nutrients

Article Oral Administration of Ginger-Derived Lipid Nanoparticles and Dmt1 siRNA Potentiates the Effect of Dietary Iron Restriction and Mitigates Pre-Existing in Hamp KO Mice

Xiaoyu Wang 1,2,†, Mingzhen Zhang 3,4,† , Regina R. Woloshun 2, Yang Yu 2 , Jennifer K. Lee 2 , Shireen R. L. Flores 2, Didier 3,5 and James F. Collins 2,*

1 Key Laboratory of Precision Nutrition and Food Quality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; [email protected] 2 Food Science & Human Nutrition Department, University of Florida, Gainesville, FL 32611, USA; regina.woloshun@ufl.edu (R.R.W.); yuyang2@ufl.edu (Y.Y.); leejennifer@ufl.edu (J.K.L.); srflores@ufl.edu (S.R.L.F.) 3 Center for Diagnostics and Therapeutics, Institute for Biomedical Science, Georgia State University, Atlanta, GA 30303, USA; [email protected] (M.Z.); [email protected] (D.M.) 4 School of Basic Medical Science, Health Science Center, Institute of Medical Engineering, Xi’an Jiaotong University, Xi’an 710049, China   5 Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA * Correspondence: jfcollins@ufl.edu Citation: Wang, X.; Zhang, M.; † These authors contributed equally to this work. Woloshun, R.R.; Yu, Y.; Lee, J.K.; Flores, S.R.L.; Merlin, D.; Collins, J.F. Abstract: Intestinal iron transport requires an iron importer (Dmt1) and an iron exporter (Fpn1). The Oral Administration of hormone regulates iron absorption by modulating Fpn1 levels on the basolateral Ginger-Derived Lipid Nanoparticles surface of duodenal enterocytes. In the genetic, iron-loading disorder hereditary hemochromatosis and Dmt1 siRNA Potentiates the (HH), hepcidin production is low and Fpn1 protein expression is elevated. High Fpn1-mediated Effect of Dietary Iron Restriction and iron export depletes intracellular iron, causing a paradoxical increase in Dmt1-mediated iron import. Mitigates Pre-Existing Iron Overload in Hamp KO Mice. Nutrients 2021, 13, Increased activity of both transporters causes excessive iron absorption, thus initiating body iron 1686. https://doi.org/10.3390/ loading. Logically then, silencing of intestinal Dmt1 or Fpn1 could be an effective therapeutic nu13051686 intervention in HH. It was previously established that Dmt1 knock down prevented iron-loading in weanling Hamp (encoding hepcidin) KO mice (modeling type 2B HH). Here, we tested the hypothesis Academic Editor: Gladys that Dmt1 silencing combined with dietary iron restriction (which may be recommended for HH Oluyemisi Latunde-Dada patients) will mitigate iron loading once already established. Accordingly, adult Hamp KO mice were switched to a low-iron (LFe) diet and (non-toxic) folic acid-coupled, ginger nanoparticle-derived lipid Received: 12 March 2021 vectors (FA-GDLVs) were used to deliver negative-control (NC) or Dmt1 siRNA by oral, intragastric Accepted: 12 May 2021 gavage daily for 21 days. The LFe diet reduced body iron burden, and experimental interventions Published: 15 May 2021 potentiated iron losses. For example, Dmt1 siRNA treatment suppressed duodenal Dmt1 mRNA expression (by ~50%) and reduced serum and liver non-heme iron levels (by ~60% and >85%, Publisher’s Note: MDPI stays neutral respectively). Interestingly, some iron-related parameters were repressed similarly by FA-GDLVs with regard to jurisdictional claims in carrying either siRNA, including 59Fe (as FeCl ) absorption (~20% lower), pancreatic non-heme iron published maps and institutional affil- 3 iations. (reduced by ~65%), and serum (decreased 40–50%). Ginger may thus contain bioactive lipids that also influence iron homeostasis. In conclusion, the combinatorial approach of FA-GDLV and Dmt1 siRNA treatment, with dietary iron restriction, mitigated pre-existing iron overload in a murine model of HH.

Copyright: © 2021 by the authors. Keywords: hepcidin antimicrobial peptide; hereditary hemochromatosis; iron overload; divalent Licensee MDPI, Basel, Switzerland. This article is an open access article metal-ion transporter 1; Slc11a2; nanoparticles; ginger; GDLVs distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https:// creativecommons.org/licenses/by/ 4.0/).

Nutrients 2021, 13, 1686. https://doi.org/10.3390/nu13051686 https://www.mdpi.com/journal/nutrients Nutrients 2021, 13, 1686 2 of 16

1. Introduction Iron is an essential trace mineral with numerous notable biological functions. Iron homeostasis is unique among the dietary minerals as iron-excretory systems do not exist in humans or other mammals. Modulation of the rate of intestinal transport is thus critical to maintain overall body iron levels. Sophisticated homeostatic regulators of iron absorption have developed through evolutionary time, which collectively influence iron absorption in relation to body iron stores, erythroid demand, tissue oxygenation, and during infection and inflammation [1]. Hepcidin is a principal regulator of iron absorption. This liver- derived, peptide hormone controls serum iron levels by regulating iron efflux from cells that absorb (duodenal enterocytes) and store (reticuloendothelial macrophages) iron [2]. The Hamp (encoding hepcidin) is transactivated in hepatocytes when iron stores are replete and during inflammation; increased hepcidin suppresses intestinal iron absorption and lowers serum iron. During iron depletion, hypoxia and when erythroid demand is elevated, Hamp transcription is downregulated, thus increasing intestinal iron transport and raising serum iron. Transactivation of Hamp is impaired in the genetic disease hereditary hemochromatosis (HH) [3], causing low hepcidin, elevated iron absorption and pathological tissue iron overload [4]. Management of HH is principally by phlebotomy [5]; however, this treatment removes iron along with other essential nutrients and biomolecules from the body (i.e., it is not specific for iron). Moreover, some patients are averse to this clinical procedure. Development of new treatment approaches is thus warranted. Dietary interventions may also impact disease severity in HH, and patients may thus be advised to avoid foods rich in highly bioavailable iron, iron-fortified foods and dietary supplements containing extra iron. For example, the Hemochromatosis Society Netherlands, in their publication entitled, “Dietary Advice in HFE-Hemochromatosis” states that: “ ... it is likely that the combination of phlebotomy and diet modifications is the best treatment for hemochromatosis. Changes in nutrition can perhaps reduce the number of required phlebotomies. That this preventive method for iron overload gets virtually no attention is quite remarkable because–although (almost) no research is done–it is expected that bloodletting also has an impact on other health aspects, such as the vitamin status of patients”. Moreover, a recent systematic review on this topic concludes that: “Despite the limited quantitative evidence and the lack of randomized, prospec- tive trials, dietary interventions that modify iron intake and bioavailability may affect iron accumulation in HH patients. Although this measure may be welcome in patients willing to contribute to their disease management, limited data exist on the clinical and quality-of-life benefit.” [6]. Additional experimental validation of nutritional interventions that may mitigate iron loading in HH is clearly worthy of further consideration. Since absorption of dietary and supplemental iron is inappropriately elevated in HH, one logical therapeutic intervention involves targeting (and silencing) the main intestinal iron transporters, including: divalent metal-ion transporter 1 (Dmt1) (the main importer of dietary non-heme [or inorganic] iron) and 1 (Fpn1) (the main iron exporter). The activity of both transporters is inappropriately elevated in HH [7–9]. Low hepcidin increases Fpn1 protein levels on the enterocyte basolateral membrane which secondarily causes a paradoxical increase in Dmt1 protein levels on the brush-border membrane. The latter likely occurs via intracellular depletion of iron (due to high Fpn1-mediated iron export), which stabilizes the Dmt1 transcript via the IRE/IRP iron-regulatory system [10], thus increasing protein expression levels. We previously demonstrated that in vivo knock down of intestinal Dmt1 prevented iron loading in a weanling mouse model of early onset (type 2B) HH (i.e., Hamp KO mice). Here, we again focused on Dmt1, but in this case, we sought to test the hypothesis that Dmt1 silencing would mitigate iron loading once already established in older Hamp KO mice. We further postulated that Dmt1 knock down would Nutrients 2021, 13, 1686 3 of 16

be most effective when combined with dietary iron restriction. Accordingly, we used folic acid-coupled, ginger nanoparticle-derived lipid vectors (FA-GDLVs) [11] to deliver Dmt1 (and negative control) siRNAs to the intestinal epithelium of fully iron-loaded, Hamp−/− mice fed a low-iron diet. Subsequent experimental analyses assessed iron status and quantified intestinal iron absorption and tissue distribution.

2. Materials and Methods 2.1. Preparation of Folic Acid-Conjugated Ginger Nanoparticle-derived Lipid Vectors (FA-GDLVs) Ginger-derived nanoparticles were isolated according to established protocols [11–14]. Subsequently, folic acid-conjugated ginger nanoparticle-derived lipid vectors (FA-GDLVs) were fabricated using previously established methods [11]. Folic acid coupling allowed GDLV uptake in the upper small intestine [11], presumably by targeting the proton-coupled folate transporter (PCFT; also named heme-carrier protein 1 [HCP1]), which is highly expressed in the duodenum [15,16]. HPLC-purified, in vivo Dmt1-siRNA (cat. no. 4457308) or negative control siRNA (NC-siRNA) (cat. no. 4457289) (ThermoFisher Scientific) were loaded into FA-GDLVs as described previously [11]. In brief, Dmt1-siRNA or NC-siRNA (3.75 nmol) were mixed with 6 µL of Turbofect in vivo transfection reagent (cat. no. R0541) (ThermoFisher Scientific) and then added to the lipid film of FA-GDLVs under sonication.

2.2. Animal Studies All animal studies were approved by Institutional Animal Care and Use Committee (IACUC) of the University of Florida. Hepcidin KO (Hamp−/−) mice on the C57BL/6 genetic background were bred in our animal facility. Female Hamp−/− mice were weaned onto a standard chow diet and then, at 5 weeks of age, were switched to low-iron diet (2–6 ppm Fe; Envigo: TD.80396) for the remainder of the experiment (a schematic of the experimental design is provided in Figure1). These diets contain only inorganic (non-heme) iron. At 5 weeks of age, when consuming a chow diet, these mice are significantly iron loaded, with hepatic non-heme iron levels >20× higher than sex- and age-matched WT mice [11]. Dietary iron restriction was utilized for two reasons: first, we postulated that knock down of Dmt1 would likely be more effective at attenuating absorption when dietary iron was low. This was particularly relevant since the extent of in vivo Dmt1 silencing was uncertain, and modest decreases in Dmt1 activity would likely be more impactful when enteral iron was lower (since the ratio of substrate ions [Fe] to transporter molecules [Dmt1] would also be lower); and second, we sought to assess the impact of dietary iron restriction on the iron-loading in Hamp−/− mice (since this is a logical approach to reduce body iron burden in HH). One day after the dietary switch, mice were given saline (control) or freshly made NC-siRNA or Dmt1-siRNA loaded FA-GDLVs daily (3.75 nmol/dose) by oral, intragastric gavage for 21 days. On each day of the experiment, mice were fasted for three hours before receiving the gavage treatment and then given food immediately thereafter. Water was always provided ad libitum. Our previous studies demonstrated that orally administered GDLVs/siRNAs are non-toxic and non-inflammatory in mice [11,12,17]. At the end of the intervention, half of the mice were euthanized to harvest blood and tissues Nutrients 2021, 13, x FOR PEER REVIEW(liver, spleen, heart, kidney, pancreas, bone marrow and different segments of intestine),4 of 16

and the other half were used for an iron (59Fe) absorption study (as described below).

Figure 1. Overview of experimental design.

2.3. Hematologic and Iron Parameters

Mice were anesthetized by CO2 exposure and killed by cervical dislocation. Hemo- globin (Hb) and hematocrit (Hct) levels in whole blood were determined by standard methods [18]. Plasma non-heme iron levels were measured using a colorimetric method [19]. A previously described colorimetric method was used to quantify total iron-binding capacity (TIBC) [19]. saturation was calculated as (serum iron/TIBC × 100). Serum ferritin levels were assessed using the Ferritin Mouse ELISA Kit (Abcam, ab157713). Non-heme iron levels in tissues (liver, spleen, heart, kidney, and pancreas) were determined using a standard acid digestion, chromogen-based colorimetric assay as described before [19]; data were normalized by tissue weight.

2.4. Iron Absorption Study Intestinal iron absorption and body iron distribution in experimental mice were de- termined after oral administration of 59Fe, as previously described [18]. Briefly, mice were provided their final dose of saline, or FA-GDLVs/siRNA, and then three hours later, food was removed from cages (but mice were allowed free access to water during this time). The transport solution consisted of 2.5 µCi 59FeCl3 (500 µCi; 16.26 mCi/mL, Perkin Elmer) diluted into 0.2 mL of a solution containing 0.5 M ascorbic acid, 0.15 M NaCl and 5 µg of Fe from (NH4)2Fe(SO4)2. After the three-hour fast, mice were given the transport solution by oral, intragastric gavage. Mice were then immediately given free access to a standard semi-purified (control) diet (with ⁓48 ppm Fe) (Envigo; TD.80394) and euthanized 24 hours later. The control diet was utilized here as we sought to assess iron absorption when dietary intake was normal (i.e., to increase physiological relevance). The 24-h time point was consistent with our previous studies [18,20], and was chosen as a time when all en- teral iron would be absorbed (including the slower phase of iron transport that occurs 12 to ⁓20 h after dosing) [21] and before significant non-specific iron losses had occurred (e.g., via desquamation of epidermal cells, exfoliation of intestinal epithelial cells, and in urine and sweat). Whole-carcass, blood, and tissue radioactivity was measured with a WIZ- ARD2 automatic gamma counter (Perkin Elmer). Intestinal iron absorption was calculated as follows: radioactivity in the carcass minus radioactivity in the entire gut (esophagus to anus) divided by the amount of radioactivity in the oral gavage solution (×100). Blood and tissue radioactive counts were normalized by volume or weight, respectively.

2.5. MPO Assay A small piece of duodenum was freshly harvested and immediately homogenized with Myeloperoxidase (MPO) Assay buffer. MPO activity was quantified using the MPO Activity Assay Kit (Abcam; ab105136) according to the manufacturer’s instruction. Results are shown as relative MPO activity (normalized by the control group).

2.6. Western Blotting Isolated duodenal enterocytes [22] were homogenized in HEM buffer (20 mM Hepes, 1 mM EDTA, 300 mM mannitol, pH 7.4) plus protease inhibitors. After a slow-speed spin to remove undigested material, the homogenate was then centrifuged at 100,000× g for 30

Nutrients 2021, 13, 1686 4 of 16

2.3. Hematologic and Iron Parameters

Mice were anesthetized by CO2 exposure and killed by cervical dislocation. Hemoglobin (Hb) and hematocrit (Hct) levels in whole blood were determined by standard methods [18]. Plasma non-heme iron levels were measured using a colorimetric method [19]. A previously described colorimetric method was used to quantify total iron-binding capacity (TIBC) [19]. Transferrin saturation was calculated as (serum iron/TIBC × 100). Serum ferritin levels were assessed using the Ferritin Mouse ELISA Kit (Abcam, ab157713). Non-heme iron levels in tissues (liver, spleen, heart, kidney, and pancreas) were determined using a standard acid digestion, chromogen-based colorimetric assay as described before [19]; data were normalized by tissue weight.

2.4. Iron Absorption Study Intestinal iron absorption and body iron distribution in experimental mice were determined after oral administration of 59Fe, as previously described [18]. Briefly, mice were provided their final dose of saline, or FA-GDLVs/siRNA, and then three hours later, food was removed from cages (but mice were allowed free access to water during this 59 time). The transport solution consisted of 2.5 µCi FeCl3 (500 µCi; 16.26 mCi/mL, Perkin Elmer) diluted into 0.2 mL of a solution containing 0.5 M ascorbic acid, 0.15 M NaCl and 5 µg of Fe from (NH4)2Fe(SO4)2. After the three-hour fast, mice were given the transport solution by oral, intragastric gavage. Mice were then immediately given free access to a standard semi-purified (control) diet (with ~48 ppm Fe) (Envigo; TD.80394) and euthanized 24 h later. The control diet was utilized here as we sought to assess iron absorption when dietary intake was normal (i.e., to increase physiological relevance). The 24-h time point was consistent with our previous studies [18,20], and was chosen as a time when all enteral iron would be absorbed (including the slower phase of iron transport that occurs 12 to ~20 h after dosing) [21] and before significant non-specific iron losses had occurred (e.g., via desquamation of epidermal cells, exfoliation of intestinal epithelial cells, and in urine and sweat). Whole-carcass, blood, and tissue radioactivity was measured with a WIZARD2 automatic gamma counter (Perkin Elmer). Intestinal iron absorption was calculated as follows: radioactivity in the carcass minus radioactivity in the entire gut (esophagus to anus) divided by the amount of radioactivity in the oral gavage solution (×100). Blood and tissue radioactive counts were normalized by volume or weight, respectively.

2.5. MPO Assay A small piece of duodenum was freshly harvested and immediately homogenized with Myeloperoxidase (MPO) Assay buffer. MPO activity was quantified using the MPO Activity Assay Kit (Abcam; ab105136) according to the manufacturer’s instruction. Results are shown as relative MPO activity (normalized by the control group).

2.6. Western Blotting Isolated duodenal enterocytes [22] were homogenized in HEM buffer (20 mM Hepes, 1 mM EDTA, 300 mM mannitol, pH 7.4) plus protease inhibitors. After a slow-speed spin to remove undigested material, the homogenate was then centrifuged at 100,000× g for 30 min at 4 ◦C. The resulting pellet was then resuspended in HEM buffer plus protease inhibitors. Protein concentrations were quantified using the Pierce BCA protein assay kit (ThermoFisher Scientific; Waltham, MA). Thirty micrograms of protein were loaded into each lane of 8% polyacrylamide gels. Subsequent to gel running, were transferred to PVDF membranes and then blocked in Odyssey blocking buffer (Licor; Chattanooga, TN, USA). Membranes were then incubated with rabbit anti-DMT1 primary antibody (1:2000) (kindly provided by Dr. François Canonne-Hergaux; French National Institute of Health and Medical Research (INSERM), Digestive Health Research Institute (IRSD), Toulouse, France), mouse anti-FPN1 primary antibody (1:500) (kindly provided by Dr. Mitchell Knutson, University of Florida) or mouse anti-Na/K ATPase antibody (after stripping the membranes) (1:500; cat. no. sc21712; Santa Cruz Biotechnology; Dallas, TX, USA). Blots Nutrients 2021, 13, 1686 5 of 16

were then incubated with IRDye 800CW donkey anti-rabbit secondary antibody (1:10,000; cat. no. 925-32213; Licor) or IRDye 680 donkey anti-mouse secondary antibody (1:10,000; cat. no. 926-68072; Licor). Blots were subsequently imaged and protein bands were quantified using a Licor Odyssey CLx immunofluorescent instrument (model- 9141-02V). DMT1 and FPN1 immunoreactive protein band intensities were then normalized to band intensities of Na/K ATPase.

2.7. Quantitative Real-Time PCR Total RNA was isolated by RNAzol® RT reagent (Molecular Research Center, Inc.; Cincinnati, OH) following the manufacturer’s recommended protocol. A Nanodrop spec- trophotometer was used to measure RNA concentration and assess RNA integrity. SYBR- Green qRT-PCR was performed based on a well-established protocol [23,24]. The expression of experimental was normalized to expression of the housekeeping gene Cyclophilin A (CypA). Primer sequences are listed in Table1.

Table 1. qRT-PCR Primer Sequence (50 to 30)

Dmt1 GTGATCCTGACCCGGTCTATCG TGAGGATGGGTATGAGAGCAAAGG Epo ATGAAGACTTGCAGCGTGGA AGGCCCAGAGGAATCAGTAG Erfe ACTCACCAAGCAGCCAAGAA TTCTCCAGCCCCATCACAGT TNF-α CACAAGATGCTGGGACAGTGA TCCTTGATGGTGGTGCATGA IL-6 CTGCAAGAGACTTCCATCCAGTT AGGGAAGGCCGTGGTTGT CypA CTTACGACAAGCAGCCCTTCATG AGCTGTTTTTAACTCACTGCTGTTGTA

2.8. Statistical Analyses Results are depicted as box plots displaying the minimum, the lower (25th percentile), the median (50th percentile), the upper (75th percentile), and the maximum ranked sam- ple. The mean value is indicated by a ‘+’ sign. Statistical analysis was performed using GraphPad Prism (v 8.0) software. Prior to data analysis, the Brown–Forsythe test was used to confirm equal variance. Data were then analyzed by one-way ANOVA. If significant main effects were noted, then by a Tukey’s multiple comparisons test was also performed to assess differences between individual groups. p < 0.05 was considered statistically significant.

3. Results 3.1. FA-GDLVs/siRNA Exposure Had No Detrimental Effects on Experimental Mice We first sought to determine whether oral administration of FA-GDLVs caused intesti- nal inflammation or other untoward effects on experimental mice. We did not expect such negative outcomes as previous work has shown that GDLVs and FA-GDLVs are non-toxic and well tolerated by mice [25,26]. Consistent with this, in the current investigation, none of the experimental treatments altered body weights, or the relative mass of the liver, spleen, heart, or kidneys in experimental mice (Figure2). Blood hemoglobin and hematocrit levels were also not altered by the experimental treatments (Figure3). Consistent with this, biomarkers of erythropoietic demand, including bone marrow erythroferrone (Erfe) [27] and renal erythropoietin (Epo) mRNA expression, were unaffected by FA-GDLVs/siRNA exposure (Figure3). Note that the magnitude of mRNA expression of Epo and Erfe accurately reflects circulating levels of the functional hormones since both genes are regulated transcriptionally (Erfe probably by Epo/Epo receptor signaling [27], and Epo by hypoxia-inducible trans-acting factors [28]). Nutrients 2021, 13, x FOR PEER REVIEW 6 of 16

Nutrients 2021, 13, x FOR PEER REVIEW 6 of 16 Nutrients 2021, 13, 1686 6 of 16

Figure 2. Body weight and tissue weights were not affected by FA-GDLV/siRNA treatments. Body weight (A), and relative liver (B), spleen (C), heart (D), and kidney (E) weights (as a percentage of total body weight) were quantified in experimental mice. Data are presented as box plots for n = 4- 5 mice per group. No statistically significant differences were noted between groups (as determined by one-way ANOVA). NC, negative control. +, mean value.

Blood hemoglobin and hematocrit levels were also not altered by the experimental treatments (Figure 3). Consistent with this, biomarkers of erythropoietic demand, includ- ing bone marrow erythroferrone (Erfe) [27] and renal erythropoietin (Epo) mRNA expres- sion, wereFigure unaffected 2. BodyFigure weightby 2. FA-GDLVs/siRNABody and weight tissue and weights tissue weightsex wereposure were not (Figure notaffected affected 3). by by NoteFA-GDLV/siRNA FA-GDLV/siRNA that the magnitude treatments. treatments. Body Body weight (A), and relative liver (B), spleen (C), heart (D), and kidney (E) weights (as a percentage of of mRNAweight expression (A), and of relative Epo and liver Erfe (B ),accurately spleen (C), reflects heart (D circulating), and kidney levels (E) weights of the functional (as a percentage of total body weight) were quantified in experimental mice. Data are presented as box plots for n = 4–5 total body weight) were quantified in experimental mice. Data are presented as box plots for n = 4- hormones since bothmice genes per group. are No regulated statistically transcriptionally significant differences were(Erfe noted probably between groupsby Epo/Epo (as determined re- by ceptor signaling5 mice per [27], group.one-way and No ANOVA).Epo statistically by NC,hypoxia-inducible negative significant control. diffe +, meanrences trans value.-acting were noted factors between [28]). groups (as determined by one-way ANOVA). NC, negative control. +, mean value.

Blood hemoglobin and hematocrit levels were also not altered by the experimental treatments (Figure 3). Consistent with this, biomarkers of erythropoietic demand, includ- ing bone marrow erythroferrone (Erfe) [27] and renal erythropoietin (Epo) mRNA expres- sion, were unaffected by FA-GDLVs/siRNA exposure (Figure 3). Note that the magnitude of mRNA expression of Epo and Erfe accurately reflects circulating levels of the functional hormones since both genes are regulated transcriptionally (Erfe probably by Epo/Epo re- ceptor signaling [27], and Epo by hypoxia-inducible trans-acting factors [28]).

Figure 3. HematologicalFigure parameters 3. Hematological in the parameters blood and in the erythropoietic blood and erythropoietic demand demandwere not were affected not affected in in experimental mice byexperimental Fa-GDLVs/siRNA mice by Fa-GDLVs/siRNA exposure. Blood, exposure. bone Blood,marrow bone and marrow kidney and were kidney collected were collected and analyzed. Hb (Aand) and analyzed. Hct (B Hb) in (A whole) and Hct blood (B) in were whole measured. blood were measured.Erfe mRNA Erfe expression mRNA expression in bone in bone marrow (C) and Epomarrow mRNA (C )expression and Epo mRNA in kidney expression (D in) kidneywere also (D) were analyzed. also analyzed. Data Dataare arepresented presented as as box box plots for n = 4–5plots mice for pern = 4–5group. mice perNo group. statistically No statistically significant significant differences differences were were noted noted between between groups groups (as determined(as determinedby one-way by ANOVA). one-way ANOVA). NC, negative NC, negative control; control; +, +,mean mean value. value.

Figure 3. Hematological parameters in the blood and erythropoietic demand were not affected in experimental mice by Fa-GDLVs/siRNA exposure. Blood, bone marrow and kidney were collected and analyzed. Hb (A) and Hct (B) in whole blood were measured. Erfe mRNA expression in bone marrow (C) and Epo mRNA expression in kidney (D) were also analyzed. Data are presented as box plots for n = 4–5 mice per group. No statistically significant differences were noted between groups (as determined by one-way ANOVA). NC, negative control; +, mean value.

Nutrients 2021, 13, x FOR PEER REVIEW 7 of 16 Nutrients 2021, 13, 1686 7 of 16

Moreover,Moreover, myeloperoxidase myeloperoxidase (MPO) (MPO) activity activity inin duodenal duodenal epithelial epithelial tissue tissue was similarwas similar amongamong all experimental all experimental groups, groups, indicating indicating no no localized inflammatory inflammatory response response (Figure (Figure4 ). 4). SupportingSupporting this, this,hepatic hepatic IL-6 IL-6 and and TNF TNFαα mRNAmRNA expressionexpression was was not not different different between between groupsgroups (Figure (Figure 4). 4 ).

FigureFigure 4. Inflammatory 4. Inflammatory biomarkers biomarkers did did not not change change afte afterr treatments. treatments. MPO MPO activi activityty in in duodenal duodenal epi- thelial epithelialtissue was tissue measured was measured (A). IL-6 (A). mRNA IL-6 mRNA expression expression (B ()B )and and TNF- TNF-αα mRNAmRNA expression expression (C) (C) in liver werein liver also were analyzed. also analyzed. Data are Data presented are presented as box asplots box for plots n = for 4–5n mice= 4–5 per mice group. per group. No statistically No significantstatistically differences significant were differences noted between were noted grou betweenps (as groupsdetermined (as determined by one-way by one-way ANOVA). ANOVA). NC, neg- ative control;NC, negative +, mean control; value. +, mean value.

Overall, then, no pathophysiologic effects of FA-GDLVs/siRNA exposure were noted, Overall, then, no pathophysiologic effects of FA-GDLVs/siRNA exposure were as all measured outcomes did not vary from saline (control) administration. These obser- noted,vations as all measured are congruent outcomes with previous did not studies vary that from have saline exemplified (control) the administration. safe use of orally These observationsadministered are congruent GDLVs and with FA-GDLVs previous for intestinal studies siRNA that have delivery exemplified [11,12,17]. the safe use of orally administered GDLVs and FA-GDLVs for intestinal siRNA delivery [11,12,17]. 3.2. FA-GDLV-Mediated siRNA Delivery Downregulated Duodenal Dmt1 mRNA Expression 3.2. FA-GDLV-MediatedShort-interfering siRNA RNAs Delivery (siRNAs) Down are double-strandedregulated Duodenal RNA moleculesDmt1 mRNA that function Expression by RNA interference (RNAi), in which specific transcripts are targeted for degradation [29]. Short-interferingThe expected experimental RNAs (siRNAs) outcome ofare the double-stranded current study then RNA would molecules be decreased that Dmt1 function by RNAmRNA interference expression (RNAi), in tissues/cells in which targeted speci byfic FA-GDLVs transcripts when are provided targeted by oral, for intragas-degradation [29]. Thetric gavage.expected As experimental intestinal iron absorption outcome occursof the mainly current in thestudy duodenum then would [1], we be coupled decreased Dmt1 GDLVsmRNA with expression folic acid in to increasetissues/cells uptake targ ineted the proximal by FA-GDLVs small intestine when [ 11provided]. Here, this by oral, intragastricapproach gavage. seemed As to intestinal be effective, iron as Dmt1 absorption mRNA expression occurs mainly was reduced in the in duodenum the duodenal [1], we epithelium (>50%), but no change was seen in jejunum, colon, bone marrow, liver, or kidney coupled(Figure GDLVs5). Orally with administered folic acid to FA-GDLVs increase thusuptake specifically in the targetedproximal the small proximal intestine small [11]. Here, intestine,this approach and extra-intestinal seemed to effectsbe effective, were not as observed. Dmt1 mRNA These observations expression are was consistent reduced in the duodenalwith our epithelium previously published (>50%), workbut no with change FA-GDLVs was seen in mice in [jejunum,11]. colon, bone marrow, liver, or kidney (Figure 5). Orally administered FA-GDLVs thus specifically targeted the 3.3. Duodenal DMT1 and FPN1 Protein Levels Were Unaffected by FA-GDLV-siRNA Delivery proximal small intestine, and extra-intestinal effects were not observed. These observa- tions are consistentsiRNA-mediated with our suppression previously of Dmt1 published mRNA expressionwork with would FA-GDLVs be expected in mice to also [11]. decrease DMT1 protein levels, and possibly decrease intestinal iron absorption (since DMT1 is the main iron uptake mechanism in mice) [30]. We thus quantified DMT1 protein levels in experimental mice by Western blot analysis. Also, since iron transfer across the enterocyte basolateral membrane is thought to be the rate-limiting step in mucosal iron transfer [1], it was also important to quantify FPN1 protein expression levels. The DMT1 and FPN1 antibodies we used, which were provided by leading investigators in the iron biology field, have been previously validated for use in mice [31–33]. To increase specificity, duodenal enterocytes were isolated (using an established procedure) and total membrane proteins were purified and run on the separating gels. The DMT1 antibody produced single, diffuse bands in each lane of the blots, with the molecular weight of the protein band being in the 75–85 kDa range (Figure6A), consistent with previous publications [ 32]. The FPN1 antibody detected several non-specific bands (or lower m.w. degradation products) [33], as

Nutrients 2021, 13, 1686 8 of 16

well as a diffuse band at ~65–70 kDa which is the immunoreactive FPN1 protein (Figure6B) (established by using proteins isolated from Fpn1 KO mice; personal communication, Dr. Mitchell Knutson). Using this experimental approach, however, we were unable to detect differences in duodenal DMT1 or FPN1 protein levels between experimental groups of mice (Figure6). This outcome was unexpected, at least for DMT1, since duodenal mRNA expression was suppressed. High inter-animal variability was apparent, as band intensities varied more than 10-fold (i.e., relative protein expression was >100 in one animal and <10 in another). This clearly contributed to the negative outcome. Nonetheless, since immunoreactive protein on an immunoblot may or may not accurately reflect functional Nutrients 2021, 13, x FOR PEER REVIEW 8 of 16 protein levels, it was imperative to quantify intestinal iron absorption in vivo (which should mainly reflect DMT1 activity).

Figure Figure5. FA-GDLVs 5. FA-GDLVs loaded loaded with with Dmt1 Dmt1 siRNA siRNA decreased decreased Dmt1 Dmt1 mRNA mRNA expression expression in duodenum. in duodenum. −/− FemaleFemale HampHamp-/- mice weremice weaned were weaned onto onto a standard a standard chow chow di dietet andand then then switched switched to a to low-iron a low-iron diet diet 1 day before1 day before NP treatment. NP treatment. Starting Starting at at 5 weeks of of age, age, mice mice were were orally orally gavaged gavaged with saline with (control), saline (con- trol), negativenegative control-siRNA (NC-siRNA) (NC-siRNA) or DMT1-siRNA or DMT1-siRNA FA-GDLVs FA-GDLVs daily for three daily weeks. for three Dmt1 weeks. mRNA Dmt1 expression was quantified in duodenum (A), jejunum (B), colon (C), bone marrow (D), liver (E), and mRNA expression was quantified in duodenum (A), jejunum (B), colon (C), bone marrow (D), liver kidney (F). Expression of experimental genes was normalized to expression of Cyclophilin. Data are (E), and kidney (F). Expression of experimental genes was normalized to expression of Cyclophilin. presented as box plots for n = 4–5 mice per group. Statistical significance was assessed by one-way Data are presented as box plots for n = 4–5 mice per group. Statistical significance was assessed by ANOVA (A–F), followed by a Tukey’s multiple comparisons test (A,C). Groups labeled with different one-way ANOVA (A–F), followed by a Tukey’s multiple comparisons test (A, C). Groups labeled letters differ significantly. p = 0.0037 (A), p = 0.0239 (C). NC, negative control; +, mean value. with different letters differ significantly. p = 0.0037 (A), p = 0.0239 (C). NC, negative control; +, mean value.

3.3. Duodenal DMT1 and FPN1 Protein Levels Were Unaffected by FA-GDLV-siRNA Delivery siRNA-mediated suppression of Dmt1 mRNA expression would be expected to also decrease DMT1 protein levels, and possibly decrease intestinal iron absorption (since DMT1 is the main iron uptake mechanism in mice) [30]. We thus quantified DMT1 protein levels in experimental mice by Western blot analysis. Also, since iron transfer across the enterocyte basolateral membrane is thought to be the rate-limiting step in mucosal iron transfer [1], it was also important to quantify FPN1 protein expression levels. The DMT1 and FPN1 antibodies we used, which were provided by leading investigators in the iron biology field, have been previously validated for use in mice [31–33]. To increase specific- ity, duodenal enterocytes were isolated (using an established procedure) and total mem- brane proteins were purified and run on the separating gels. The DMT1 antibody pro- duced single, diffuse bands in each lane of the blots, with the molecular weight of the protein band being in the 75–85 kDa range (Figure 6A), consistent with previous publica- tions [32]. The FPN1 antibody detected several non-specific bands (or lower m.w. degra- dation products) [33], as well as a diffuse band at ⁓65–70 kDa which is the immunoreactive FPN1 protein (Figure 6B) (established by using proteins isolated from Fpn1 KO mice; per- sonal communication, Dr. Mitchell Knutson). Using this experimental approach, however, we were unable to detect differences in duodenal DMT1 or FPN1 protein levels between experimental groups of mice (Figure 6). This outcome was unexpected, at least for DMT1, since duodenal mRNA expression was suppressed. High inter-animal variability was ap- parent, as band intensities varied more than 10-fold (i.e., relative protein expression was >100 in one animal and <10 in another). This clearly contributed to the negative outcome. Nonetheless, since immunoreactive protein on an immunoblot may or may not accurately

Nutrients 2021, 13, x FOR PEER REVIEW 9 of 16

Nutrients 2021, 13, 1686 9 of 16 reflect functional protein levels, it was imperative to quantify intestinal iron absorption in vivo (which should mainly reflect DMT1 activity).

Figure 6.6.DMT1 DMT1 and and FPN1 FPN1 protein protein expression expression in the in duodenal the duodenal epithelium epithelium was unaffected was unaffected by experi- by exper- mentalimental treatments. treatments. Total Total membrane membrane proteins proteins were were isolated isolated from duodenalfrom duodenal enterocytes, enterocytes, and equal and equal quantities were were run run on on gels gels and and transferred transferred to membranes. to membranes. Anti-DMT1 Anti-DMT1 and -FPN1 and antibodies-FPN1 antibodies were were used toto detect detect proteins proteins on on the the membranes. membranes. A Na-K A Na-K ATPase ATPase antibody antibody was used was as used a control. as a control. DMT1 DMT1 ((AA)) andand FPN1 FPN1 ( B()B protein) protein levels levels were were analyzed analyzed and and normalized normalized by Na-K by ATPaseNa-K ATPase protein protein levels. levels. Quantitative data data are are mean mean± SD± SD for forn = n 3 = mice 3 mice per groupper group and wereand were analyzed analyzed by one-way by one-way ANOVA ANOVA (no(no statisticalstatistical differences differences were were noted). noted). The The asterisk asterisk demarcates demarcates the full the length full length immunoreactive immunoreactive Fpn1 Fpn1 band ((BB).). To To aide aide in in interpretation, interpretation the, entirethe entire blots blots are shown are shown (A,B). ( NC,A, B negative). NC, negative control. control.

3.4. GDLV/siRNAGDLV/siRNA Exposure Exposure Decreased Decreased Liver Liver and Pancreasand Pancreas Non-heme Non-heme Iron Content Iron Content InIn this this investigation, investigation, our our experimental experimental approach approach was towas assess to assess the utility the ofutility dietary of dietary ironiron restrictionrestriction combined combined with with attenuation attenuation of of intestinal intestinal DMT1 DMT1 iron iron import import activity activity for for mit- mitigating iron loading in a murine model of severe HH. To exemplify how the low-iron igating iron loading in a murine model of severe HH. To exemplify how the low-iron diet diet impacted body iron burden, we compared the current data to our previously published dataimpacted on 7–9-week-old body ironfemale burden,Hamp weKO compared mice (and the littermate current controls) data to fedour a previously chow diet with published

~200data ppmon 7–9-week-old Fe [11]. In the currentfemale study,Hamp dietary KO mice iron(and restriction littermate reduced controls) liver iron fed levels a chow from diet with >1250⁓200 ppmµg/g Fe (in [11]. chow In fed theHamp currentKO mice) study, [11 dietar] to ~150y ironµg/g. restriction With concurrent reduced knockdown liver iron levels offrom intestinal >1250 Dmt1, µg/g liver(in chow iron content fed Hamp was KO reduced mice) to 25–30[11] toµ g/g~150 (Figure µg/g.7 WithA), which concurrent is close knock- todown the ~40 of intestinalµg/g which Dmt1, was previouslyliver iron documentedcontent was in reduced age- and to sex-matched 25–30 µg/g WT (Figure mice [7A),11]. which Inis close pancreas, to the iron ~40 restriction µg/g which reduced was non-heme previously iron documented levels by ~2.5-fold in age- (from and ~300sex-matchedµg/g WT tomice 125 [11].µg/g), In andpancreas, FA-GDLV iron (plus restriction either siRNA)reduced exposure non-heme further iron decreasedlevels by levels~2.5-fold to (from µ µ ~35~300 g/g,µg/g which to 125 is closerµg/g), to and WT FA-GDLV values of ~10 (plusg/g either (Figure siRNA)7B). In heartexposure (Figure further7C), low- decreased iron intake reduced non-heme iron levels from ~110 µg/g (in chow fed Hamp KO mice) [11] levels to ~35 µg/g, which is closer to WT values of ~10 µg/g (Figure 7B). In heart (Figure to ~40 µg/g, which was closer to WT values (i.e., ~25 µg/g), with FA-GDLV/siRNA exposure7C), low-iron having intake no additional reduced non-heme effect. Moreover, iron levels dietary from iron ~110 restriction µg/g (in reduced chow fed renal Hamp KO non-hememice) [11] iron to levels~40 µg/g, from ~125whichµg/g was (in HampcloserKO to mice) WT tovalues values (i.e., typically ~25 seenµg/g), in age- with FA- andGDLV/siRNA sex-matched exposure WT mice (i.e.,having 20–25 noµ additionalg/g) (Figure ef7D)fect. [ 11 Moreover,]. Renal non-heme dietary ironiron content restriction re- wasduced not renal further non-heme reduced byiron experimental levels from interventions.~125 µg/g (in Finally,Hamp KO spleen mice) non-heme to values iron typically contentseen in wasage- uniformly and sex-matched low in all WT mice mice (20–25 (i.e.,µg/g), 20–25 with µg/g) FA-GDLV (Figure exposure7D) [11].having Renal non-heme no furtheriron content effects. was These not values further are reduced ~50% lower by thanexpe inrimental age- and interventions. sex-matched mice Finally, from spleen our non- previousheme iron study content (i.e., 50–60was uniformlyµg/g) [11]. Spleenlow in ironall mice depletion (20–25 has µg/g), been previously with FA-GDLV described exposure −/− inhavingHamp no furthermice, whicheffects. is These presumably values dueare ~ to50% high lower Fpn1-mediated than in age- iron and export sex-matched from mice splenic macrophages (which store and recycle iron obtained by erythrophagocytosis) [34]. from our previous study (i.e., 50–60 µg/g) [11]. Spleen iron depletion has been previously described in Hamp-/- mice, which is presumably due to high Fpn1-mediated iron export from splenic macrophages (which store and recycle iron obtained by erythrophagocyto- sis) [34].

Nutrients 2021, 13, x FOR PEER REVIEW 10 of 16

In sum, dietary iron restriction reduced hepatic non-heme iron levels in Hamp-/- mice, but concurrent knockdown of Dmt1 was necessary to bring values into the same range as in WT controls. In other tissues, low iron intake reduced tissue iron levels closer to WT levels, with FA-GDLV administration potentiating the effect of the low-iron diet only in pancreas (but not in heart or kidneys). Silencing intestinal Dmt1 is thus effective at reduc- ing hepatic iron stores, while FA-GDLV/NC siRNA exposure reduced pancreatic iron loading. The latter observation suggested that ginger contains bioactive lipids [35] that influence iron absorption and homeostasis, since it is highly unlikely that the effects were due to the NC siRNA (which, according to the manufacturer, has no significant sequence similarity to mouse, rat, or human gene sequences, and has been shown to have minimal influences on global as determined by microarray analysis). This outcome Nutrients 2021was, 13 ,unanticipated, 1686 as FA-GDLV/NC siRNA administration was without effect in our pre-10 of 16 vious study [11].

Figure 7. FA-GDLVs/siRNAFigure 7. FA-GDLVs/siRNA administration decreased administration liv decreaseder and pancreatic liver and pancreaticiron levels. iron Shown levels. Shownis is non-heme Fe contentnon-heme in liver Fe (A content), pancreas in liver (B (A),), heart pancreas (C), (B and), heart kidney (C), and (D). kidney Data ( Dwere). Data analyzed were analyzed by by one-way ANOVA (A–D), followed by Tukey’s multiple comparisons test (A,B). Data are presented one-way ANOVA (A–D), followed by Tukey’s multiple comparisons test (A, B). Data are pre- as box plots for n = 4–5 mice per group. Groups labeled with different letters differ significantly. sented as box plots for n = 4–5 mice per group. Groups labeled with different letters differ signifi- p = 0.0048 (A), p = 0.0264 (B). NC, negative control; +, mean value. cantly. p = 0.0048 (A), p = 0.0264 (B). NC, negative control; +, mean value. In sum, dietary iron restriction reduced hepatic non-heme iron levels in Hamp−/− 3.5. Dmt1 Knock Downmice, Reduced but concurrent Parenchymal knockdown Iron of Loading Dmt1 was and necessary Serum Non-heme to bring values Iron into Levels the same range as in WT controls. In other tissues, low iron intake reduced tissue iron levels closer Additional experimentsto WT levels, withwere FA-GDLV carried administrationout to quantify potentiating biomarkers the effect of iron of the status, low-iron in- diet cluding serum ferritinonly in and pancreas non-heme (but not iron, in heart TIBC or kidneys). and blood Silencing transferrin intestinal saturation Dmt1 is thus (TSAT). effective at In the absence of inflammationreducing hepatic ironor malignant stores, while disease, FA-GDLV/NC serum siRNA ferritin exposure is a reliable reduced pancreaticindicator iron of body iron storesloading. [36]. TheMoreover, latter observation increases suggested in serum that non-heme ginger contains iron, bioactive and consequent lipids [35] that influence iron absorption and homeostasis, since it is highly unlikely that the effects were increases in transferrindue to thesaturation, NC siRNA typify (which, iron-loading according to the disorders manufacturer, like hasHH. no Successful significant sequence mit- igation of iron loadingsimilarity would to mouse, thus rat, best or human be exemplified gene sequences, by reductions and has been in shown serum to have ferritin, minimal serum non-heme influencesiron, and on TSAT. global In gene our expression previous as determinedstudy [11], by serum microarray ferritin analysis). was reported This outcome to be ~1000 ng/mLwas inunanticipated, 7-week-old female as FA-GDLV/NC Hamp-/- mice. siRNA Here, administration control was(i.e., without saline effectgavage) in our previous study [11]. mice had similar serum ferritin levels of ~1000 ng/ml, indicating that dietary iron re- striction did not decrease3.5. Dmt1 Knockthese Down levels. Reduced FA-GDLVs/NC Parenchymal Iron siRNA Loading treatment and Serum Non-heme reduced Iron serum Levels ferritin by ~20%, whileAdditional FA-GDLVs/Dmt1 experiments siRNA were carried exposure out to further quantify reduced biomarkers it (by of iron ~50%) status, to in- just over 500 ng/mlcluding (although serum the ferritin difference and non-heme between iron, the TIBC treatment and blood transferringroups was saturation not statis- (TSAT). tically different) (FigureIn the absence 8A). ofIn inflammation these mice or(with malignant no inflammation), disease, serum ferritin a 50% is areduction reliable indicator in of body iron stores [36]. Moreover, increases in serum non-heme iron, and consequent increases in transferrin saturation, typify iron-loading disorders like HH. Successful mit- igation of iron loading would thus best be exemplified by reductions in serum ferritin, serum non-heme iron, and TSAT. In our previous study [11], serum ferritin was reported to be ~1000 ng/mL in 7-week-old female Hamp−/− mice. Here, control (i.e., saline gav- age) mice had similar serum ferritin levels of ~1000 ng/mL, indicating that dietary iron restriction did not decrease these levels. FA-GDLVs/NC siRNA treatment reduced serum Nutrients 2021, 13, 1686 11 of 16

Nutrients 2021, 13, x FOR PEER REVIEW 11 of 16

ferritin by ~20%, while FA-GDLVs/Dmt1 siRNA exposure further reduced it (by ~50%) to just over 500 ng/mL (although the difference between the treatment groups was not serumstatistically ferritin should different) reflect (Figure a reduction8A). In these in micebody (with iron nocontent inflammation), of a similar a 50% magnitude. reduction Se- rum innon-heme serum ferritin iron was should also reflect reduced a reduction by low-iron in body feeding iron content (from of ~310 a similar µg/dL magnitude. in chow fed HampSerum-/- mice non-heme [11] to ~220 iron wasµg/dL) also (Figure reduced 8B), by low-iron with Dmt1 feeding siRNA (from administration ~310 µg/dL in chow further fed re- Hamp−/− µ ducing it to ~60mice µg/dL. [11] to TIBC ~220 wasg/dL) not (Figure different8B), withamong Dmt1 experimental siRNA administration groups. Moreover, further reducing it to ~60 µg/dL. TIBC was not different among experimental groups. Moreover, TSAT trended lower in Dmt1 siRNA-exposed mice, but statistical significance was not TSAT trended lower in Dmt1 siRNA-exposed mice, but statistical significance was not achievedachieved due due to tohigh high inter-animal inter-animal variability;variability; in fact,in fact, TSAT TSAT values values varied 50–100-foldvaried 50– (from100-fold (from100% 100% to to 1–2%) 1–2%) (Figure (Figure8C). Nonetheless,8C). Nonetheless, these data these demonstrate data demonstrate that GDLVs/Dmt1 that GDLVs/Dmt1 siRNA siRNAtreatment treatment decreased decreased body body iron burden iron burden in Hamp in− /Hamp− mice.-/- mice.

FigureFigure 8. FA-GDLVs/DMT1 8. FA-GDLVs/DMT1 siRNA siRNA treatment treatment decreased decreased wholewhole body body and and serum serum iron iron content. content. Shown Shownare are serum serum ferritin ferritin levels levels (A), serum (A), serum non-heme non-heme Fe content Fe (B content), and blood (B), TSATand blood (C). Data TSAT were (C analyzed). Data were byanalyzed one-way by ANOVA one-way (A– ANOVAC), followed (A by–C Tukey’s), followed multiple by Tukey’s comparisons multiple test (A comparisons,B). Data are presented test (A, B). Data areas box presented plots for asn =box 4–5 plots mice for per ngroup. = 4–5 mice Groups per labeled group. with Groups different labeled letters with differ different significantly. letters differp significantly.= 0.0433 (A), p p= = 0.0036 0.0433 (B ().A NC,), p = negative 0.0036 ( control;B). NC, +, negative mean value. control; +, mean value. 3.6. FA-GDLVs/Dmt1 siRNA Administration Blunted Intestinal Iron (59Fe) Absorption and 3.6. FA-GDLVs/Dmt1Altered Tissue Iron DistributionsiRNA Administration Blunted Intestinal Iron (59Fe) Absorption and Altered TissueSo far, Iron we Distribution have demonstrated that FA-GDLV/Dmt1 siRNA treatment suppressed SoDmt1 far, mRNA we have expression demonstrated in duodenum that (byFA-GDLV/Dmt1 ~50%) and decreased siRNA non-heme treatment iron suppressed levels in Dmt1serum mRNA (by expression ~50%) and liverin duodenum (by ~85%). (by Serum ~50%) ferritin and and decreased pancreatic non-heme non-heme iron iron levels were in reduced (by ~50%) in mice receiving FA-GDLVs carrying either siRNA (i.e., NC or Dmt1). serum (by ~50%) and liver (by ~85%). Serum ferritin and pancreatic non-heme iron were Given these observations, the next obvious step was to assess intestinal iron absorption. reducedA well-established, (by ~50%) in mice oral receiving gavage, radiotracer FA-GDLVs approach carrying was either thus siRNA utilized. (i.e.,Hamp NCKO or mice Dmt1). Givenhave these elevated observations, intestinal the iron next absorption obvious [37 ],step presumably was to assess due to intestinal high expression/activity iron absorption. A well-established,of the intestinal ironoral transportersgavage, radi DMT1otracer and approach FPN1. Consistent was thus with utilized. this, controlHamp KOHamp mice have KOelevated mice in intestinal the current iron investigation absorption absorbed [37], presumably >45% of the due radioactive to high ironexpression/activity dose, indica- of thetive intestinal of elevated iron iron transporters absorption DMT1 (since and WT FPN1. mice typically Consistent absorb with 10–15% this, control [or less] Hamp of the KO mice dose)in the [ 38current]. Iron absorptioninvestigation decreased absorbed by ~10% >45% in of mice the treatedradioactive with FA-GDLV/NCiron dose, indicative siRNA of 59 elevatedand iron ~20% absorption in mice exposed (since to WT FA-GDLV/Dmt1 mice typically siRNA absorb (Figure 10–15%9A). [or less]Fe activity of the dose) in liver [38]. was also reduced in both FA-GDLV treatment groups (NC, ~40%; Dmt1, ~70%) (Figure9B) . Iron absorption decreased by ~10% in mice treated with FA-GDLV/NC siRNA and ~20% Surprisingly, FA-GDLV/Dmt1 siRNA treatment increased 59Fe activity in heart, muscle, 59 in miceand exposed bone, while to FA-GDLV/Dmt1 both experimental siRNA treatments (Figure increased 9A).59 FeFe activity activity in in blood liver and was kidney also re- duced(Figure in both9C–G) FA-GDLV. 59Fe activity treatment in spleen groups and (NC, pancreas ~40%; was Dmt1, not ~70%) influenced (Figure by any9B). of Surpris- the ingly,treatments FA-GDLV/Dmt1(Figure9H, siRNA I) . These treatment results increased demonstrated 59Fe specific activity effects in heart, of Dmt1 muscle, siRNA and and bone, whilealso both direct experimental effects of FA-GDLVs, treatments again increased suggesting 59Fe activity that ginger in blood contains and bioactive kidney lipids (Figure 9C–G).that 59Fe can activity influence in iron spleen homeostasis and pancreas (likely bywas modulating not influenced the magnitude by any of of the intestinal treatments iron (Figureabsorption). 9H, I). These results demonstrated specific effects of Dmt1 siRNA and also direct effects of FA-GDLVs, again suggesting that ginger contains bioactive lipids that can influ- ence iron homeostasis (likely by modulating the magnitude of intestinal iron absorption).

Nutrients 2021Nutrients, 13, x2021 FOR, 13 PEER, 1686 REVIEW 12 of 1612 of 16

−/− 59 FigureFigure 9. FA-GDLVs 9. FA-GDLVs loaded loaded with with DMT1 DMT1 siRN siRNAA decreased decreased ironiron absorption absorption in Hampin Hamp-/-mice. mice. 59FeFe ab- 59 sorptionabsorption (A), and (A ),59Fe and radioactivityFe radioactivity in liver in liver (B), (B blood), blood (C (C),), kidney kidney (D(D),), heart heart (E ),(E muscle), muscle (F), bone(F), bone (G), spleen(G), spleen (H), (andH), andpancreas pancreas (I) (Iwas) was measured. measured. Data Data were analyzedanalyzed by by one-way one-way ANOVA ANOVA (A–I ),(A–I), followedfollowed by Tukey’s by Tukey’s multiple multiple comparisons comparisons test test ( (AA––G). DataData are are presented presented as boxas box plots plots for n for= 4–5 n = 4–5 mice permice group. per group. Groups Groups labeled labeled with with different different letters letters differ significantly. significantly.p = p 0.0276 = 0.0276 (A), (pA=), 0.0161p = 0.0161 (B), p (=B ),0.0064p = 0.0064 (C), (pC ),= p0.0003= 0.0003 (D (),D ),p p= =0.0103 0.0103 ( E), pp= = 0.00410.0041 (F (),Fp),= p 0.0089= 0.0089 (G). ( NC,G). negativeNC, negative control; control; +, +, meanmean value. value. 4. Discussion 4. DiscussionInappropriately elevated intestinal iron absorption, due to impaired hepatic Hamp Inappropriatelyexpression, underlies elevated iron loading intestinal in HH. iron Blocking absorption, iron absorption due tois impaired thus a plausible hepatic ther- Hamp expression,apeutic underlies intervention. iron Targeting loading intestinal in HH. Dmt1 Bloc hasking been iron previously absorption considered, is thus asa smallplausible therapeuticmolecule intervention. inhibitors have Targeting been identified intestinal [39–44 Dmt1]. To our has knowledge, been previously however, considered, demonstra- as tion of the effectiveness of Dmt1 blockers in vivo in models of iron-related disease has not small molecule inhibitors have been identified [39–44]. To our knowledge, however, been reported to date. We previously demonstrated that FA-GDLV/Dmt1 siRNA treatment demonstrationprevented ironof the loading effectiveness in weanling of Dmt1Hamp− blockers/− mice [ 11in], vivo providing in models proof-of of iron-related concept that dis- ease hasDmt1 not knock been down reported can blunt to irondate. absorption We previously and alter bodydemonstrated iron levels. that Here, FA-GDLV/Dmt1 we sought to siRNAextend treatment this previous prevented investigation iron loading to include in weanling the more relevant Hamp-/- pathophysiological mice [11], providing state ofproof- of conceptpre-existing that Dmt1 iron overload. knock down can blunt iron absorption and alter body iron levels. Here, we Wesought previously to extend established this previous that GDLVs investigation [25,26] and FA-GDLVs to include are the non-toxic more torelevant cells and path- ophysiologicalnon-inflammatory state of [11 pre-existing]. Assays used iron to establish overload. this included biocompatibility assessments using an MTT cell viability assay in mouse colon-26 cells and electrical cell-substrate We previously established that GDLVs [25,26] and FA-GDLVs are non-toxic to cells impedance sensing (which measures real-time barrier function) in Caco2-BBE cells. It and non-inflammatorywas also previously established[11]. Assays that used FA-GDLVs to establish do not escapethis included the gut whenbiocompatibility provided by as- sessmentsoral gavage, using asan demonstratedMTT cell viability by using assay a co-administered in mouse colon-26 fluorescent cells dye and and electrical an IVIS cell- substratein vivo impedanceimaging system sensing [11 ].(which Robust measures fluorescence real-time was detected barrier in thefunction) GI tract, in but Caco2-BBE none cells. It was also previously established that FA-GDLVs do not escape the gut when pro- vided by oral gavage, as demonstrated by using a co-administered fluorescent dye and an IVIS in vivo imaging system [11]. Robust fluorescence was detected in the GI tract, but none was observed in kidney, spleen, lung, liver, or heart. Finally, there was no increase in myeloperoxidase (MPO) activity in duodenal tissue extracted from experimental mice treated with FA-GLDVs, indicating a lack of an inflammatory response [45]. Consistent

Nutrients 2021, 13, 1686 13 of 16

was observed in kidney, spleen, lung, liver, or heart. Finally, there was no increase in myeloperoxidase (MPO) activity in duodenal tissue extracted from experimental mice treated with FA-GLDVs, indicating a lack of an inflammatory response [45]. Consistent with lack of toxicity, in the current study, body and relative organ weights, myeloperoxidase activity in duodenum, and hepatic IL-6 and TNFα mRNA expression were unaffected by experimental treatments. Blood hemoglobin and hematocrit levels and biomarkers of erythroid demand (i.e., bone marrow erythroferrone and renal erythropoietin mRNA expression) were also unaltered, indicating that Dmt1 siRNA treatment did not cause iron deficiency or decrease iron delivery to the erythron. As part of this investigation, iron absorption was quantified using a well-established experimental approach. Interpretation of 59Fe radiotracer absorption studies can be chal- lenging, since this method offers a ‘snapshot’ of where dietary iron is being distributed during a single 24-h period (and may not reflect total iron accumulation in blood and tissues over a longer period). Since percent iron absorption is calculated from whole body 59Fe activity (minus 59Fe in the GI tract), reductions in absorption are reflected by decreases in whole-body 59Fe accumulation. Mice treated with FA-GDLVs/Dmt1 siRNA assimilated less 59Fe from the gavage dose than controls, indicating that vectorial iron transport was reduced. Consistent with this, 59Fe activity in the liver was decreased; however, 59Fe levels were paradoxically elevated in blood and several tissues tested. The significance of this latter observation is unclear, but nonetheless, iron transfer from the gut to the body was inhibited by FA-GDLV and Dmt1 siRNA treatment. Our experimental approach here utilized dietary iron restriction (which could improve iron status in HH patients) [6], plus suppression of duodenal Dmt1 expression. We initiated our studies in 5-week-old female Hamp−/− mice that were fully iron loaded (i.e., hepatic iron levels are >25-fold higher than in age- and sex-matched WT mice) [11]. Dietary iron restriction reduced body iron levels, with Dmt1 siRNA administration potentiating iron losses. FA-GDLV/Dmt1 siRNA treatment reduced Dmt1 mRNA expression in duodenum (by >2-fold) and decreased intestinal iron absorption by ~20% while 59Fe activity in liver was reduced by ~3-fold. FA-GDLVs carrying the NC siRNA also influenced many of the iron-related parameters reported here (although typically, the magnitude was less than the effects when Dmt1 siRNA was included). Mitigation of iron loading in Hamp−/− mice was thus most likely due to suppression of duodenal Dmt1 iron import activity and a concomitant reduction in iron transfer to the portal blood circulation. We say this is most likely based upon two facts: (1) DMT1 is the predominant (and probably the only) transporter of enteral non-heme iron in mice (which is the only form of iron present in mouse chow). This is nicely exemplified by the phenotype of the intestine specific Dmt1 KO [30]. These mice are severely anemic and die prematurely at about 6 months of age due to severe iron depletion (i.e., blood hemoglobin levels are lower than most other rodent models of iron deficiency [<5 g/dL; normal is 14–15 g/dL]). It was proposed that the only reason these mice live for ~6 months is due to hepatic iron stores present at birth, and/or minimal absorption of dietary iron due to mosaicism of the Dmt1 KO (as has been documented when using the villin-CRE conditional KO system); and (2) siRNA delivery via oral administration of GDLVs has been shown to specifically target the intestinal epithelium, with no escape of the nanoparticles from the gut [12]. Additionally, in the current investigation, iron absorption may also have reduced by unidentified bioactive components contained within the FA-GDLVs (which may or may not have directly influenced DMT1-mediated iron import).

5. Conclusions This investigation has demonstrated that intestinal Dmt1 is a valid therapeutic target to mitigate iron loading once already established in a murine model of early-onset, severe HH. Attenuation of duodenal Dmt1 expression and inhibition of vectorial iron transfer across the intestinal mucosa potentiated the iron-lowering effect of dietary iron restriction. Unique chemical properties of FA-GDLVs may have also contributed to the positive outcomes of Nutrients 2021, 13, 1686 14 of 16

this investigation; however, this is currently not clear and requires further investigation. This combinatorial approach of dietary iron restriction and inhibition of intestinal iron absorption could be further developed and utilized as an adjunctive treatment in HH (or other genetic iron-loading disorders), possibly reducing reliance on phlebotomy.

Author Contributions: X.W., M.Z., R.R.W., Y.Y., J.K.L. and S.R.L.F. designed and performed exper- iments. X.W., M.Z., D.M., and J.F.C. conceptualized the study and interpreted data. X.W. drafted the manuscript, performed statistical analyses, and made the figures. X.W. and J.F.C. edited the manuscript. J.F.C. and D.M. provided funding for this investigation. All authors have read and agreed to the published version of the manuscript. Funding: This investigation was funded by grant DK074867 from the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (J.F.C.), and grant DK109717 from NIDDK and the Office of Dietary Supplements (J.F.C.). D.M. is a recipient of a Senior Research Career Scientist Award from the Department of Veterans Affairs (BX004476). X.W. is supported by the National Natural Science Foundation of China (NSFC 32001676). Institutional Review Board Statement: Not applicable. Informed Consent Statement: Not applicable. Conflicts of Interest: The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results. Institutional Animal Care and Use (IACUC) Statement: All animal experiments were approved the University of Florida IACUC. The ethic approval code is 201909452.

References 1. Gulec, S.; Anderson, G.J.; Collins, J.F. Mechanistic and regulatory aspects of intestinal iron absorption. Am. J. Physiol. Gastrointest. Liver Physiol. 2014, 307, G397–G409. [CrossRef][PubMed] 2. Nemeth, E.; Tuttle, M.S.; Powelson, J.; Vaughn, M.B.; Donovan, A.; Ward, D.M.; Ganz, T.; Kaplan, J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004, 306, 2090–2093. [CrossRef] 3. Brissot, P.; Pietrangelo, A.; Adams, P.C.; De Graaff, B.; McLaren, C.E.; Loreal, O. Haemochromatosis. Nat. Rev. Dis. Prim. 2018, 4, 18016. [CrossRef][PubMed] 4. Brissot, P.; Troadec, M.B.; Loreal, O. Intestinal absorption of iron in HFE-1 hemochromatosis: Local or systemic process? J. Hepatol. 2004, 40, 702–709. [CrossRef][PubMed] 5. Kowdley, K.V.; Brown, K.E.; Ahn, J.; Sundaram, V. ACG Clinical guideline: Hereditary hemochromatosis. Am. J. Gastroenterol. 2019, 114, 1202–1218. [CrossRef] 6. Moretti, D.; Van Doorn, G.M.; Swinkels, D.W.; Melse-Boonstra, A. Relevance of dietary iron intake and bioavailability in the management of HFE hemochromatosis: A systematic review. Am. J. Clin. Nutr. 2013, 98, 468–479. [CrossRef][PubMed] 7. Griffiths, W.J.; Sly, W.S.; Cox, T.M. Intestinal iron uptake determined by divalent metal transporter is enhanced in HFE-deficient mice with hemochromatosis. Gastroenterology 2001, 120, 1420–1429. [CrossRef][PubMed] 8. Fleming, R.E.; Migas, M.C.; Zhou, X.; Jiang, J.; Britton, R.S.; Brunt, E.M.; Tomatsu, S.; Waheed, A.; Bacon, B.R.; Sly, W.S. Mechanism of increased iron absorption in murine model of hereditary hemochromatosis: Increased duodenal expression of the iron transporter DMT1. Proc. Natl. Acad. Sci. USA 1999, 96, 3143–3148. [CrossRef] 9. Stuart, K.A.; Anderson, G.J.; Frazer, D.M.; Powell, L.W.; McCullen, M.; Fletcher, L.M.; Crawford, D.H. Duodenal expression of iron transport molecules in untreated haemochromatosis subjects. Gut 2003, 52, 953–959. [CrossRef][PubMed] 10. Hubert, N.; Hentze, M.W. Previously uncharacterized isoforms of divalent metal transporter (DMT)-1: Implications for regulation and cellular function. Proc. Natl. Acad. Sci. USA 2002, 99, 12345–12350. [CrossRef] 11. Wang, X.; Zhang, M.; Flores, S.R.L.; Woloshun, R.R.; Yang, C.; Yin, L.; Xiang, P.; Xu, X.; Garrick, M.D.; Vidyasagar, S.; et al. Oral gavage of ginger nanoparticle-derived lipid vectors carrying Dmt1 siRNA blunts iron loading in murine hereditary hemochromatosis. Mol. Ther. 2019, 27, 493–506. [CrossRef][PubMed] 12. Zhang, M.; Wang, X.; Han, M.K.; Collins, J.F.; Merlin, D. Oral administration of ginger-derived nanolipids loaded with siRNA as a novel approach for efficient siRNA drug delivery to treat ulcerative colitis. Nanomedicine 2017, 12, 1927–1943. [CrossRef] [PubMed] 13. Sung, J.; Yang, C.; Collins, J.F.; Merlin, D. Preparation and characterization of ginger lipid-derived nanoparticles for colon-targeted siRNA delivery. Bio-Protocol 2020, 10, e3685. [CrossRef][PubMed] 14. Sung, J.; Yang, C.; Viennois, E.; Zhang, M.; Merlin, D. Isolation, purification, and characterization of ginger-derived nanoparticles (GDNPs) from ginger, rhizome of zingiber officinale. Bio-Protocol 2019, 9, e3390. [CrossRef][PubMed] Nutrients 2021, 13, 1686 15 of 16

15. Shayeghi, M.; Latunde-Dada, G.O.; Oakhill, J.S.; Laftah, A.H.; Takeuchi, K.; Halliday, N.; Khan, Y.; Warley, A.; McCann, F.E.; Hider, R.C.; et al. Identification of an intestinal heme transporter. Cell 2005, 122, 789–801. [CrossRef][PubMed] 16. Qiu, A.; Jansen, M.; Sakaris, A.; Min, S.H.; Chattopadhyay, S.; Tsai, E.; Sandoval, C.; Zhao, R.; Akabas, M.H.; Goldman, I.D. Identification of an intestinal folate transporter and the molecular basis for hereditary folate malabsorption. Cell 2006, 127, 917–928. [CrossRef][PubMed] 17. Zhang, M.; Collins, J.F.; Merlin, D. Do ginger-derived nanoparticles represent an attractive treatment strategy for inflammatory bowel diseases? Nanomedicine 2016, 11, 3035–3037. [CrossRef] 18. Doguer, C.; Ha, J.H.; Gulec, S.; Vulpe, C.D.; Anderson, G.J.; Collins, J.F. Intestinal potentiates iron absorption in weanling, adult, and pregnant mice under physiological conditions. Blood Adv. 2017, 1, 1335–1346. [CrossRef] 19. Ha, J.H.; Doguer, C.; Collins, J.F. Consumption of a high-iron diet disrupts homeostatic regulation of intestinal copper absorption in adolescent mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 313, G535–G360. [CrossRef][PubMed] 20. Gulec, S.; Collins, J.F. Investigation of iron in mice expressing a mutant Menke’s copper transporting ATPase (Atp7a) protein with diminished activity (Brindled; Mo (Br) (/y)). PLoS ONE 2013, 8, e66010. [CrossRef][PubMed] 21. Wheby, M.S.; Jones, L.G.; Crosby, W.H. Studies on iron absorption. Intestinal regulatory mechanisms. J. Clin. Investig. 1964, 43, 1433–1442. [CrossRef] 22. Jiang, L.; Ranganathan, P.; Lu, Y.; Kim, C.; Collins, J.F. Exploration of the copper-related compensatory response in the Belgrade rat model of genetic iron deficiency. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 301, G877–G886. [CrossRef][PubMed] 23. Gulec, S.; Collins, J.F. Silencing the Menkes copper-transporting ATPase (Atp7a) gene in rat intestinal epithelial (IEC-6) cells increases iron flux via transcriptional induction of ferroportin 1 (Fpn1). J. Nutr. 2014, 144, 12–19. [CrossRef][PubMed] 24. Xie, L.; Collins, J.F. Transcription factors Sp1 and Hif2alpha mediate induction of the copper-transporting ATPase (Atp7a) gene in intestinal epithelial cells during hypoxia. J. Biol. Chem. 2013, 288, 23943–23952. [CrossRef] 25. Zhang, M.; Viennois, E.; Prasad, M.; Zhang, Y.; Wang, L.; Zhang, Z.; Han, M.K.; Xiao, B.; Xu, C.; Srinivasan, S.; et al. Edible ginger-derived nanoparticles: A novel therapeutic approach for the prevention and treatment of inflammatory bowel disease and colitis-associated cancer. Biomaterials 2016, 101, 321–340. [CrossRef] 26. Zhang, M.; Xiao, B.; Wang, H.; Han, M.K.; Zhang, Z.; Viennois, E.; Xu, C.; Merlin, D. Edible ginger-derived nano-lipids loaded with doxorubicin as a novel drug-delivery approach for colon cancer therapy. Mol. Ther. 2016, 24, 1783–1796. [CrossRef][PubMed] 27. Kautz, L.; Jung, G.; Valore, E.V.; Rivella, S.; Nemeth, E.; Ganz, T. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat. Genet. 2014, 46, 678–684. [CrossRef][PubMed] 28. Semenza, G.L. Hypoxia-inducible factors in physiology and medicine. Cell 2012, 148, 399–408. [CrossRef][PubMed] 29. Carthew, R.W.; Sontheimer, E.J. Origins and mechanisms of miRNAs and siRNAs. Cell 2009, 136, 642–655. [CrossRef] 30. Shawki, A.; Anthony, S.R.; Nose, Y.; Engevik, M.A.; Niespodzany, E.J.; Barrientos, T.; Ohrvik, H.; Worrell, R.T.; Thiele, D.J.; Mackenzie, B. Intestinal DMT1 is critical for iron absorption in the mouse but is not required for the absorption of copper or manganese. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 309, G635–G647. [CrossRef] 31. Canonne-Hergaux, F.; Gruenheid, S.; Ponka, P.; Gros, P. Cellular and subcellular localization of the Nramp2 iron transporter in the intestinal brush border and regulation by dietary iron. Blood 1999, 93, 4406–4417. [CrossRef][PubMed] 32. Gruenheid, S.; Canonne-Hergaux, F.; Gauthier, S.; Hackam, D.J.; Grinstein, S.; Gros, P. The iron transport protein NRAMP2 is an integral membrane glycoprotein that colocalizes with transferrin in recycling endosomes. J. Exp. Med. 1999, 189, 831–841. [CrossRef][PubMed] 33. Knutson, M.D.; Oukka, M.; Koss, L.M.; Aydemir, F.; Wessling-Resnick, M. Iron release from macrophages after erythrophago- cytosis is up-regulated by ferroportin 1 overexpression and down-regulated by hepcidin. Proc. Natl. Acad. Sci. USA 2005, 102, 1324–1328. [CrossRef] 34. Lesbordes-Brion, J.C.; Viatte, L.; Bennoun, M.; Lou, D.Q.; Ramey, G.; Houbron, C.; Hamard, G.; Kahn, A.; Vaulont, S. Targeted disruption of the hepcidin 1 gene results in severe hemochromatosis. Blood 2006, 108, 1402–1405. [CrossRef][PubMed] 35. Mao, Q.Q.; Xu, X.Y.; Cao, S.Y.; Gan, R.Y.; Corke, H.; Beta, T.; Li, H.B. Bioactive compounds and bioactivities of ginger (Zingiber officinale Roscoe). Foods 2019, 8, 185. [CrossRef][PubMed] 36. Wang, W.; Knovich, M.A.; Coffman, L.G.; Torti, F.M.; Torti, S.V. Serum ferritin: Past, present and future. Biochim. Biophys. Acta 2010, 1800, 760–769. [CrossRef][PubMed] 37. Masaratana, P.; Laftah, A.H.; Latunde-Dada, G.O.; Vaulont, S.; Simpson, R.J.; McKie, A.T. Iron absorption in hepcidin1 knockout mice. Br. J. Nutr. 2011, 105, 1583–1591. [CrossRef][PubMed] 38. Brissot, P.; Ropert, M.; Le Lan, C.; Loreal, O. Non-transferrin bound iron: A key role in iron overload and iron toxicity. Biochim. Biophys. Acta 2012, 1820, 403–410. [CrossRef][PubMed] 39. Cadieux, J.A.; Zhang, Z.; Mattice, M.; Brownlie-Cutts, A.; Fu, J.; Ratkay, L.G.; Kwan, R.; Thompson, J.; Sanghara, J.; Zhong, J.; et al. Synthesis and biological evaluation of substituted pyrazoles as blockers of divalent metal transporter 1 (DMT1). Bioorg. Med. Chem. Lett. 2012, 22, 90–95. [CrossRef] 40. Wetli, H.A.; Buckett, P.D.; Wessling-Resnick, M. Small-molecule screening identifies the selanazal drug ebselen as a potent inhibitor of DMT1-mediated iron uptake. Chem. Biol. 2006, 13, 965–972. [CrossRef][PubMed] 41. Zhang, Z.; Kodumuru, V.; Sviridov, S.; Liu, S.; Chafeev, M.; Chowdhury, S.; Chakka, N.; Sun, J.; Gauthier, S.J.; Mattice, M.; et al. Discovery of benzylisothioureas as potent divalent metal transporter 1 (DMT1) inhibitors. Bioorg. Med. Chem. Lett. 2012, 22, 5108–5113. [CrossRef][PubMed] Nutrients 2021, 13, 1686 16 of 16

42. Montalbetti, N.; Simonin, A.; Simonin, C.; Awale, M.; Reymond, J.L.; Hediger, M.A. Discovery and characterization of a novel non-competitive inhibitor of the divalent metal transporter DMT1/SLC11A2. Biochem. Pharmacol. 2015, 96, 216–224. [CrossRef] [PubMed] 43. Turcu, A.L.; Versini, A.; Khene, N.; Gaillet, C.; Caneque, T.; Muller, S.; Rodriguez, R. DMT1 inhibitors kill cancer stem cells by blocking lysosomal iron translocation. Chemistry 2020, 26, 7369–7373. [CrossRef][PubMed] 44. Manatschal, C.; Pujol-Gimenez, J.; Poirier, M.; Reymond, J.L.; Hediger, M.A.; Dutzler, R. Mechanistic basis of the inhibition of SLC11/NRAMP-mediated metal ion transport by bis-isothiourea substituted compounds. Elife 2019, 8, e51913. [CrossRef] [PubMed] 45. Pulli, B.; Ali, M.; Forghani, R.; Schob, S.; Hsieh, K.L.; Wojtkiewicz, G.; Linnoila, J.J.; Chen, J.W. Measuring myeloperoxidase activity in biological samples. PLoS ONE 2013, 8, e67976. [CrossRef][PubMed]