential Function for Cellular Viability through an Interaction with a Preformed ISCU/NFS1/ISD11 Iron-Sulfur Assembly Complex. PLOS ONE. 2011;6(1):e16199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Mochel F, Knight MA, Tong W-H, Hernandez D, Ayyad K, Taivassalo T, et al. Splice Mutation in the Iron-Sulfur Cluster Scaffold Protein ISCU Causes Myopathy with Exercise Intolerance. The American Journal of Human Genetics. 2008;82(3):652–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.La Rosa P, Petrillo S, Turchi R, Berardinelli F, Schirinzi T, Vasco G, et al. The Nrf2 induction prevents ferroptosis in Friedreich’s Ataxia. Redox Biol. 2021;38:101791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.La Rosa P, Petrillo S, Fiorenza MT, Bertini ES, Piemonte F. Ferroptosis in Friedreich’s Ataxia: A Metal-Induced Neurodegenerative Disease. Biomolecules. 2020; 10(11): 1551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Ayala A, Munoz MF, Arguelles S. Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. 2014;2014:360438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Chang L-C, Chiang S-K, Chen S-E, Yu Y-L, Chou R-H, Chang W-C. Heme oxygenase-1 mediates BAY 11–7085 induced ferroptosis. Cancer Letters. 2018;416:124–37. [DOI] [PubMed] [Google Scholar]
- 142.#039, Oria V, Petrini S, Travaglini L, Priori C, Piermarini E, et al. Frataxin Deficiency Leads to Reduced Expression and Impaired Translocation of NF-E2-Related Factor (Nrf2) in Cultured Motor Neurons. International Journal of Molecular Sciences. 2013; 14(4):7853–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Petrillo S, Piermarini E, Pastore A, Vasco G, Schirinzi T, Carrozzo R, et al. Nrf2-Inducers Counteract Neurodegeneration in Frataxin-Silenced Motor Neurons: Disclosing New Therapeutic Targets for Friedreich’s Ataxia. Int J Mol Sci. 2017; 18(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Chiang S, Braidy N, Maleki S, Lai S, Richardson DR, Huang ML. Mechanisms of impaired mitochondrial homeostasis and NAD(+) metabolism in a model of mitochondrial heart disease exhibiting redox active iron accumulation. Redox Biol. 2021. ;46:102038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Tai G, Corben LA, Yiu EM, Milne SC, Delatycki MB. Progress in the treatment of Friedreich ataxia. Neurologia i Neurochirurgia Polska. 2018;52(2): 129–39. [DOI] [PubMed] [Google Scholar]
- 146.Johnson J, Mercado-Ayon E, Clark E, Lynch D, Lin H. Drp1-dependent peptide reverse mitochondrial fragmentation, a homeostatic response in Friedreich ataxia. Pharmacol Res Perspect. 2021. ;9(3):e00755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443(7113):787–95. [DOI] [PubMed] [Google Scholar]
- 148.Bates GP, Dorsey R, Gusella JF, Hayden MR, Kay C, Leavitt BR, et al. Huntington disease. Nature Reviews Disease Primers. 2015;1(1). [DOI] [PubMed] [Google Scholar]
- 149.Agrawal S, Fox J, Thyagarajan B, Fox JH. Brain mitochondrial iron accumulates in Huntington’s disease, mediates mitochondrial dysfunction, and can be removed pharmacologically. Free Radic Biol Med. 2018;120:317–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Djousse L, Knowlton B, Cupples LA, Marder K, Shoulson I, Myers RH. Weight loss in early stage of Huntington’s disease. Neurology. 2002;59(9):1325–30. [DOI] [PubMed] [Google Scholar]
- 151.Mena NP, Urrutia PJ, Lourido F, Carrasco CM, Nunez MT. Mitochondrial iron homeostasis and its dysfunctions in neurodegenerative disorders. Mitochondrion. 2015;21:92–105. [DOI] [PubMed] [Google Scholar]
- 152.Muller M, Leavitt BR. Iron dysregulation in Huntington’s disease. Journal of Neurochemistry. 2014;130(3):328–50. [DOI] [PubMed] [Google Scholar]
- 153.Bartzokis G, Cummings J, Perlman S, Hance DB, Mintz J. Increased basal ganglia iron levels in Huntington disease. Arch Neurol. 1999;56(5):569–74. [DOI] [PubMed] [Google Scholar]
- 154.Guidetti P, Charles V, Chen EY, Reddy PH, Kordower JH, Whetsell WO Jr., et al. Early degenerative changes in transgenic mice expressing mutant huntingtin involve dendritic abnormalities but no impairment of mitochondrial energy production. Exp Neurol. 2001;169(2):340–50. [DOI] [PubMed] [Google Scholar]
- 155.Oliveira JM. Nature and cause of mitochondrial dysfunction in Huntington’s disease: focusing on huntingtin and the striatum. J Neurochem. 2010;114(1):1–12. [DOI] [PubMed] [Google Scholar]
- 156.Lumsden AL, Henshall TL, Dayan S, Lardelli MT, Richards RI. Huntingtin-deficient zebrafish exhibit defects in iron utilization and development. Hum Mol Genet. 2007. ;16(16):1905–20. [DOI] [PubMed] [Google Scholar]
- 157.Brennan WA Jr., Bird ED, Aprille JR. Regional mitochondrial respiratory activity in Huntington’s disease brain. J Neurochem. 1985;44(6):1948–50. [DOI] [PubMed] [Google Scholar]
- 158.Benchoua A, Trioulier Y, Zala D, Gaillard MC, Lefort N, Dufour N, et al. Involvement of mitochondrial complex II defects in neuronal death produced by N-terminus fragment of mutated huntingtin. Mol Biol Cell. 2006;17(4):1652–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Tabrizi SJ, Cleeter MW, Xuereb J, Taanman JW, Cooper JM, Schapira AH. Biochemical abnormalities and excitotoxicity in Huntington’s disease brain. Ann Neurol. 1999;45(1):25–32. [DOI] [PubMed] [Google Scholar]
- 160.Milakovic T, Johnson GV. Mitochondrial respiration and ATP production are significantly impaired in striatal cells expressing mutant huntingtin. J Biol Chem. 2005;280(35):30773–82. [DOI] [PubMed] [Google Scholar]
- 161.Cui L, Jeong H, Borovecki F, Parkhurst CN, Tanese N, Krainc D. Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell. 2006;127(1):59–69. [DOI] [PubMed] [Google Scholar]
- 162.Hodges A, Strand AD, Aragaki AK, Kuhn A, Sengstag T, Hughes G, et al. Regional and cellular gene expression changes in human Huntington’s disease brain. Hum Mol Genet. 2006;15(6):965–77. [DOI] [PubMed] [Google Scholar]
- 163.Luthi-Carter R, Strand A, Peters NL, Solano SM, Hollingsworth ZR, Menon AS, et al. Decreased expression of striatal signaling genes in a mouse model of Huntington’s disease. Hum Mol Genet. 2000;9(9):1259–71. [DOI] [PubMed] [Google Scholar]
- 164.Shirendeb UP, Calkins MJ, Manczak M, Anekonda V, Dufour B, McBride JL, et al. Mutant huntingtin’s interaction with mitochondrial protein Drp1 impairs mitochondrial biogenesis and causes defective axonal transport and synaptic degeneration in Huntington’s disease. Hum Mol Genet. 2012;21(2):406–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Song W, Chen J, Petrilli A, Liot G, Klinglmayr E, Zhou Y, et al. Mutant huntingtin binds the mitochondrial fission GTPase dynamin-related protein-1 and increases its enzymatic activity. Nat Med. 2011;17(3):377–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Johri A, Chaturvedi RK, Beal MF. Hugging tight in Huntington’s. Nat Med. 2011;17(3):245–6. [DOI] [PubMed] [Google Scholar]
- 167.Dominguez JF, Ng AC, Poudel G, Stout JC, Churchyard A, Chua P, et al. Iron accumulation in the basal ganglia in Huntington’s disease: cross-sectional data from the IMAGE-HD study. J Neurol Neurosurg Psychiatry. 2016;87(5):545–9. [DOI] [PubMed] [Google Scholar]
- 168.van Bergen JM, Hua J, Unschuld PG, Lim IA, Jones CK, Margolis RL, et al. Quantitative Susceptibility Mapping Suggests Altered Brain Iron in Premanifest Huntington Disease. AJNR Am J Neuroradiol. 2016;37(5):789–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Dexter DT, Jenner P, Schapira AH, Marsden CD. Alterations in levels of iron, ferritin, and other trace metals in neurodegenerative diseases affecting the basal ganglia. The Royal Kings and Queens Parkinson’s Disease Research Group. Ann Neurol. 1992;32 Suppl:S94–100. [DOI] [PubMed] [Google Scholar]
- 170.Simmons DA, Casale M, Alcon B, Pham N, Narayan N, Lynch G. Ferritin accumulation in dystrophic microglia is an early event in the development of Huntington’s disease. Glia. 2007;55(10):1074–84. [DOI] [PubMed] [Google Scholar]
- 171.Sapp E, Kegel KB, Aronin N, Hashikawa T, Uchiyama Y, Tohyama K, et al. Early and progressive accumulation of reactive microglia in the Huntington disease brain. J Neuropathol Exp Neurol. 2001;60(2):161–72. [DOI] [PubMed] [Google Scholar]
- 172.Chen J, Marks E, Lai B, Zhang Z, Duce JA, Lam LQ, et al. Iron accumulates in Huntington’s disease neurons: protection by deferoxamine. PLoS One. 2013;8(10):e77023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Niu L, Ye C, Sun Y, Peng T, Yang S, Wang W, et al. Mutant huntingtin induces iron overload via up-regulating IRP1 in Huntington’s disease. Cell Biosci. 2018;8:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Firdaus WJ, Wyttenbach A, Giuliano P, Kretz-Remy C, Currie RW, Arrigo AP. Huntingtin inclusion bodies are iron-dependent centers of oxidative events. FEBS J. 2006;273(23):5428–41. [DOI] [PubMed] [Google Scholar]
- 175.Nguyen T, Hamby A, Massa SM. Clioquinol down-regulates mutant huntingtin expression in vitro and mitigates pathology in a Huntington’s disease mouse model. Proc Natl Acad Sci U S A. 2005;102(33):11840–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Narayanan M, Huynh JL, Wang K, Yang X, Yoo S, McElwee J, et al. Common dysregulation network in the human prefrontal cortex underlies two neurodegenerative diseases. Mol Syst Biol. 2014;10:743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Bowling AC, Beal MF. Bioenergetic and oxidative stress in neurodegenerative diseases. Life Sci. 1995;56(14):1151–71. [DOI] [PubMed] [Google Scholar]
- 178.Roze E, Saudou F, Caboche J. Pathophysiology of Huntington’s disease: from huntingtin functions to potential treatments. Curr Opin Neurol. 2008;21(4):497–503. [DOI] [PubMed] [Google Scholar]
- 179.Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med. 2010;362(4):329–44. [DOI] [PubMed] [Google Scholar]
- 180.Mandel S, Amit T, Bar-Am O, Youdim MB. Iron dysregulation in Alzheimer’s disease: multimodal brain permeable iron chelating drugs, possessing neuroprotective-neurorescue and amyloid precursor protein-processing regulatory activities as therapeutic agents. Prog Neurobiol. 2007;82(6):348–60. [DOI] [PubMed] [Google Scholar]
- 181.Nakamura M, Shishido N, Nunomura A, Smith MA, Perry G, Hayashi Y, et al. Three histidine residues of amyloid-beta peptide control the redox activity of copper and iron. Biochemistry. 2007;46(44):12737–43. [DOI] [PubMed] [Google Scholar]
- 182.Rottkamp CA, Raina AK, Zhu X, Gaier E, Bush AI, Atwood CS, et al. Redox-active iron mediates amyloid-beta toxicity. Free Radic Biol Med. 2001;30(4):447–50. [DOI] [PubMed] [Google Scholar]
- 183.Zecca L, Youdim MB, Riederer P, Connor JR, Crichton RR. Iron, brain ageing and neurodegenerative disorders. Nat Rev Neurosci. 2004;5(11):863–73. [DOI] [PubMed] [Google Scholar]
- 184.Rogers JT, Randall JD, Cahill CM, Eder PS, Huang X, Gunshin H, et al. An iron-responsive element type II in the 5’-untranslated region of the Alzheimer’s amyloid precursor protein transcript. J Biol Chem. 2002;277(47):45518–28. [DOI] [PubMed] [Google Scholar]
- 185.Cho HH, Cahill CM, Vanderburg CR, Scherzer CR, Wang B, Huang X, et al. Selective translational control of the Alzheimer amyloid precursor protein transcript by iron regulatory protein-1. J Biol Chem. 2010;285(41):31217–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Rogers JT, Bush AI, Cho HH, Smith DH, Thomson AM, Friedlich AL, et al. Iron and the translation of the amyloid precursor protein (APP) and ferritin mRNAs: riboregulation against neural oxidative damage in Alzheimer’s disease. Biochem Soc Trans. 2008;36(Pt 6):1282–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Avramovich-Tirosh Y, Amit T, Bar-Am O, Weinreb O, Youdim MB. Physiological and pathological aspects of Abeta in iron homeostasis via 5’UTR in the APP mRNA and the therapeutic use of iron-chelators. BMC Neurosci. 2008;9 Suppl 2:S2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Perry G, Sayre LM, Atwood CS, Castellani RJ, Cash AD, Rottkamp CA, et al. The role of iron and copper in the aetiology of neurodegenerative disorders: therapeutic implications. CNS Drugs. 2002;16(5):339–52. [DOI] [PubMed] [Google Scholar]
- 189.Yamamoto A, Shin RW, Hasegawa K, Naiki H, Sato H, Yoshimasu F, et al. Iron (III) induces aggregation of hyperphosphorylated tau and its reduction to iron (II) reverses the aggregation: implications in the formation of neurofibrillary tangles of Alzheimer’s disease. J Neurochem. 2002;82(5):1137–47. [DOI] [PubMed] [Google Scholar]
- 190.Lei P, Ayton S, Finkelstein DI, Spoerri L, Ciccotosto GD, Wright DK, et al. Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export. Nat Med. 2012;18(2):291–5. [DOI] [PubMed] [Google Scholar]
- 191.Guo C, Wang P, Zhong ML, Wang T, Huang XS, Li JY, et al. Deferoxamine inhibits iron induced hippocampal tau phosphorylation in the Alzheimer transgenic mouse brain. Neurochem Int. 2013;62(2):165–72. [DOI] [PubMed] [Google Scholar]
- 192.Wang T, Xu SF, Fan YG, Li LB, Guo C. Iron Pathophysiology in Alzheimer’s Diseases. Adv Exp Med Biol. 2019;1173:67–104. [DOI] [PubMed] [Google Scholar]
- 193.Lane DJR, Ayton S, Bush AI. Iron and Alzheimer’s Disease: An Update on Emerging Mechanisms. J Alzheimers Dis. 2018;64(s1):S379–S95. [DOI] [PubMed] [Google Scholar]
- 194.Wong BX, Tsatsanis A, Lim LQ, Adlard PA, Bush AI, Duce JA. beta-Amyloid precursor protein does not possess ferroxidase activity but does stabilize the cell surface ferrous iron exporter ferroportin. PLoS One. 2014;9(12):e114174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Ritchie CW, Bush AI, Mackinnon A, Macfarlane S, Mastwyk M, MacGregor L, et al. Metal-protein attenuation with iodochlorhydroxyquin (clioquinol) targeting Abeta amyloid deposition and toxicity in Alzheimer disease: a pilot phase 2 clinical trial. Arch Neurol. 2003;60(12):1685–91. [DOI] [PubMed] [Google Scholar]
- 196.Lannfelt L, Blennow K, Zetterberg H, Batsman S, Ames D, Harrison J, et al. Safety, efficacy, and biomarker findings of PBT2 in targeting Abeta as a modifying therapy for Alzheimer’s disease: a phase IIa, double-blind, randomised, placebo-controlled trial. Lancet Neurol. 2008;7(9):779–86. [DOI] [PubMed] [Google Scholar]
- 197.Mouton-Liger F, Paquet C, Dumurgier J, Bouras C, Pradier L, Gray F, et al. Oxidative stress increases BACE1 protein levels through activation of the PKR-eIF2alpha pathway. Biochim Biophys Acta. 2012;1822(6):885–96. [DOI] [PubMed] [Google Scholar]
- 198.Pratico D, Uryu K, Leight S, Trojanoswki JQ, Lee VM. Increased lipid peroxidation precedes amyloid plaque formation in an animal model of Alzheimer amyloidosis. J Neurosci. 2001;21(12):4183–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Horowitz MP, Greenamyre JT. Mitochondrial iron metabolism and its role in neurodegeneration. J Alzheimers Dis. 2010;20 Suppl 2:S551–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Bartzokis G, Cummings JL, Markham CH, Marmarelis PZ, Treciokas LJ, Tishler TA, et al. MRI evaluation of brain iron in earlier- and later-onset Parkinson’s disease and normal subjects. Magnetic Resonance Imaging. 1999; 17(2):213–22. [DOI] [PubMed] [Google Scholar]
- 201.Ayton S, Lei P, Duce JA, Wong BXW, Sedjahtera A, Adlard PA, et al. Ceruloplasmin dysfunction and therapeutic potential for Parkinson disease. Annals of Neurology. 2013;73(4):554–9. [DOI] [PubMed] [Google Scholar]
- 202.Jiang H, Wang J, Rogers J, Xie J. Brain Iron Metabolism Dysfunction in Parkinson’s Disease. Mol Neurobiol. 2017;54(4):3078–101. [DOI] [PubMed] [Google Scholar]
- 203.Jiang H, Song N, Jiao Q, Shi L, Du X. Iron Pathophysiology in Parkinson Diseases. Adv Exp Med Biol. 2019;1173:45–66. [DOI] [PubMed] [Google Scholar]
- 204.Jellinger K, Paulus W, Grundke-lqbal I, Riederer P, Youdim MB. Brain iron and ferritin in Parkinson’s and Alzheimer’s diseases. J Neural Transm Park Dis Dement Sect. 1990;2(4):327–40. [DOI] [PubMed] [Google Scholar]
- 205.Schipper HM, Liberman A, Stopa EG. Neural heme oxygenase-1 expression in idiopathic Parkinson’s disease. Exp Neurol. 1998; 150(1 ):60–8. [DOI] [PubMed] [Google Scholar]
- 206.Heikkila R, Hess A, Duvoisin R. Dopaminergic neurotoxicity of 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine in mice. Science. 1984;224(4656):1451–3. [DOI] [PubMed] [Google Scholar]
- 207.Nicklas WJ, Vyas I, Heikkila RE. Inhibition of NADH-linked oxidation in brain mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite of the neurotoxin, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. Life Sciences. 1985;36(26):2503–8. [DOI] [PubMed] [Google Scholar]
- 208.Bindoff LA, Birch-Machin M, Cartlidge NEF, Parker WD, Turnbull DM. Mitochondrial Function in Parkinson’s Disease. The Lancet. 1989;334(8653). [DOI] [PubMed] [Google Scholar]
- 209.Mizuno Y, Ohta S, Tanaka M, Takamiya S, Suzuki K, Sato T, et al. Deficiencies in complex I subunits of the respiratory chain in Parkinson’s disease. Biochem Biophys Res Commun. 1989; 163(3): 1450–5. [DOI] [PubMed] [Google Scholar]
- 210.Urrutia PJ, Mena NP, Nðñez MT. The interplay between iron accumulation, mitochondrial dysfunction, and inflammation during the execution step of neurodegenerative disorders. Frontiers in Pharmacology. 2014;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Devos D, Moreau C, Devedjian JC, Kluza J, Petrault M, Laloux C, et al. Targeting chelatable iron as a therapeutic modality in Parkinson’s disease. Antioxid Redox Signal. 2014;21 (2): 195–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Borie C, Gasparini F, Verpillat P, Bonnet A-M, Agid Y, Hetet G, et al. Association study between iron-related genes polymorphisms and Parkinson’s disease. Journal of Neurology. 2002;249(7):801–4. [DOI] [PubMed] [Google Scholar]
- 213.Deplazes J, Spiegel J, Becker G, Riess O, Berg D, Schöbel K, et al. Screening for mutations of the IRP2 gene in Parkinson?s disease patients with hyperechogenicity of the substantia nigra. Journal of Neural Transmission. 2004; 111 (4):515–21. [DOI] [PubMed] [Google Scholar]
- 214.Foglieni B, Ferrari F, Goldwurm S, Santambrogio P, Castiglioni E, Sessa M, et al. Analysis of ferritin genes in Parkinson disease. Clinical Chemical Laboratory Medicine. 2007;45(11). [DOI] [PubMed] [Google Scholar]
- 215.He Q, Du T, Yu X, Xie A, Song N, Kang Q, et al. DMT1 polymorphism and risk of Parkinson’s disease. Neuroscience Letters. 2011;501 (3): 128–31. [DOI] [PubMed] [Google Scholar]
- 216.Dexter DT, Wells FR, Lee AJ, Agid F, Agid Y, Jenner P, et al. Increased Nigral Iron Content and Alterations in Other Metal Ions Occurring in Brain in Parkinson’s Disease. Journal of Neurochemistry. 1989;52(6): 1830–6. [DOI] [PubMed] [Google Scholar]
- 217.Salazar J, Mena N, Hunot S, Prigent A, Alvarez-Fischer D, Arredondo M, et al. Divalent metal transporter 1 (DMT1) contributes to neurodegeneration in animal models of Parkinson’s disease. Proc Natl Acad Sci USA. 2008; 105(47): 18578–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.NBIA DISORDERS [Available from: http://nbiacure.org/learn/nbia-disorders/.
- 219.Overview of NBIA Disorders NBIA Disorders Association [Available from: https://www.nbiadisorders.org/about-nbia/overview-of-nbia-disorders.
- 220.Levi S, Cozzi A, Santambrogio P. Iron Pathophysiology in Neurodegeneration with Brain Iron Accumulation. Adv Exp Med Biol. 2019; 1173:153–77. [DOI] [PubMed] [Google Scholar]
- 221.Jeong SY, David S. Glycosylphosphatidylinositol-anchored ceruloplasmin is required for iron efflux from cells in the central nervous system. J Biol Chem. 2003;278(29):27144–8. [DOI] [PubMed] [Google Scholar]
- 222.Harris ZL, Takahashi Y, Miyajima H, Serizawa M, MacGillivray RT, Gitlin JD. Aceruloplasminemia: molecular characterization of this disorder of iron metabolism. Proc Natl Acad Sci U S A. 1995;92(7):2539–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Kono S, Yoshida K, Tomosugi N, Terada T, Hamaya Y, Kanaoka S, et al. Biological effects of mutant ceruloplasmin on hepcidin-mediated internalization of ferroportin. Biochim Biophys Acta. 2010;1802(11):968–75. [DOI] [PubMed] [Google Scholar]
- 224.Hellman NE, Kono S, Mancini GM, Hoogeboom AJ, De Jong GJ, Gitlin JD. Mechanisms of copper incorporation into human ceruloplasmin. J Biol Chem. 2002;277(48):46632–8. [DOI] [PubMed] [Google Scholar]
- 225.Marchi G, Busti F, Lira Zidanes A, Castagna A, Girelli D. Aceruloplasminemia: A Severe Neurodegenerative Disorder Deserving an Early Diagnosis. Front Neurosci. 2019;13:325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Vidal R, Ghetti B, Takao M, Brefel-Courbon C, Uro-Coste E, Glazier BS, et al. Intracellular ferritin accumulation in neural and extraneural tissue characterizes a neurodegenerative disease associated with a mutation in the ferritin light polypeptide gene. J Neuropathol Exp Neurol. 2004;63(4):363–80. [DOI] [PubMed] [Google Scholar]
- 227.Cozzi A, Rovelli E, Frizzale G, Campanella A, Amendola M, Arosio P, et al. Oxidative stress and cell death in cells expressing L-ferritin variants causing neuroferritinopathy. Neurobiol Dis. 2010;37(1):77–85. [DOI] [PubMed] [Google Scholar]
- 228.Barbeito AG, Garringer HJ, Baraibar MA, Gao X, Arredondo M, Nunez MT, et al. Abnormal iron metabolism and oxidative stress in mice expressing a mutant form of the ferritin light polypeptide gene. J Neurochem. 2009;109(4):1067–78. [DOI] [PMC free article] [PubMed] [Google Scholar]