Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2021 May 14;16(5):e0250606. doi: 10.1371/journal.pone.0250606

Iron activates microglia and directly stimulates indoleamine-2,3-dioxygenase activity in the N171-82Q mouse model of Huntington’s disease

David W Donley 1,2, Marley Realing 3, Jason P Gigley 4, Jonathan H Fox 1,2,*
Editor: Jane Foster5
PMCID: PMC8121302  PMID: 33989290

Abstract

Huntington’s disease (HD) is a neurodegenerative disorder caused by a dominant CAG-repeat expansion in the huntingtin gene. Microglial activation is a key feature of HD pathology, and is present before clinical disease onset. The kynurenine pathway (KP) of tryptophan degradation is activated in HD, and is thought to contribute to disease progression. Indoleamine-2,3-dioxygenase (IDO) catalyzes the first step in this pathway; this and other pathway enzymes reside with microglia. While HD brain microglia accumulate iron, the role of iron in promoting microglial activation and KP activity is unclear. Here we utilized the neonatal iron supplementation model to investigate the relationship between iron, microglial activation and neurodegeneration in adult HD mice. We show in the N171-82Q mouse model of HD microglial morphologic changes consistent with immune activation. Neonatal iron supplementation in these mice promoted neurodegeneration and resulted in additional microglial activation in adults as determined by increased soma volume and decreased process length. We further demonstrate that iron activates IDO, both in brain lysates and purified recombinant protein (EC50 = 1.24 nM). Brain IDO activity is increased by HD. Neonatal iron supplementation further promoted IDO activity in cerebral cortex, altered KP metabolite profiles, and promoted HD neurodegeneration as measured by brain weights and striatal volumes. Our results demonstrate that dietary iron is an important activator of microglia and the KP pathway in this HD model, and that this occurs in part through a direct effect on IDO. The findings are relevant to understanding how iron promotes neurodegeneration in HD.

Introduction

Huntington’s disease (HD) is an autosomal-dominant neurodegenerative disorder caused by a CAG-repeat expansion in exon 1 of the huntingtin gene (HTT) and results from expression of mutant huntingtin protein (mHTT) which misfolds in neurons and microglial cells [1,2]. Mutant HTT results in altered transcription, axonal transport, mitochondrial function, vesicular trafficking, inflammation, and oxidative stress are some of the mechanisms implicated in HD [3]. Exactly how these pathways are related is not fully understood, but they result in progressive neuronal dysfunction and loss, eventually leading to clinical disease. Symptoms of HD encompass a hyperkinetic motor disorder, cognitive decline, and psychiatric problems [4]. Currently, there are no interventions that delay HD onset or progression.

Numerous studies have shown that iron accumulates in the brains of HD patients. Magnetic resonance imaging–based approaches have provided evidence that iron accumulation starts before clinical onset and progresses based on CAG expansion size [57]. Significant iron accumulation in HD patients occurs in the striatum and cerebral cortex, regions that show early degeneration, suggesting that iron accumulation may have a role in neurodegeneration [5,8]. Post-mortem studies also demonstrate increases in regional iron levels in brains from individuals with advanced HD [810]. However, the approaches used in these studies could not determine the cell types involved and do not reliably differentiate between the accumulation of iron in a safe form, such as ferritin-bound, and in a potentially toxic labile form. Thus it is not clear how accumulated iron contributes to HD pathogenesis.

Huntingtin protein is involved in iron homeostasis [11,12], but the mechanisms through which brain iron accumulates in HD are incompletely understood. Microglia have an important role in brain iron metabolism [13]; being phagocytic, they clear dying cells, which can result in iron uptake [14,15]. As the primary brain immune cell, microglial cells also accumulate iron in response to inflammation [1618]. Microglial iron accumulation is present in human HD brains and is associated with an activated microglial morphology [19]. Further, HD mouse microglia have increased ferritin expression and detectable iron (III) stores [19,20]. Iron within the ferritin shell is considered a safe form suggesting that microglia in HD demonstrate a protective response to brain iron accumulation and degeneration. Microglial iron promotes oxygen radical generation that potentiates inflammation and neurotoxicity in models of Parkinson’s disease [21,22]. However, in HD it is unclear if microglial iron also drives disease progression.

The kynurenine pathway (KP) of tryptophan metabolism is segregated between microglia and astrocytes, and is activated in HD [23,24]. The first step is the oxidation of tryptophan to kynurenine by the microglial enzyme indoleamine-2,3-dioxygenase (IDO). IDO is upregulated by microglial activation [25,26].and pathway activity is thought to contribute to HD progression via generation of increased amounts of neurotoxic intermediates 3-hydroxykynurenine (3-HK) and quinolinic acid [23,24,2729]. Iron modifies KP activity by activating 3-hydroxyanthanilic acid dioxygenase (3-HAO) and promoting synthesis of neurotoxic quinolinic acid [30]. Despite this, it is unknown if iron has a key regulatory role in the KP and if it modulates gatekeeping steps such as IDO-mediated oxidation of tryptophan.

Even though HD is caused only by CAG expansion in the huntingtin gene, an estimated 60% of the non-CAG variance in age of onset is explained by environmental factors [31]. We have previously used neonatal iron supplementation (NIS) to assess the possible role of high dietary iron intake as an environmental modifier in mouse models of HD. This model is relevant, as in most populations human infants are supplemented with iron to prevent deficiency. Recommended daily allowances for iron have been developed to prevent iron deficiency, and upper limits were established to avoid short-term adverse effects [32]; however, long-term consequences of early-life NIS are not understood [33]. We have previously shown that NIS in R6/2 and YAC128 mouse models of HD exacerbates disease progression [34,35]. We have additionally shown that NIS promotes markers of mitochondrial dysfunction in R6/2 mice [36], although the cell-specific mechanisms by which NIS promotes HD in mice are unknown.

HD patients develop presymptomatic microglial activation and elevated blood cytokine levels [3739]. Activated microglia display morphological changes that correlate with increased production of proinflammatory cytokines such as IL-1β and increased phagocytic activity [3941]. In HD mouse models, the detrimental role of inflammation is supported by adverse effects of proinflammatory responses to lipopolysaccharide and Toxoplasma gondii [37,42]. Despite evidence for a role of microglial in promoting HD, the role of iron as an activator of microglial is unclear. Here we used NIS in the N171-82Q model of human HD to investigate the relationship between iron, microglial and kynurenine pathway activation, and neurodegeneration. These mice develop neurodegeneration as evidenced by loss of brain and striatal volume, and neuronal loss [43]. We report that NIS preferentially activates microglial cells in HD mice, directly activates IDO enzymatic activity, and promotes neurodegeneration. The findings demonstrate that microglial are sensitive to iron-mediated activation and that this may promote neurodegeneration in HD.

Results

Elevated indoleamine-2,3-dioxygenase activity in HD mouse brain is decreased by iron chelation

We measured brain IDO activity in the striatum and cortex of WT and HD mice with and without NIS. IDO activity was significantly increased in striata (F(1,42) = 7.77, p = 0.0075) and cortices (F(1,42) = 13.57, p < 0.001) of HD mice regardless of NIS (Fig 1A and 1B). In HD but not WT cortices, NIS significantly increased IDO activity (p = 0.016). IDO activity in HD mice that received NIS was significantly decreased by the potent iron (III) chelator deferoxamine added ex vivo in striata (p = 0.0067) and cortices (p = 0.0028) (Fig 1A and 1B). IDO has two isoforms, IDO1 and IDO2. IDO1 is highly expressed in brain microglia as well as in peripheral tissues and is a regulator of KP activity during inflammation [4446]. The second isoform, IDO2, is more narrowly expressed, primarily in peripheral tissues, and has much lower activity as compared with IDO1 [46,47]. Therefore, we measured Ido1 and Ido2 transcript levels relative to beta-actin in WT and HD brains. Ido1 was not significantly increased in the striatum (F(1,20) = 1.36, p = 0.2581) but was increased in the cortex of HD mice (F(1,20) = 5.86, p = 0.0251) relative to WT mice regardless of NIS (Fig 1C and 1D). We found in both the striatum (F(1,20) = 4.42, p = 0.0484) and cortex (F(1,20) = 4.82, p = 0.0402) that NIS alone significantly increased Ido2 expression irrespective of mouse genotype (Fig 1E and 1F). Together, these data demonstrate that iron may have a direct role in activating enzyme activity and that Ido expression does not account for all the changes in enzyme activity.

Fig 1. Elevated IDO activity in HD mouse brains is decreased by ex-vivo iron chelation.

Fig 1

Mouse pups were supplemented with carbonyl iron from postnatal days 10–17 and then were sacrificed at 14 weeks of age. A. Striatal IDO activity is increased in N171-82Q HD mice and significantly decreased by ex vivo iron (III) chelation (50 μM deferoxamine). B. Cortical IDO activity is increased in iron-supplemented HD mice compared to non-supplemented HD mice and activity is decreased by ex vivo iron chelation. C. There is no difference in relative transcript levels of IDO1 in striatum. D. IDO1 mRNA is increased in cortex of HD mice at 14-weeks of age. E, F. Iron increases levels of IDO2 in striatum (E) and cortex (F) but there was no significant effect of HD on IDO2 mRNA. A, B. Bars represent means ± 95% CI. n = 9 wild-type and 11 HD mice in each group. *p<0.05, **p<0.01. C, F. Bars represent means ± SE, n = 6; *p<0.05.

Iron directly activates IDO

To test whether iron stimulates IDO enzymatic activity, we incubated mouse brain extracts with iron (II). Iron increased brain IDO activity (F(3,16) = 14.00, p < 0.001) in a dose-dependent manner (Fig 2A). To determine if iron directly activates IDO, we used purified human IDO1. We demonstrated a dose-dependent increase in IDO activity (F(5,10) = 6.42, p = 0.0064) with an EC50 = 1.24 nM for iron activation of IDO1 (Fig 2B). We then studied the effect of increased iron on cultured mouse microglial cells. After 2 hours of iron incubation in serum-free medium, we measured significantly increased IDO activity (F(6,14) = 6.04, p = 0.0027) (Fig 2C). To study whether this effect was a post-translational mechanism, we supplemented iron into microglial cultures in the presence of the protein translation inhibitor cycloheximide. We performed this experiment in serum-containing medium to reduce toxicity due to protein translation inhibition. Iron significantly increased IDO activity (F(3,19) = 14.11, p < 0.001); however, we found no difference in IDO activity with cycloheximide treatment, supporting a direct, post-translational activation of IDO by iron (Fig 2D).

Fig 2. Nanomolar levels of iron activates IDO.

Fig 2

The effect of iron on IDO activity was determined in brain extracts, purified protein, and the immortalized EOC20 mouse microglial cell line. A. Iron (II) ammonium sulfate increases IDO activity in protein extracts from 14-week-old WT mouse brains in a dose-dependent manner. n = 5. B. Iron (II) activates purified IDO. n = 3. C. Iron(II) added to cultured microglia in serum-free medium for 2 hours increased IDO activity. n = 3. D. Iron(II) was added to the medium of cultured microglia for 2 hours with and without addition of cycloheximide, and IDO activity was determined. IDO activity was significantly increased by iron but there was no effect of cycloheximide. n = 6. A, D. Data are shown as the mean ± 95% CI. B, C. Data are shown as the mean ± SE. *p<0.05, **p<0.01, ***p<0.001. A-C. Asterisks represent comparisons to samples without iron addition.

Neonatal iron supplementation exacerbates neurodegeneration in N171-82Q HD mice

NIS in R6/2 and YAC128 HD mouse models exacerbates neurodegeneration [34,35]. Consistent with this, both N171-82Q HD mice (F(1,61) = 77.70, p < 0.001) and NIS (F(1,61) = 4.28, p = 0.0428) significantly decreased brain weights (Fig 3A). There was a significant interaction (F(1,61) = 4.05, p = 0.0486) between HD and NIS (Fig 3A); iron-supplemented HD mice had significantly decreased brain weights compared with control HD mice (p = 0.0072) and iron-supplemented WT mice (p < 0.001). In addition, striatal volumes were significantly decreased in HD mice (F(1,40) = 11.70, p = 0.0015) (F(1,40) = 2.89, p = 0.0970) (Fig 3B). No difference in striatal volume was found comparing control HD mice with control WT mice (p = 0.3559). However, iron-supplemented HD mice had significantly decreased striatal volume compared with control HD mice (p = 0.0206) and with iron-supplemented WT mice (p = 0.0011). There was no difference among groups with respect to striatal neuron cell body volume (Fig 3C).

Fig 3. Neonatal iron supplementation exacerbates HD in the N171-82Q mouse model.

Fig 3

Mouse pups were administered neonatal iron from postnatal days 10–17 then were sacrificed at 14 weeks of age. A. Total brain wet weight decreased in HD mice at 14 weeks of age. n = 15 WT control, 19 WT NIS, 13 HD control, 17 HD NIS. B. Striatal volume measured using the Cavalieri method was decreased in iron-supplemented HD mice compared with both control HD mice and iron-supplemented WT mice. C. Cell body volume of striatal neurons was not significantly different among groups. Data are shown as the mean ± SE. **p<0.01, ***p<0.001. B, C. n = 10 WT control, 11 WT NIS, 12 HD control, 11 HD NIS.

Neonatal iron supplementation induces microglial activation in HD mice

Activated microglia demonstrate altered morphology and are present in HD [19,48]. We therefore quantified microglial morphology as an indicator of activation status. Microglia were characterized as ramified, primed, amoeboid, or reactive [48,49] (Fig 4A). To address whether iron supplementation and/or mouse genotype is associated with microglial activation we performed logistic regression. This allowed us to statistically test differences in the blinded, categorical assignment of microglial activation. Ramified cells are considered to be non-activated and perform a different function than activated microglia whereas the primed morphology is an intermediate functional and morphological phenotype between the non-activated ramified cells and the highly activated amoeboid and reactive microglia [50,51]. Therefore, we first compared the proportion of ramified microglia to the combined proportion of primed, amoeboid, and reactive cells. We found that iron significantly increased the probability of an activated microglial morphology. The proportion of ramified and primed microglia was then compared to highly activated amoeboid and reactive microglia to determine the probability of a microglial cell exhibiting a fully activated morphology. We again found that iron significantly increased microglial activation and that HD mice had a higher proportion of highly activated microglia (amoeboid or reactive) when supplemented with iron (p-values in Fig 4B). The percentages of the four morphologies relative to total microglial cells are illustrated in Fig 4B for comparison purposes. To extend these findings, we also quantified morphological characteristics using a quantitative neurolucida-based approach. NIS increased cell body volumes (F(1,124) = 8.74, p = 0.0037), but there was no difference between iron-supplemented WT and HD microglia (Fig 4C). We also found a significant NIS-HD interaction with respect to the number of microglial processes (F(3,40) = 4.8, p = 0.006); these were significantly increased in iron-supplemented HD mice relative to control HD mice (p = 0.0024) (Fig 4D). In contrast, HD mice had decreased process lengths (F(1,124) = 6.09, p = 0.0149), which were unaffected by iron supplementation (Fig 4E).

Fig 4. Neonatal iron supplementation and HD differentially alter microglial morphology.

Fig 4

Microglia in brain sections from 14-week-old N171-82Q mice were labeled with anti-Iba1, and their morphology was evaluated. A. Representative micrographs of microglial cells labeled with anti-Iba1 were identified and characterized as having one of four morphologies, amoeboid, primed, ramified, and reactive. Neurolucida traces are shown below each image. Scale bars = 20 μm. B. Iron increased the frequency of reactive microglia in iron-supplemented HD mice. The table shows p-values from logistic regression analysis of microglial morphologies. Iron significantly increased the probability of activated microglial morphologies. R = ramified; P = primed; A = amoeboid; Re = reactive. Each morphology is presented as a percent of total microglial cells within each mouse. The percentage of cells with each morphology relative to all microglial cells in a given mouse are shown. C. The microglial soma volume was significantly increased by NIS. D. Iron-supplemented HD mice have a significantly increased number of processes relative to vehicle-treated HD mice. E. The total process length was significantly decreased in HD mice relative to WT mice but was not changed by iron supplementation. B-F. n = 10 WT control, 11 WT NIS, 12 HD control, 11 HD NIS. Data are shown as the mean ± SE. *p<0.05, **p<0.01.

Labile iron accumulates in HD microglia

HD microglia accumulate ferritin-bound iron, which is considered non-pathogenic but is elevated in response to increased labile iron [19,52,53]. Labile iron has the potential to drive oxidative and inflammatory processes. We used a flow cytometry to identify brain microglial cells [54] and measure cytoplasmic labile iron using the fluorescent dye calcein AM. HD mice had increased labile iron in microglial cells (F(1,24) = 4.67, p = 0.0409), but there was no effect of NIS on labile iron accumulation (Fig 5A and 5B). To assess whether iron promoted increased generalized neuroinflammation, we quantified expression of the gene encoding kynurenine monooxygenase (Kmo) and the chemokine CCL2 (Ccl2). The KMO enzyme is downstream of IDO in the KP, and its upregulation favors production of neurotoxic pathway intermediates including 3-hydroxykynurenine and quinolinic acid, which are associated with neuroinflammation [44]. The chemokine CCL2 (MCP-1), expressed by both neurons and glia, recruits microglia and peripheral monocytes to sites of inflammation in the central nervous system. Expression of CCL2 is associated with microglial activation, but is an indicator of a more generalized, sustained inflammatory response [5456]. Kmo transcripts were increased both in HD mice (F(1,38) = 11.29, p = 0.0018) and by NIS (F(1,38) = 5.65, p = 0.0226) in the striatum and cortex (Fig 6A and 6B). In the striatum, non-supplemented HD mice showed an increase in Kmo transcripts compared with WT mice (p = 0.0447) and, in the cortex, iron-supplemented HD mice had increased levels of Kmo transcripts compared with non-supplemented HD mice (p = 0.007) and iron-supplemented WT mice (p = 0.0015). No differences in Ccl2 expression were found in the striatum or cortex (Fig 6C and 6D).

Fig 5. Labile iron accumulates in HD microglia.

Fig 5

Microglia, defined as CD11b+CD45+CX3CR1+ cells, were extracted from mouse brains at 14 weeks of age and labeled with calcein AM. A. Representative histograms of calcein AM fluorescence in brain microglia incubated with (red) or without (blue) deferiprone. B. Labile iron, defined as the difference in mean fluorescence intensity (MFI) between cells incubated with and without deferiprone, was significantly increased in HD microglia. n = 6–8. Data are shown as the mean ± SE. *p<0.05.

Fig 6. Kmo transcripts are increased by HD and NIS.

Fig 6

Gene expression was determined at 14 weeks of age. A. Kmo expression was increased in the mouse HD striatum relative to WT but NIS had not effect. B. Kmo expression in iron-supplemented HD mice was significantly increased in the cortex. C-D. Ccl2 expression was unchanged in the striatum (C) and cortex (D). n = 6.Data are shown as the mean ± SE. *p<0.05.

Kynurenine metabolism is increased by NIS and HD

KP metabolites provide additional markers of changes in pathway activity in response to NIS and HD. We used LC-MS to measure several metabolites simultaneously in brain. There were no changes in tryptophan levels among groups in either the cortex or striatum (Fig 7A and 7E). Kynurenine levels were increased by both neonatal iron (F(1,80) = 5.23, p = 0.0249) and the HD genotype (F(1,80) = 9.95, p = 0.0023) in the cortex and striatum, consistent with IDO activation (Fig 7B and 7F). We also found a significant effect of both neonatal iron (F(1,80) = 4.23, p = 0.043) and the HD genotype (F(1,80) = 12.7, p = 0.0006) on 3-hydroxykynurenine levels in the cortex and striatum (Fig 7C and 7G). Although we did not detect an effect of iron supplementation or the HD genotype alone on kynurenic acid levels in either the cortex or striatum, we did find a significant NIS-HD interaction (F(1,80) = 4.06, p = 0.0472) (Fig 7D and 7H). We also compared metabolite ratios to address the balance of kynurenine metabolism (Fig 7I–7L). The ratio of 3-hydroxykynurenine (3-HK) to kynurenic acid is important in determining the relative toxicity of 3-HK, with an increase in the 3-HK/kynurenic acid ratio indicating an increase in the production of neurotoxic kynurenine metabolites [27]. In both the cortex and striatum, we found that 3-HK/kynurenic acid ratios were increased in HD mice (F(1,40) = 7.07, p = 0.0112) but showed only a trend toward being increased by iron supplementation (F(1,40) = 3.09, p = 0.0864) (Fig 7K and 7L). In cortex, iron-supplemented HD mice had an increased 3-HK/kynurenic acid ratio compared with iron-supplemented WT mice (p = 0.0031) (Fig 7K).

Fig 7. Brain kynurenine metabolism is altered by the HD genotype and NIS.

Fig 7

Mouse pups were supplemented with iron from postnatal days 10–17. KP metabolite levels were measured in the cortex (A-D) and striatum (E-H) at 14 weeks of age and are presented per milligram of soluble brain protein. A. Cortical tryptophan levels were unchanged. B. Cortical kynurenine was increased in iron-supplemented WT mice and in HD mice relative to control WT mice. C. HD mice had elevated 3-HK in the cortex. D. Kynurenic acid levels in the cortex were unchanged as a result of iron supplementation or the HD genotype. E. Striatal tryptophan levels were unchanged. F. Kynurenine was increased in the striatum of iron-supplemented WT mice and in HD mice relative to control WT mice. G. Striatal 3-HK was elevated by iron supplementation in WT mice and by the HD genotype H. Iron-supplemented HD mice had decreased striatal kynurenic acid compared with iron-supplemented WT mice. I. Cortical kynurenine/tryptophan (kyn/trp) ratios were increased in iron-supplemented WT and in HD mice. J. The kyn/trp ratio in the striatum was increased by iron supplementation and in HD mice relative to control WT mice. K. Iron-supplemented HD mice had increased 3-hydroxykynurenine/kynurenic acid (3-HK/kyna) ratios in the cortex compared with iron-supplemented WT mice. L. Striatal 3-HK/kyna ratios were increased by the HD only in the presence of iron supplementation. A-L. Data are shown as the mean ± SE. n = 10.

Discussion

Microglia are key immune cells in the brain and adapt to their environment by transitioning through a range of activation states. Neuroinflammation promotes microglial iron uptake [17] and microglial iron is increased in post-mortem human HD brain [19]. However, the effect of brain iron stress on microglial activation status in HD is less clear. By using the NIS model of promotion in a genetic mouse HD model we demonstrate that iron challenge promotes morphologic and biochemical evidence of microglial activation which is linked to neurodegeneration. The findings support a role of microglia in mediating the effects of iron on disease promotion in HD.

Neonatal iron supplementation activates microglial cells and promotes neurodegeneration in HD mice

Neonatal iron supplementation (NIS) is relevant to understanding the mechanisms through which iron promotes HD neurodegeneration because it models an iron-fortified diet that a human infant may receive, and as there is no detectable short-term toxicity [33]. Here we used the N171-82Q mouse model of HD for the first time to study the effects of NIS on microglial activation and neurodegeneration. We demonstrate that NIS potentiates neurodegeneration in these mice which is consistent with our finding in two other HD mouse models [34,35]. Microglial activation results in a graded transition of morphologic changes from ramified cells, which have many processes and small somas, to phagocytic cells, with their large somas and short processes [49]. To assess the role of a microglial pathway in promotion of neurodegeneration we used blinded categorical and continuous methods in parallel to assess microglial morphology. We demonstrate that N171-82Q mice have morphologic evidence of microglial activation as has been reported in the R6/2 model and human HD [19]. Importantly, activation status was further enhanced by NIS. The categorical assignment of brain microglia (ramified, primed, amoeboid, reactive) is the sum of several changes including increased soma size, a change in the number of processes, and a decrease in process length. NIS in WT mice led to increased soma volume and did not change the categorical assignment of microglia. However, iron-supplemented HD mice displayed increased soma volume and increased numbers of processes (resulting from NIS), and had increased microglial activation according to categorical assessment. These data indicate that microglial activation in iron-supplemented HD mice results from a synergism between iron and HD.

Cellular iron status is sensed by the central regulators of iron metabolism, iron-regulatory proteins 1 and 2 (IRP1/IRP2) [57]. Reports of decreased IRP1/IRP2 combined with increased microglial ferritin are consistent with iron stress in HD mice [19,20]. Iron stress is thought to result from labile iron which can generate hydroxyl radicals via redox chemistry, and modify iron-sensitive enzymes [58]. By isolating brain microglial cells and using flow cytometry, we have demonstrated for the first time that HD microglia have increased labile iron which supports the presence of iron stress in these cells. The calcein AM ester used to quantify labile iron is primarily a cytoplasmic iron marker because the esterases that trap the calcein after cleavage of the acetoxy-methyl group are predominantly cytoplasmic [59]. The failure to identify an effect of NIS on labile microglial iron could therefore also be explained by location of the iron pool, or possibly insufficient experimental power.

Labile iron directly activates IDO

The kynurenine pathway of tryptophan degradation is involved in the progression of HD and other brain disorders [44]. Activation of this pathway occurs early in human HD [23,24]. In addition, inhibition provides protection in HD model systems [29,60]. IDO is a cytoplasmic enzyme that is expressed in microglia and has an important role in regulating pathway activity. We have previously shown that IDO activity is increased in N171-82Q HD mice and that additional activation occurs in the presence of an infectious neuroinflammatory stimulus [42]. Consistent with a role in stimulating KP activity, we show that iron activated IDO in cell culture. Ido expression is primarily driven by pro-inflammatory signals such as lipopolysaccharide acting through the NLRP3 inflammasome [61], and IFNγ [25]. As iron can be pro-inflammatory we assessed the effect of NIS on Ido expression. Consistent with our prior study [42], we found increased cortical Ido1 in non-supplemented N171-82Q HD mice only. We additionally demonstrate using recombinant IDO, cultured microglia and brain homogenates that iron results in direct activation of IDO (Fig 2). The labile iron pool has been reported to be as high as 100–300 nM, but fluctuates based on cell type and environment [53,62,63]. We found that iron activates recombinant IDO with an EC50 of 1.24 nM which is in the physiological range for iron activation to occur in cells. Furthermore, supporting a direct effect of iron on IDO activity in vivo activation by NIS in HD mice was decreased by iron chelation ex vivo (Fig 1). Together, these data are consistent with a model whereby IDO activity in HD microglia results from basal gene upregulation by inflammatory signaling and direct activation by labile iron. IDO is inhibited at the post-translational level by nitric oxide [64,65]. However, to our knowledge, this is the first report of a post-translational activator of IDO. Previous structural analyses have identified allosteric sites on IDO. Based on our data showing that IDO activity is decreased by iron chelation in brain tissue, it is possible that iron acts at an allosteric site to increase activity [66,67]. One prevailing hypothesis of how IDO works is that tryptophan oxidation is accomplished through stepwise addition of oxygen molecules followed by a ring-break [68,69]. Therefore, it is also possible that free iron around the active site contributes to the ferryl oxygen and/or the tryptophan-epoxide intermediates during the nucleophilic addition of the second oxygen. Supporting diverse effects of iron on the KP, another study demonstrated that 3-hydroxyanthranilic acid dioxygenase, a downstream KP enzyme, is also activated by iron [30].

Neonatal iron supplementation activates kynurenine metabolism

To further investigate KP pathway responses in HD and NIS we quantified Kmo transcript and four key KP pathway metabolite levels. Despite demonstrating increased IDO activity in HD mice (Fig 1) similar increases in kynurenine, the product of this reaction, were not found suggesting increased activity of the downstream pathway. In one branch of the KP. KMO catalyzes the conversion of kynurenine to 3-HK in the first step of quinolinic acid synthesis, a pathway that promotes neurotoxicity [44]. We assessed this step by analysis of Kmo transcript and 3-HK levels; elevation of both in striata of NIS wild-type mice is consistent with activation of this step. However, Kmo and 3HK levels in cortex were discordant across experimental groups demonstrating a complex relationship between transcript and 3HK. CCL2 is expressed in neurons and glia and has a role in the activation and recruitment of immune cells, including microglia [55,56]. The failure to find significant changes in Ccl2 transcript levels suggests that there is not a generalized inflammatory response in HD/NIS mice, but a more specific microglial response involving the KP. In the second and neuroprotective branch of the KP, kynurenic acid is synthesized from kynurenine [44]. 3HK/kynurenine ratios, indicating toxic/protective pathway activities were increased by HD in striata and cortices with possible trends to additional increases with NIS. These findings indicate that NIS has differential effects in wild-type versus HD striata and cortices.

Taken together this study provides data supporting a causal relationship between NIS in the N171-82Q mouse model of HD, microglial activation, and neurodegeneration. NIS activated microglial differentially in wild-type and HD mice. However, NIS only promoted neurodegeneration in HD mice demonstrating specifically vulnerability to the effects of NIS in this nutritionally-relevant model. Individuals with HD have a pro-inflammatory macrophage phenotype and elevated peripheral TNFα [37,70]. The finding of IDO activation by iron is novel and relevant to not only neurodegenerative processes but also cancer where IDO is implicated in immune regulation [71]. Additional studies could focus on the mechanism of iron activation of IDO, and how NIS alters the microglial inflammatory phenotype in HD.

Materials and methods

Materials

All reagents were from Sigma unless otherwise stated. Validation of antibody was performed by the manufacturer. There are no restrictions on availability.

Mouse husbandry and breeding

N171-82Q Huntington’s disease mice (Jackson Labs, Bar Harbor, ME; strain B6C3-Tg(HD82Gln)81Gschi/J) were maintained as described [42]. Briefly, mice were group housed with ad-libitum access to food and water and were maintained in a 12-hour light/dark cycle. Sentinel mice were evaluated every 6 months by comprehensive serology panel screening for murine infectious diseases at Charles River Laboratories and were free of all tested diseases. Mice received daily welfare checks. At the designated endpoint, mice were humanely sacrificed using intraperitoneal injection of B-euthanasia solution in the home cage. This study and all procedures used were approved by the University of Wyoming Institutional Animal Care and Use Committee (UW-IACUC) (protocol number 20150813JF00190-01) in accordance with the National Institutes of Health guidelines.

Experimental design

N171-82Q HD mice were used for all experiments. The N171-82Q mouse is a transgenic model of HD that expresses the N-terminal 171 amino acids of human huntingtin protein with 82 poly-glutamine repeats under the control of the prion protein promotor [72]. When both males and females were used for experiments, the sex of the mice was included in the statistical analysis. Mouse pups received carbonyl iron supplementation or vehicle control by oral gavage from postnatal day 10–17 as described [34,35]. HD and WT littermate mice were assigned a unique 3-digit identifiers and systematically assigned to experimental groups at weaning to balance their ages and sex and to minimize effects of litter of origin. Experiments were set up with a 2 × 2 factorial design with genotype and iron treatment as the factors. Endpoints and outcomes were pre-determined. All researchers were blinded to the genotype and treatment group at weaning and remained blinded until data analysis. All experimental animals are accounted for in the data presented with the exception of three spontaneous mouse deaths. One iron-supplemented WT mouse (at 7 weeks of age) and two control HD mice (at 11 and 13 weeks of age) died during the reported studies. This study was not pre-registered.

Brain indoleamine-2,3-dioxygenase activity

Dissected brain regions were snap frozen on dry ice and stored at −70°C until analysis. Samples were prepared and IDO activity was determined by HPLC-MS/MS as described [42]. Additionally, a separate aliquot was analyzed the same way but with addition of deferoxamine to the incubation buffer.

Brain gene expression by real-time quantitative PCR analysis

Total RNA was extracted from the cerebral cortex and striatum using phenol/chloroform and then the cDNA was prepared, and Ido expression was quantified as described [42]. Kmo and Ccl2 expression was analyzed using Applied Biosystems Taqman gene expression primer/probe combinations Mm1321343_m1 (Kmo) and Mm00441242_m1 (Ccl2). Expression was normalized to beta-actin using the Applied Biosystems Taqman gene expression primer/probe combination Mm00607939_s1. Gene expression was determined using 20 ng of cDNA per reaction.

Purified indoleamine-2,3-dioxygenase activity

Recombinant human IDO (Accession #P14902) was obtained from R&D Systems (#6030-AO-010) and frozen at −80°C until use. The incubation buffer was prepared by mixing 800 μM tryptophan, 40 mM ascorbic acid, 9000 U/ml catalase, and 40 μM methylene blue in HBSS (pH = 6.5) with 80 mM ascorbic acid in 0.405 M Tris (pH = 8.0) in equal parts and pre-warmed to 37°C. One hundred ng/ml purified protein (100 ng/ml) in HBSS was mixed in equal parts with the incubation buffer and incubated at 37°C for 10 minutes. The reaction was stopped by the addition of 20% (v/v) 5 M acetic acid and incubated at 50°C for 10 minutes. Samples were centrifuged at 12,000×g, 4°C for 10 minutes, and the supernatants were filtered through a 0.2-μm filter. Kynurenine was measured in the filtered supernatant as described [42].

Indoleamine-2,3-dioxygenase activity in cultured cells

EOC20 cells, an immortalized microglial cell line (ATCC CTRL-2469) were used. Cells were cultured according to the manufacturer’s instructions and were verified to be mycoplasma free just prior to experiments. For IDO measurements, cells were washed twice with prewarmed HBSS and then incubated with IDO incubation buffer as described [42] for 4 hours. The kynurenine concentration was quantified in the cell supernatant using the method described above. Cells were lysed, and protein levels quantified using a Bradford assay. IDO activity was defined as picomoles of kynurenine produced per microgram of protein per hour.

Brain kynurenine metabolite quantification

Dissected brain tissue was snap frozen on dry ice and stored at −80°C until analysis. Brain regions were prepared according to the protocol for IDO activity in brain tissue. Samples were prepared by mixing 450 μg brain protein 1:2 (v/v) with methanol containing 2% acetic acid and then centrifuging the mixture at 12,000×g for 10 minutes at 4°C. Supernatants were filtered through a Phenomenex Phree Phospholipid extraction column and a 0.2 μm filter. The samples were then run on a Waters Acquity UPLC-MS/MS with a methanol/2% acetic acid mobile phase gradient on a 2 × 100−mm Waters BEH C18 column. The sample (5 μl) was injected with a total flow rate of 0.3 ml/min and a total run time of 4.5 minutes. Analytes were verified using two M+H parent-daughter transitions. Tryptophan parent m/z = 205.13 and daughter m/z = 118.03 and 146.07; kynurenic acid parent m/z = 190.07 and daughter m/z = 88.96 and 116.03; kynurenine parent m/z = 209.04 and daughter m/z = 94.02 and 146.03; 3-HK parent m/z = 224.98 and daughter m/z = 110.04 and 162.05. Concentrations were quantified using QuanLynx (Waters) software from serial dilutions of standards.

Mouse neuropathology

Mice designated for stereological analysis were perfused with 4% paraformaldehyde and treated as described [34,35]. Briefly, fixed brains were sectioned at 40 μm on a freezing microtome, and serial sections were collected in 12-well plates so that one well contained every 12th brain section. Sections from one well were mounted and thionin stained. The Cavalieri method and nucleator methods in StereoInvestigator (MicroBrightField) software were used to determine striatal volume and neuronal cell body volume, respectively.

Microglial immunohistochemistry

Microglial morphology was characterized in the striatum at the level of the anterior commissure. Striatal sections were mounted, and microglia were immunolabeled with anti-Iba1 (RRID: AB_2636859, Abcam ab178846). Brain sections were permeabilized for 15 minutes in methanol containing 3% hydrogen peroxide and then blocked with 1× TBE, 0.05% (v/v) Tween-20, and 2% (v/v) normal serum. Sections were incubated for 3 days in primary antibody (1:1000) with appropriate controls in blocking buffer at 4°C. They were then washed and incubated for 24 hours with biotinylated secondary antibody prior to washing, labeling with streptavidin-HRP conjugate (Vector Labs, RRID:AB_2336827), and detection using 3,3’-diaminobenzidine per the manufacturer’s instructions. Reactions were quenched with water and then washed twice in 1× TBE prior to coverslipping with fluoromount G (Southern Biotech).

Microglial morphology characterization

Microglial cells were defined as Iba1-positive cells in brain sections. Two 50 μm image stacks were collected from the dorsal and medial portion of the striatum at the level of the anterior commissure. From these images, two independent approaches were used to quantify the microglial morphology. First, cells were classified as ramified, primed, reactive, or amoeboid as described [49]. Second, quantitative morphology parameters were determined by uploading images to Neurolucida software (MicroBrightField) and each cell was traced throughout the stack. All cells that were fully in the image stack were evaluated. Ten microglial cells or more were traced per brain.

Microglial labile iron analysis

We used flow cytometry to identify microglia purified from forebrains in combination with calcein AM detection of labile iron. In brief, mice were perfused for two minutes with cold, heparinized saline. Forebrains were dissected out and placed in cold HBSS and then diced, digested, and homogenized to isolate brain microglia as described [73,74]. Cells from each animal were plated at 1 × 106 cells/ml in four separate wells. Cells were incubated with primary antibody conjugates for 30 minutes. Microglial cells were labeled using CD11b, CD45, and CX3CR1 (Biolegend, RRID: AB_312791, AB_2563538, AB_2565700 respectively) and analyzed by flow cytometry. Microglial markers including anti-Iba1 and CD68 were not used at the risk of losing iron from the cell due to the permeabilization required for intracellular staining. Previously, calcein AM was used to characterize the labile iron pool in cells by comparing calcein AM fluorescence with and without an iron chelator added to cells [75,76]. Two wells from each brain were incubated with 10 μM deferiprone, a membrane permeable iron chelator, in HBSS for 30 minutes at room temperature, and two wells received buffer at the same time. Fixable Near-IR Live/Dead Stain (ThermoFisher Scientific) was used to exclude dead cells. To detect iron, the cells were also incubated with 20 μM calcein AM for 15 minutes. Cells were washed thoroughly, and flow cytometry data were collected using a Guava easyCyte HT flow cytometer (Millipore). Data were analyzed using FlowJo v9.3 software. The cell gating and compensation strategy was determined from no-stain, single-color, and fluorescence-minus-one controls for each fluorescence channel. The mean fluorescence intensity was determined for each sample. Labile iron in microglia was defined as the difference in the mean fluorescence intensity for cells from the same mouse that were labeled with calcein AM with and without deferiprone treatment, similar to that described previously [75,76].

Statistical analyses

Data were analyzed using SAS software version 9.2 (Cary, NC). Generalized linear modeling was used for single-time point analyses, and mixed modeling was used for repeated measures and matched data. Assumptions of normality and equal variance were verified. All main effects and interactions were investigated in the initial statistical model, and then we performed pre-planned pairwise comparisons. Non-normally distributed data were log transformed for analysis and are presented as the mean ± 95% CIs. Logistic regression was used to analyze the categorical data on microglial morphology and to determine interaction effects between treatment and genotype. Logistic regression yielded an inference on the odds of microglial activation based on an activated microglial morphology. This statistical method is powerful against non-linear effects and for data, such as microglial morphology, where the interval between variables is not constant. Sample sizes were determined based on previous experience using each outcome. Data points represent the average of technical duplicates unless otherwise indicated. Differences with p < 0.05 were considered significant.

Acknowledgments

The authors would like to thank Dr. Kenneth Gerow, University of Wyoming, for consulting on statistical analysis of microglial morphologic data.

Data Availability

Raw data is available from Mendeley Data (DOI: 10.17632/z6jnx6hjcg.1).

Funding Statement

Funding for this study was provided by NIH NINDS grants R01 NS079450 (Fox) and R56 NS097813 (Fox). The Wyoming NIH INBRE program, supported by the Institutional Development Award from NIH NIGMS 2P20GM103432, provided graduate and undergraduate fellowships to DWD and MR, respectively. The funding agencies had no input on study design, data analysis, or decision to publish.

References

  • 1.The Huntington’s Disease Collaborative Research Group. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. The Huntington’s Disease Collaborative Research Group (HDCRG). Cell. 1993;72(6):971–83. Epub 1993/03/26. 10.1016/0092-8674(93)90585-e . [DOI] [PubMed] [Google Scholar]
  • 2.JA H.P., Maurik vH, odKI C., MR T., Anna‐Aster dR, SM H., et al. Frequency of nuclear mutant huntingtin inclusion formation in neurons and glia is cell‐type‐specific. Glia. 2017;65(1):50–61. 10.1002/glia.23050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.La Spada AR, Weydt P, Pineda VV. Frontiers in Neuroscience. Huntington’s Disease Pathogenesis: Mechanisms and Pathways. In: Lo DC, Hughes RE, editors. Neurobiology of Huntington’s Disease: Applications to Drug Discovery. Boca Raton (FL): CRC Press/Taylor & Francis Llc.; 2011. [Google Scholar]
  • 4.Vonsattel JPG, DiFiglia M. Huntington disease. Journal of Neuropathology and Experimental Neurology. 1998;57(5):369–84. 10.1097/00005072-199805000-00001 [DOI] [PubMed] [Google Scholar]
  • 5.Bartzokis G, Cummings J, Perlman S, Hance DB, Mintz J. Increased basal ganglia iron levels in huntington disease. Archives of Neurology. 1999;56(5):569–74. 10.1001/archneur.56.5.569 [DOI] [PubMed] [Google Scholar]
  • 6.Vymazal J, Klempíř J, Jech R, Židovská J, Syka M, Růžička E, et al. MR relaxometry in Huntington’s disease: Correlation between imaging, genetic and clinical parameters. Journal of the Neurological Sciences. 2007;263(1):20–5. 10.1016/j.jns.2007.05.018 [DOI] [PubMed] [Google Scholar]
  • 7.Jurgens CK, Jasinschi R, Ekin A, Witjes-Ane MN, Middelkoop H, van der Grond J, et al. MRI T2 Hypointensities in basal ganglia of premanifest Huntington’s disease. PLoS currents. 2010;2. Epub 2010/09/30. 10.1371/currents.RRN1173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Rosas HD, Chen YI, Doros G, Salat DH, Chen NK, Kwong KK, et al. Alterations in brain transition metals in Huntington disease: an evolving and intricate story. Arch Neurol. 2012;69(7):887–93. Epub 2012/03/07. 10.1001/archneurol.2011.2945 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.van Bergen JMG, Hua J, Unschuld PG, Lim IAL, Jones CK, Margolis RL, et al. Quantitative susceptibility mapping suggests altered brain iron in premanifest Huntington’s disease. AJNR American journal of neuroradiology. 2016;37(5):789–96. 10.3174/ajnr.A4617 PMC4867278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dexter DT, Carayon A, Javoy-Agid F, Agid Y, Wells FR, Daniel SE, et al. Alterations in the levels of iron, ferritin and other trace metals in Parkinson’s disease and other neurodegenerative diseases affecting the basal ganglia. Brain: a journal of neurology. 1991;114 (Pt 4):1953–75. Epub 1991/08/01. 10.1093/brain/114.4.1953 . [DOI] [PubMed] [Google Scholar]
  • 11.Dietrich P, Johnson IM, Alli S, Dragatsis I. Elimination of huntingtin in the adult mouse leads to progressive behavioral deficits, bilateral thalamic calcification, and altered brain iron homeostasis. PLOS Genetics. 2017;13(7):e1006846. 10.1371/journal.pgen.1006846 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lumsden AL, Henshall TL, Dayan S, Lardelli MT, Richards RI. Huntingtin-deficient zebrafish exhibit defects in iron utilization and development. Human Molecular Genetics. 2007;16(16):1905–20. 10.1093/hmg/ddm138 [DOI] [PubMed] [Google Scholar]
  • 13.Andersen HH, Johnsen KB, Moos T. Iron deposits in the chronically inflamed central nervous system and contributes to neurodegeneration. Cellular and Molecular Life Sciences. 2014;71(9):1607–22. 10.1007/s00018-013-1509-8 PMC3983878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Yoshida T, Tanaka M, Sotomatsu A, Hirai S. Activated microglia cause superoxide-mediated release of iron from ferritin. Neuroscience letters. 1995;190(1):21–4. Epub 1995/04/28. 10.1016/0304-3940(95)11490-n . [DOI] [PubMed] [Google Scholar]
  • 15.Rathnasamy G, Ling EA, Kaur C. Iron and iron regulatory proteins in amoeboid microglial cells are linked to oligodendrocyte death in hypoxic neonatal rat periventricular white matter through production of proinflammatory cytokines and reactive oxygen/nitrogen species. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2011;31(49):17982–95. Epub 2011/12/14. 10.1523/jneurosci.2250-11.2011 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lopes KO, Sparks DL, Streit WJ. Microglial dystrophy in the aged and Alzheimer’s disease brain is associated with ferritin immunoreactivity. Glia. 2008;56(10):1048–60. Epub 2008/04/30. 10.1002/glia.20678 . [DOI] [PubMed] [Google Scholar]
  • 17.McCarthy RC, Sosa JC, Gardeck AM, Baez AS, Lee CH, Wessling-Resnick M. Inflammation-induced iron transport and metabolism by brain microglia. J Biol Chem. 2018;293(20):7853–63. Epub 2018/04/04. 10.1074/jbc.RA118.001949 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Castelnau PA, Garrett RS, Palinski W, Witztum JL, Campbell IL, Powell HC. Abnormal iron deposition associated with lipid peroxidation in transgenic mice expressing interleukin-6 in the brain. J Neuropathol Exp Neurol. 1998;57(3):268–82. Epub 1998/05/26. 10.1097/00005072-199803000-00008 . [DOI] [PubMed] [Google Scholar]
  • 19.Simmons Danielle A, 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. 10.1002/glia.20526 [DOI] [PubMed] [Google Scholar]
  • 20.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. 10.1371/journal.pone.0077023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Zhang W, Yan Z-f, Gao J-h, Sun L, Huang X-y, Liu Z, et al. Role and Mechanism of Microglial Activation in Iron-Induced Selective and Progressive Dopaminergic Neurodegeneration. Molecular neurobiology. 2014;49(3):1153–65. 10.1007/s12035-013-8586-4 PMC4878835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Mehlhase J, Gieche J, Widmer R, Grune T. Ferritin levels in microglia depend upon activation: Modulation by reactive oxygen species. Biochimica et 10.1016/j.bbamcr.2006.04.012 2006;1763(8):854–9. 10.1016/j.bbamcr.2006.04.012. [DOI] [PubMed] [Google Scholar]
  • 23.Beal MF, Matson WR, Swartz KJ, Gamache PH, Bird ED. Kynurenine pathway measurements in Huntington’s disease striatum: evidence for reduced formation of kynurenic acid. Journal of neurochemistry. 1990;55(4):1327–39. Epub 1990/10/01. 10.1111/j.1471-4159.1990.tb03143.x . [DOI] [PubMed] [Google Scholar]
  • 24.Guidetti P, Hemachandra Reddy P, Tagle DA, Schwarcz R. Early kynurenergic impairment in Huntington’s Disease and in a transgenic animal model. Neuroscience letters. 2000;283(3):233–5. 10.1016/s0304-3940(00)00956-3 [DOI] [PubMed] [Google Scholar]
  • 25.Yadav MC, Burudi EME, Alirezaei M, Flynn CC, Watry DD, Lanigan CM, et al. IFN-γ induced IDO and WRS expression in microglia is differentially regulated by IL-4. Glia. 2007;55(13):1385–96. 10.1002/glia.20544 PMC2486430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Corona AW, Norden DM, Skendelas JP, Huang Y, O’Connor JC, Lawson M, et al. Indoleamine 2,3-dioxygenase inhibition attenuates lipopolysaccharide induced persistent microglial activation and depressive-like complications in fractalkine receptor (CX3CR1)-deficient mice. Brain, Behavior, and Immunity. 2013;31:134–42. 10.1016/j.bbi.2012.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Campesan S, Green EW, Breda C, Sathyasaikumar KV, Muchowski PJ, Schwarcz R, et al. The kynurenine pathway modulates neurodegeneration in a Drosophila model of Huntington’s disease. Current biology: CB. 2011;21(11):961–6. Epub 2011/06/04. 10.1016/j.cub.2011.04.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Guidetti P, Luthi-Carter RE, Augood SJ, Schwarcz R. Neostriatal and cortical quinolinate levels are increased in early grade Huntington’s disease. Neurobiology of Disease. 2004;17(3):455–61. 10.1016/j.nbd.2004.07.006 [DOI] [PubMed] [Google Scholar]
  • 29.Zwilling D, Huang S-Y, Sathyasaikumar Korrapati V, Notarangelo Francesca M, Guidetti P, Wu H-Q, et al. Kynurenine 3-Monooxygenase Inhibition in Blood Ameliorates Neurodegeneration. Cell. 2011;145(6):863–74. 10.1016/j.cell.2011.05.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Stachowski EK, Schwarcz R. Regulation of quinolinic acid neosynthesis in mouse, rat and human brain by iron and iron chelators in vitro. Journal of neural transmission (Vienna, Austria: 1996). 2012;119(2):123–31. Epub 2011/08/13. 10.1007/s00702-011-0694-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wexler NS. Venezuelan kindreds reveal that genetic and environmental factors modulate Huntington’s disease age of onset. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(10):3498–503. 10.1073/pnas.0308679101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Micronutrients IoMPo. Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc.: National Academies Press (US); 2001. [cited 2018]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK222310/. [PubMed] [Google Scholar]
  • 33.Agrawal S, Berggren KL, Marks E, Fox JH. Impact of high iron intake on cognition and neurodegeneration in humans and in animal models: a systematic review. Nutrition Reviews. 2017;75(6):456–70. 10.1093/nutrit/nux015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Berggren KL, Chen J, Fox J, Miller J, Dodds L, Dugas B, et al. Neonatal iron supplementation potentiates oxidative stress, energetic dysfunction and neurodegeneration in the R6/2 mouse model of Huntington’s disease. Redox Biology. 2015;4:363–74. 10.1016/j.redox.2015.02.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Berggren KL, Lu Z, Fox JA, Dudenhoeffer M, Agrawal S, Fox JH. Neonatal Iron Supplementation Induces Striatal Atrophy in Female YAC128 Huntington’s Disease Mice. Journal of Huntington’s disease. 2016;5(1):53–63. Epub 2016/04/16. 10.3233/JHD-150182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Agrawal S, Fox J, Thyagarajan B, Fox J. Brain mitochondrial iron accumulates in Huntington’s disease, mediates mitochondrial dysfunction, and can be removed pharmacologically. Free Radical Biology and Medicine. 2018. 10.1016/j.freeradbiomed.2018.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Björkqvist M, Wild EJ, Thiele J, Silvestroni A, Andre R, Lahiri N, et al. A novel pathogenic pathway of immune activation detectable before clinical onset in Huntington’s disease. The Journal of Experimental Medicine. 2008;205(8):1869–77. 10.1084/jem.20080178 PMC2525598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Tai YF, Pavese N, Gerhard A, Tabrizi SJ, Barker RA, Brooks DJ, et al. Microglial activation in presymptomatic Huntington’s disease gene carriers. Brain: a journal of neurology. 2007;130(Pt 7):1759–66. Epub 2007/04/03. 10.1093/brain/awm044 . [DOI] [PubMed] [Google Scholar]
  • 39.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. Epub 2001/03/29. 10.1093/jnen/60.2.161 . [DOI] [PubMed] [Google Scholar]
  • 40.Fernández-Arjona MdM, Grondona JM, Granados-Durán P, Fernández-Llebrez P, López-Ávalos MD. Microglia Morphological Categorization in a Rat Model of Neuroinflammation by Hierarchical Cluster and Principal Components Analysis. Frontiers in Cellular Neuroscience. 2017;11(235). 10.3389/fncel.2017.00235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Davis BM, Salinas-Navarro M, Cordeiro MF, Moons L, De Groef L. Characterizing microglia activation: a spatial statistics approach to maximize information extraction. Scientific Reports. 2017;7(1):1576. 10.1038/s41598-017-01747-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Donley DW, Olson AR, Raisbeck MF, Fox JH, Gigley JP. Huntingtons Disease Mice Infected with Toxoplasma gondii Demonstrate Early Kynurenine Pathway Activation, Altered CD8+ T-Cell Responses, and Premature Mortality. PLOS ONE. 2016;11(9):e0162404. 10.1371/journal.pone.0162404 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ferrante RJ. Mouse models of Huntington’s disease and methodological considerations for therapeutic trials. Biochim Biophys Acta. 2009;1792(6):506–20. Epub 2009/04/14. 10.1016/j.bbadis.2009.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Schwarcz R, Bruno JP, Muchowski PJ, Wu H-Q. Kynurenines in the mammalian brain: when physiology meets pathology. Nature Reviews Neuroscience. 2012;13:465. 10.1038/nrn3257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lee A, Kanuri N, Zhang Y, Sayuk GS, Li E, Ciorba MA. IDO1 and IDO2 Non-Synonymous Gene Variants: Correlation with Crohn’s Disease Risk and Clinical Phenotype. PLOS ONE. 2014;9(12):e115848. 10.1371/journal.pone.0115848 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ball HJ, Sanchez-Perez A, Weiser S, Austin CJD, Astelbauer F, Miu J, et al. Characterization of an indoleamine 2,3-dioxygenase-like protein found in humans and mice. Gene. 2007;396(1):203–13. 10.1016/j.gene.2007.04.010 [DOI] [PubMed] [Google Scholar]
  • 47.Metz R, DuHadaway JB, Kamasani U, Laury-Kleintop L, Muller AJ, Prendergast GC. Novel Tryptophan Catabolic Enzyme IDO2 Is the Preferred Biochemical Target of the Antitumor Indoleamine 2,3-Dioxygenase Inhibitory Compound D-1-Methyl-Tryptophan. Cancer Research. 2007;67(15):7082–7. 10.1158/0008-5472.CAN-07-1872 [DOI] [PubMed] [Google Scholar]
  • 48.Kraft AD, Kaltenbach LS, Lo DC, Harry GJ. Activated microglia proliferate at neurites of mutant huntingtin-expressing neurons. Neurobiol Aging. 2012;33(3):621.e17–33. 10.1016/j.neurobiolaging.2011.02.015 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Torres-Platas SG, Comeau S, Rachalski A, Bo GD, Cruceanu C, Turecki G, et al. Morphometric characterization of microglial phenotypes in human cerebral cortex. Journal of Neuroinflammation. 2014;11:12–. 10.1186/1742-2094-11-12 PMC3906907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kierdorf K, Prinz M. Factors regulating microglia activation. Frontiers in Cellular Neuroscience. 2013;7(44). 10.3389/fncel.2013.00044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lawson LJ, Perry VH, Dri P, Gordon S. Heterogeneity in the distribution and morphology of microglia in the normal adult mouse brain. Neuroscience. 1990;39(1):151–70. Epub 1990/01/01. 10.1016/0306-4522(90)90229-w . [DOI] [PubMed] [Google Scholar]
  • 52.Picard V, Epsztejn S, Santambrogio P, Cabantchik ZI, Beaumont C. Role of Ferritin in the Control of the Labile Iron Pool in Murine Erythroleukemia Cells. Journal of Biological Chemistry. 1998;273(25):15382–6. 10.1074/jbc.273.25.15382 [DOI] [PubMed] [Google Scholar]
  • 53.Konijn AM, Glickstein H, Vaisman B, Meyron-Holtz EG, Slotki IN, Cabantchik ZI. The Cellular Labile Iron Pool and Intracellular Ferritin in K562 Cells. Blood. 1999;94(6):2128–34. [PubMed] [Google Scholar]
  • 54.Mizutani M, Pino PA, Saederup N, Charo IF, Ransohoff RM, Cardona AE. The fractalkine receptor but not CCR2 is present on microglia from embryonic development throughout adulthood(). Journal of immunology (Baltimore, Md: 1950). 2012;188(1):29–36. 10.4049/jimmunol.1100421 PMC3244524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Conductier G, Blondeau N, Guyon A, Nahon JL, Rovere C. The role of monocyte chemoattractant protein MCP1/CCL2 in neuroinflammatory diseases. Journal of neuroimmunology. 2010;224(1–2):93–100. Epub 2010/08/04. 10.1016/j.jneuroim.2010.05.010 . [DOI] [PubMed] [Google Scholar]
  • 56.Hinojosa AE, Garcia-Bueno B, Leza JC, Madrigal JL. CCL2/MCP-1 modulation of microglial activation and proliferation. J Neuroinflammation. 2011;8:77. Epub 2011/07/07. 10.1186/1742-2094-8-77 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Klausner RD, Rouault TA, Harford JB. Regulating the fate of mRNA: The control of cellular iron metabolism. Cell. 1993;72(1):19–28. 10.1016/0092-8674(93)90046-s [DOI] [PubMed] [Google Scholar]
  • 58.Wang B, Lu J, Dubey KD, Dong G, Lai W, Shaik S. How do Enzymes Utilize Reactive OH Radicals? Lessons from Nonheme HppE and Fenton Systems. Journal of the American Chemical Society. 2016;138(27):8489–96. 10.1021/jacs.6b03555 [DOI] [PubMed] [Google Scholar]
  • 59.Tenopoulou M, Kurz T, Doulias P-T, Galaris D, Brunk Ulf T. Does the calcein-AM method assay the total cellular ‘labile iron pool’ or only a fraction of it? Biochemical Journal. 2007;403(Pt 2):261–6. 10.1042/BJ20061840 PMC1874234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Giorgini F, Guidetti P, Nguyen Q, Bennett SC, Muchowski PJ. A genomic screen in yeast implicates kynurenine 3-monooxygenase as a therapeutic target for Huntington’s disease. Nature genetics. 2005;37(5):526–31. 10.1038/ng1542 PMC1449881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zhang S, Zong Y, Ren Z, Hu J, Wu X, Xiao H, et al. Regulation of indoleamine 2, 3-dioxygenase in hippocampal microglia by NLRP3 inflammasome in lipopolysaccharide-induced depressive-like behaviors. Eur J Neurosci. 2020;52(11):4586–601. Epub 2020/10/25. 10.1111/ejn.15016 . [DOI] [PubMed] [Google Scholar]
  • 62.Breuer W, Epsztejn S, Cabantchik ZI. Dynamics of the cytosolic chelatable iron pool of K562 cells. FEBS letters. 1996;382(3):304–8. Epub 1996/03/18. 10.1016/0014-5793(96)00190-1 . [DOI] [PubMed] [Google Scholar]
  • 63.Epsztejn S, Kakhlon O, Glickstein H, Breuer W, Cabantchik ZI. Fluorescence Analysis of the Labile Iron Pool of Mammalian Cells. Analytical Biochemistry. 1997;248(1):31–40. 10.1006/abio.1997.2126 [DOI] [PubMed] [Google Scholar]
  • 64.Fujigaki H, Seishima M, Saito K. Posttranslational modification of indoleamine 2,3-dioxygenase. Analytical and Bioanalytical Chemistry. 2012;403(7):1777–82. 10.1007/s00216-012-5946-2 [DOI] [PubMed] [Google Scholar]
  • 65.Thomas SR, Terentis AC, Cai H, Takikawa O, Levina A, Lay PA, et al. Post-translational Regulation of Human Indoleamine 2,3-Dioxygenase Activity by Nitric Oxide. Journal of Biological Chemistry. 2007;282(33):23778–87. 10.1074/jbc.M700669200 [DOI] [PubMed] [Google Scholar]
  • 66.Lewis-Ballester A, Pham KN, Batabyal D, Karkashon S, Bonanno JB, Poulos TL, et al. Structural insights into substrate and inhibitor binding sites in human indoleamine 2,3-dioxygenase 1. Nature Communications. 2017;8(1):1693. 10.1038/s41467-017-01725-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Pearson JT, Siu S, Meininger DP, Wienkers LC, Rock DA. In Vitro Modulation of Cytochrome P450 Reductase Supported Indoleamine 2,3-Dioxygenase Activity by Allosteric Effectors Cytochrome b5 and Methylene Blue. Biochemistry. 2010;49(12):2647–56. 10.1021/bi100022c [DOI] [PubMed] [Google Scholar]
  • 68.Booth ES, Basran J, Lee M, Handa S, Raven EL. Substrate Oxidation by Indoleamine 2,3-Dioxygenase: Evidence for a Common Reaction Mechanism. Journal of Biological Chemistry. 2015. 10.1074/jbc.M115.695684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Capece L, Lewis-Ballester A, Yeh S-R, Estrin DA, Marti MA. Complete Reaction Mechanism of Indoleamine 2,3-Dioxygenase as Revealed by QM/MM Simulations. The Journal of Physical Chemistry B. 2012;116(4):1401–13. 10.1021/jp2082825 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Di Pardo A, Alberti S, Maglione V, Amico E, Cortes EP, Elifani F, et al. Changes of peripheral TGF-β1 depend on monocytes-derived macrophages in Huntington disease. Molecular Brain. 2013;6:55–. 10.1186/1756-6606-6-55 PMC4029620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bilir C, Sarisozen C. Indoleamine 2,3-dioxygenase (IDO): Only an enzyme or a checkpoint controller? Journal of Oncological Sciences. 2017;3(2):52–6. 10.1016/j.jons.2017.04.001. [DOI] [Google Scholar]
  • 72.Schilling G, Klevytska A, Tebbenkamp AT, Juenemann K, Cooper J, Gonzales V, et al. Characterization of huntingtin pathologic fragments in human Huntington disease, transgenic mice, and cell models. J Neuropathol Exp Neurol. 2007;66(4):313–20. Epub 2007/04/07. 10.1097/nen.0b013e318040b2c8 . [DOI] [PubMed] [Google Scholar]
  • 73.Grabert K, Michoel T, Karavolos MH, Clohisey S, Baillie JK, Stevens MP, et al. Microglial brain region−dependent diversity and selective regional sensitivities to aging. Nature Neuroscience. 2016;19:504. 10.1038/nn.4222 https://www.nature.com/articles/nn.4222#supplementary-information. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Nikodemova M, Watters JJ. Efficient isolation of live microglia with preserved phenotypes from adult mouse brain. Journal of Neuroinflammation. 2012;9(1):147. 10.1186/1742-2094-9-147 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ali A, Zhang Q, Dai J, Huang X. Calcein as a fluorescent iron chemosensor for the determination of low molecular weight iron in biological fluids. Biometals. 2003;16(2):285–93. 10.1023/a:1020642808437 [DOI] [PubMed] [Google Scholar]
  • 76.Prus E, Fibach E. Flow cytometry measurement of the labile iron pool in human hematopoietic cells. Cytometry Part A. 2007;73A(1):22–7. 10.1002/cyto.a.20491 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Jane Foster

19 Mar 2021

PONE-D-21-02271

Iron activates microglia and directly stimulates indoleamine-2,3-dioxygenase activity in the N171-82Q mouse model of Huntington’s disease

PLOS ONE

Dear Dr. Fox,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please submit your revised manuscript by May 03 2021 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

We look forward to receiving your revised manuscript.

Kind regards,

Jane Foster, PhD

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2.  We note that you have stated that you will provide repository information for your data at acceptance. Should your manuscript be accepted for publication, we will hold it until you provide the relevant accession numbers or DOIs necessary to access your data. If you wish to make changes to your Data Availability statement, please describe these changes in your cover letter and we will update your Data Availability statement to reflect the information you provide.

Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

Additional Editor Comments (if provided):

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Partly

Reviewer #2: Yes

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: I Don't Know

Reviewer #2: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: Overall the manuscript is clearly written with only a few typographical errors. Statistical analysis is described but it is beyond my expertise to evaluate the statistical methods employed. Methods are well described.

Major points:

1) One conclusion, that iron promotes neurodegeneration, is supported by brain weights and striatal volumes. Is there precedence in the literature that these measurements are sufficient to support this conclusion in the absence of any data specifically implicating neurons? Or would “brain and striatal atrophy” be a more appropriate description of what is being examined?

2) It is stated “NIS alone significantly increased Ido2 104 expression irrespective of mouse genotype (Fig. 1E,F)” but there are no indications of statistical significance in Figure 1E and 1F.

3) In the text EOC 20 cells should be referred to as “an immortalized microglial cell line” and not simply “microglia”

4) The statement in lines 195/196 about CCL2 activating microglia is inaccurate and needs to be revised.

Other points:

1) Please define KP at first use in abstract

2) Please state what deferoxamine does at first use in main text results.

3) In describing studies please be more consistent with use of terms- ironII, iron, ferrous iron and ferrous ammonium sulfate are all used at one place to describe in vitro studies

Reviewer #2: In this manuscript Donley and colleagues “utilized the neonatal iron supplementation model to investigate the relationship between iron, microglial activation and neurodegeneration in adult Huntington's Disease mice”. The authors showed that “neonatal iron supplementation in these mice promoted neurodegeneration and resulted in additional microglial activation”. The authors demonstrated that “that iron activates

IDO” and that “brain IDO activity is increased by HD”.

This is a dense manuscript from J. Fox’s laboratory that works for years in HD and iron. It is a well crafted study, that provides new insights in neurodegeneration and therefore deserves publication.

Statistical analyses, review of previous works and extensive materials and methods provided.

The authors did a wonderful effort to discuss their results and provide intelligent interpretation.

Figures are informative although a little too crowded.

Minor point

Results: Figure 1E,F, since NIS significantly increased Ido2 please include asterisks in the corresponding figure bars.

Potential Concerns:

None identified

No concerns were raised about dual publication, research ethics, or publication ethics. All animal permissions and IACUC protocols exist. At the designated endpoint, mice were humanely sacrificed using intra-peritoneal injections of B-euthanasia solution in the home cage.

All data are fully available without restriction and included in the manuscript.

Funding information from NIH is also provided.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2021 May 14;16(5):e0250606. doi: 10.1371/journal.pone.0250606.r002

Author response to Decision Letter 0


4 Apr 2021

All my comments are in the response to reviewer file and are pasted below.

Dear editor and reviewers: We appreciate the input on our manuscript. Responding to this has enabled us to make further improvements. Please see the specific comments below that are responses to all the reviews comments.

We hope that you will now find this manuscript acceptable for publication in PLOS ONE.

Sincerely, Jonathan Fox

Line numbers refer to the finalized version without track changes.

Reviewer 1:

Major points:

1. The reviewer raises the question of whether loss of brain weight and striatal volume is sufficient to support the conclusion that there is neurodegeneration, or would brain/striatal atrophy be a more appropriate term to use. This a valid point. We do think there is significant precedence in the literature that our findings are sufficient to use the term “neurodegeneration”. There is overwhelming evidence in human HD that brain atrophy is driven to a large extent by neuron degeneration and loss (Curr Top Behav Neurosci. 2015;22:33-80). This may be also contributed by oligodendrocyte degeneration in white matter (PNAS May 7, 2019 116 (19) 9622-9627). The N171-82Q mouse model of HD demonstrate a number of changes that support neuron degeneration including TUNEL and silver stain positive neurons in striatum and or cortex (Curr Top Behav Neurosci. 2015;22:33-80; Neurobiology of Disease, June, 2008, 30;293-302). This is in addition to very well established loss of striatal and overall brain mass (Brain Pathol 2016 Nov;26(6):726-740). While we acknowledge that demonstrating neuron-specific changes would have enhanced the study, our interpretation of neurodegeneration is consistent with the current literature. To address this point in the text we have made the following addition. “These mice develop neurodegeneration as evidenced by loss of brain and striatal volume, and neuronal loss” and included a reference. Line 91.

2. Figs 1E and 1F have now been modified according to the reviewer’s suggestions.

3. EOC 20 has now been changed to “an immortalized microglial cell line”. See lines 135 and 395.

4. Statement about CCL activating microglia – We agree with the reviewer that the original statement was inaccurate. We have now changed this to -”The chemokine CCL2 (MCP-1), expressed by both neurons and glia, recruits microglia and peripheral monocytes to sites of inflammation in the central nervous system. Expression of CCL2 is associated with microglial activation, but is an indicator of a more generalized, sustained inflammatory response”. See lines 207-210.

Minor points:

1. KP is now defined correctly in the abstract.

2. We have now explained what deferoxamine does. We changed the sentence to “IDO activity in HD mice that received NIS was significantly decreased by the potent iron (III) chelator deferoxamine added ex vivo in striata (p = 0.0067) and cortices (p = 0.0028)”. See lines 100-101.

3. We have now made the terminology around iron more consistent. We have removed the term ferrous and used “iron (II)” instead. Most of the time we do not designate the oxidation state of iron because it is not necessary, or the form of iron is unknown.

Attachment

Submitted filename: ResponseToReviewers.pdf

Decision Letter 1

Jane Foster

12 Apr 2021

Iron activates microglia and directly stimulates indoleamine-2,3-dioxygenase activity in the N171-82Q mouse model of Huntingtons disease

PONE-D-21-02271R1

Dear Dr. Fox,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Jane Foster, PhD

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Acceptance letter

Jane Foster

4 May 2021

PONE-D-21-02271R1

Iron activates microglia and directly stimulates indoleamine-2,3-dioxygenase activity in the N171-82Q mouse model of Huntington’s disease

Dear Dr. Fox:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Jane Foster

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    Attachment

    Submitted filename: ResponseToReviewers.pdf

    Data Availability Statement

    Raw data is available from Mendeley Data (DOI: 10.17632/z6jnx6hjcg.1).


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES